51
|
Zhang L, Li Y, Qiao L, Zhao Y, Wei Y, Li Y. Protective effects of hepatic stellate cells against cisplatin-induced apoptosis in human hepatoma G2 cells. Int J Oncol 2015; 47:632-40. [PMID: 26035065 DOI: 10.3892/ijo.2015.3024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 04/24/2015] [Indexed: 11/06/2022] Open
Abstract
The effects of hepatic stellate cells (HSCs) on tumorigenicity of HCC have been previously reported. However, the detailed mechanisms responsible for these effects remain unclear. In this study, we investigated the effects of HSCs on cisplatin-induced apoptosis in human hepatoma HepG2 cell lines. HepG2 cells were treated with cisplatin alone or co-cultured with LX-2 cells 3 days before incubation with cisplatin. Cisplatin causes apoptosis in HepG2 cells and LX-2 cells protect HepG2 cells from death. The protection of LX-2 cells against cisplatin-induced cytotoxicity in HepG2 cells appeared to be related to the inhibition of apoptosis, as determined by cytotoxicity assay and nuclear staining analysis. p53 and Bax mRNA levels were elevated, and cell cycle arrest was produced after cisplatin treatment. LX-2 cells suppressed this elevation of p53 and Bax as well as the cell cycle arrest induced by cisplatin, when compared with those of the treated cells with cisplatin alone. The LX-2 cells pretreatment inhibited the cisplatin-induced apoptosis, which was related with the incomplete blockage in p53 activation. In summary, the results of our present study demonstrate that HSCs protect HepG2 cells against cisplatin-induced apoptosis and its protective effects occur via inhibiting the activation of p53, which is of critical importance for enhanced understanding of fundamental cancer biology.
Collapse
Affiliation(s)
- Lei Zhang
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou, Gansu 730030, P.R. China
| | - Yi Li
- School of Basic Medical Science, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Liang Qiao
- Storr Liver Unit at the Westmead Millennium Institute, the University of Sydney at Westmead Hospital, Westmead, NSW 2145, Australia
| | - Yongxun Zhao
- Department of Surgical Oncology, the First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Yucai Wei
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou, Gansu 730030, P.R. China
| | - Yumin Li
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou, Gansu 730030, P.R. China
| |
Collapse
|
52
|
Zuo Y, Yang J, He J, Zhao Y, He Y. An uncoordinated-5 homolog B receptor monoclonal antibody regulates A375 melanoma cell migration. Monoclon Antib Immunodiagn Immunother 2015; 33:280-6. [PMID: 25171009 DOI: 10.1089/mab.2013.0077] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Uncoordinated-5 homolog B receptor (UNC5B) was first found to mediate neural chemorepulsive effects by binding to its ligand netrin-1 in the nervous system. Newer evidence indicated that UNC5B also has functions outside the nervous system. In this study, we report on the generation of a monoclonal antibody specific to the outer-membrane immunoglobulin-like domains of UNC5B using the hybridoma technique. Western blot, immunofluorescence, and flow cytometry analyses showed that the antibody specifically bound to UNC5B protein. Interestingly, the antibody blocked the Netrin-1-induced inhibitory effect on the mobility of melanoma A375 cells by wound healing assay and transwell migration assay, whereas it had no effects on cell proliferation measured by CCK-8 assay. Thus, the functional antibody may provide a useful tool for the study of UNC5B expression profiles and functions outside the nervous system.
Collapse
Affiliation(s)
- Yuanling Zuo
- 1 Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University , Suzhou, China
| | | | | | | | | |
Collapse
|
53
|
Zhang C, Xu Y, Hao Q, Wang S, Li H, Li J, Gao Y, Li M, Li W, Xue X, Wu S, Zhang Y, Zhang W. FOXP3 suppresses breast cancer metastasis through downregulation of CD44. Int J Cancer 2015; 137:1279-90. [PMID: 25683728 DOI: 10.1002/ijc.29482] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 12/31/2014] [Accepted: 02/09/2015] [Indexed: 01/09/2023]
Abstract
Forkhead box protein 3 (FOXP3) plays an important role in breast cancer as an X-linked tumor suppressor gene. However, the biological functions and significance of FOXP3 in breast cancer metastasis remain unclear. Here, we find that, clinically, nuclear FOXP3 expression is inversely correlated with breast cancer metastasis. Moreover, we demonstrate that FOXP3 significantly inhibits adhesion, invasion and metastasis of breast cancer cells in vivo and in vitro. In addition, the adhesion molecule CD44 is found to be suppressed by FOXP3 through transcriptome sequence analysis (RNA-seq). A luciferase reporter assay, chromatin immunoprecipitation and electrophoretic mobility shift assay identify CD44 as a direct target of FOXP3. The expression of CD44 is downregulated by FOXP3 in breast cancer cells. Importantly, anti-CD44 antibody reverses the FOXP3 siRNA-induced effects on the breast cancer cells in vitro and FOXP3 expression level in the nucleus of breast cancer cells is inversely correlated with CD44 expression level in clinic breast cancer tissues. Taken together, the results from the present study suggest that FOXP3 is a suppressor of breast cancer metastasis. FOXP3 directly binds to the promoter of CD44 and inhibits its protein expression, thereby suppressing adhesion and invasion of human breast cancer cells. This finding highlights the therapeutic potential of FOXP3-CD44 signaling to inhibit breast cancer metastasis.
Collapse
Affiliation(s)
- Cun Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, the Fourth Military Medical University, 710032, Xi'an, People's Republic of China
| | - Yujin Xu
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, the Fourth Military Medical University, 710032, Xi'an, People's Republic of China
| | - Qiang Hao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, the Fourth Military Medical University, 710032, Xi'an, People's Republic of China
| | - Shuning Wang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, the Fourth Military Medical University, 710032, Xi'an, People's Republic of China
| | - Hong Li
- Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Jialin Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, the Fourth Military Medical University, 710032, Xi'an, People's Republic of China
| | - Yuan Gao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, the Fourth Military Medical University, 710032, Xi'an, People's Republic of China
| | - Meng Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, the Fourth Military Medical University, 710032, Xi'an, People's Republic of China
| | - Weina Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, the Fourth Military Medical University, 710032, Xi'an, People's Republic of China
| | - Xiaochang Xue
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, the Fourth Military Medical University, 710032, Xi'an, People's Republic of China
| | - Shouzhen Wu
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, the Fourth Military Medical University, 710032, Xi'an, People's Republic of China
| | - Yingqi Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, the Fourth Military Medical University, 710032, Xi'an, People's Republic of China
| | - Wei Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, the Fourth Military Medical University, 710032, Xi'an, People's Republic of China
| |
Collapse
|
54
|
Liu R, Yi B, Wei S, Yang WH, Hart KM, Chauhan P, Zhang W, Mao X, Liu X, Liu CG, Wang L. FOXP3-miR-146-NF-κB Axis and Therapy for Precancerous Lesions in Prostate. Cancer Res 2015; 75:1714-24. [PMID: 25712341 DOI: 10.1158/0008-5472.can-14-2109] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 12/23/2014] [Indexed: 02/07/2023]
Abstract
The tumor-suppressive activity of FOXP3 has been observed in tumor initiation, but the underlying mechanism still remains largely unknown. Here, we identified a FOXP3-microRNA-146 (miR-146)-NF-κB axis in vitro and in vivo in prostate cancer cells. We observed that FOXP3 dramatically induced the expression of miR-146a/b, which contributed to transcriptional inhibition of IRAK1 and TRAF6, in prostate cancer cell lines. Tissue-specific deletion of Foxp3 in mouse prostate caused a significant reduction of miR-146a and upregulation of NF-κB activation. In addition, prostatic intraepithelial neoplasia lesions were observed in miR-146a-mutant mice as well as in Foxp3-mutant mice. Notably, the NF-κB inhibitor bortezomib inhibited cell proliferation and induced apoptosis in prostate epithelial cells, attenuating prostatic intraepithelial neoplasia formation in Foxp3-mutant mice. Our data suggest that the FOXP3-miR-146-NF-κB axis has a functional role during tumor initiation in prostate cancer. Targeting the miR-146-NF-κB axis may provide a new therapeutic approach for prostate cancers with FOXP3 defects.
Collapse
Affiliation(s)
- Runhua Liu
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama. Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama.
| | - Bin Yi
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama. Department of Pediatric Surgery, Tongji Hospital of Huazhong University of Science and Technology, Wuhan, PR China
| | - Shi Wei
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Wei-Hsiung Yang
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia
| | - Karen M Hart
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Priyanka Chauhan
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Wei Zhang
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama. Institute for the Endemic Fluorosis Control, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, PR China
| | - Xicheng Mao
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Xiuping Liu
- Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, Texas
| | - Chang-Gong Liu
- Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, Texas
| | - Lizhong Wang
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama. Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama.
| |
Collapse
|
55
|
Ma C, Peng C, Lu X, Ding X, Zhang S, Zou X, Zhang X. Downregulation of FOXP3 inhibits invasion and immune escape in cholangiocarcinoma. Biochem Biophys Res Commun 2015; 458:234-9. [PMID: 25623530 DOI: 10.1016/j.bbrc.2015.01.067] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 01/14/2015] [Indexed: 10/24/2022]
Abstract
FOXP3 is known as a master control of regulatory T cells with recently studies indicating its expression in several tumor cells. In order to study the precise role of FOXP3 in cholangiocarcinoma, FOXP3 was knocked down in cholangiocarcinoma cell lines. Down regulation of FOXP3 inhibits tumor cell invasion by reducing the quantity of MMP-9 and MMP-2. With FOXP3 knocking down, IL-10 and TGF-β1 secreted by cancer cells diminishes and the cell survival of T cells is significant up-regulation. These results suggest that FOXP3 plays an important role in tumor malignant phenotype, especially the invasion and immune escape.
Collapse
Affiliation(s)
- Chao Ma
- The Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, Jiangsu Province 210008, PR China
| | - Chunyan Peng
- The Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, Jiangsu Province 210008, PR China
| | - Xuejia Lu
- The Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, Jiangsu Province 210008, PR China
| | - Xiwei Ding
- The Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, Jiangsu Province 210008, PR China
| | - Shu Zhang
- The Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, Jiangsu Province 210008, PR China
| | - Xiaoping Zou
- The Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, Jiangsu Province 210008, PR China.
| | - Xiaoqi Zhang
- The Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, Jiangsu Province 210008, PR China.
| |
Collapse
|
56
|
Qiu X, Ji B, Yang L, Huang Q, Shi W, Ding Z, He X, Ban N, Fan S, Zhang J, Tian Y. The role of FoxJ2 in the migration of human glioma cells. Pathol Res Pract 2015; 211:389-97. [PMID: 25661068 DOI: 10.1016/j.prp.2015.01.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 01/13/2015] [Indexed: 11/26/2022]
Abstract
Previous studies have demonstrated that FoxJ2 (forkhead box J2) is a member of Forkhead Box transcription factors and acts as an important prognostic indicator in human breast cancer. Our study aimed to assess the expression and function in human glioma. Western blot analysis and immunohistochemistry were performed in human glioma tissues. Low FoxJ2 expression was observed in 80 samples and its level was correlated with the grade of malignancy. A strongly positive correlation was observed between FoxJ2 and E-cadherin. Overexpression of FoxJ2 increased E-cadherin expression and decreased vimentin expression. The wound healing and transwell assays showed that overexpression of FoxJ2 significantly inhibited their migration in U87 cells. Consistent with this, knockdown of FoxJ2 promoted cellular motility. In a word, FoxJ2 suppressed cell migration and invasion in glioma, which might be a potential novel molecular targeted therapy for surgery and immune treatment.
Collapse
Affiliation(s)
- Xiaojun Qiu
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou 215004, People's Republic of China; Department of Radiotherapy and Oncology, The Affiliated Hospital of Nantong University, Xisi Road No. 20, Nantong 226001, People's Republic of China
| | - Bin Ji
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Nantong University, Xisi Road No. 20, Nantong 226001, People's Republic of China
| | - Lixiang Yang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215006, People's Republic of China
| | - Qingfeng Huang
- Department of Neurosurgery, The Affiliated Hospital of Nantong University, Xisi Road No. 20, Nantong 226001, People's Republic of China
| | - Wei Shi
- Department of Neurosurgery, The Affiliated Hospital of Nantong University, Xisi Road No. 20, Nantong 226001, People's Republic of China
| | - Zongmei Ding
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, Jiangsu Province 226001, People's Republic of China
| | - Xiaojuan He
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, Jiangsu Province 226001, People's Republic of China
| | - Na Ban
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, Jiangsu Province 226001, People's Republic of China
| | - Shaochen Fan
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, Jiangsu Province 226001, People's Republic of China
| | - Jianguo Zhang
- Department of Pathology, The Affiliated Hospital of Nantong University, Xisi Road No. 20, Nantong 226001, People's Republic of China
| | - Ye Tian
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou 215004, People's Republic of China.
| |
Collapse
|
57
|
Rao YF, Chen W, Liang XG, Huang YZ, Miao J, Liu L, Lou Y, Zhang XG, Wang B, Tang RK, Chen Z, Lu XY. Epirubicin-loaded superparamagnetic iron-oxide nanoparticles for transdermal delivery: cancer therapy by circumventing the skin barrier. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2015; 11:239-247. [PMID: 24925046 DOI: 10.1002/smll.201400775] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Revised: 05/24/2014] [Indexed: 06/03/2023]
Abstract
The transdermal administration of chemotherapeutic agents is a persistent challenge for tumor treatments. A model anticancer agent, epirubicin (EPI), is attached to functionalized superparamagnetic iron-oxide nanoparticles (SPION). The covalent modification of the SPION results in EPI-SPION, a potential drug delivery vector that uses magnetism for the targeted transdermal chemotherapy of skin tumors. The spherical EPI-SPION composite exhibits excellent magnetic responsiveness with a saturation magnetization intensity of 77.8 emu g(-1) . They feature specific pH-sensitive drug release, targeting the acidic microenvironment typical in common tumor tissues or endosomes/lysosomes. Cellular uptake studies using human keratinocyte HaCaT cells and melanoma WM266 cells demonstrate that SPION have good biocompatibility. After conjugation with EPI, the nanoparticles can inhibit WM266 cell proliferation; its inhibitory effect on tumor proliferation is determined to be dose-dependent. In vitro transdermal studies demonstrate that the EPI-SPION composites can penetrate deep inside the skin driven by an external magnetic field. The magnetic-field-assisted SPION transdermal vector can circumvent the stratum corneum via follicular pathways. The study indicates the potential of a SPION-based vector for feasible transdermal therapy of skin cancer.
Collapse
Affiliation(s)
- Yue-feng Rao
- The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China; Department of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Chu R, Liu SYW, Vlantis AC, van Hasselt CA, Ng EKW, Fan MD, Ng SK, Chan ABW, Du J, Wei W, Liu X, Liu Z, Chen GG. Inhibition of Foxp3 in cancer cells induces apoptosis of thyroid cancer cells. Mol Cell Endocrinol 2015; 399:228-34. [PMID: 25312920 DOI: 10.1016/j.mce.2014.10.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 09/25/2014] [Accepted: 10/08/2014] [Indexed: 12/31/2022]
Abstract
Foxp3+ regulatory T cells (Tregs) in lymphocytes facilitate the thyroid tumor growth and invasion. Very limited information is available on Foxp3 expression in thyroid cancer cells and its function is totally unknown. This study demonstrated that Foxp3 expression was increased in thyroid cancer cells. Inhibition of Foxp3 decreased cell proliferation and migration, but increased apoptosis, suggesting a positive role of Foxp3 in cancer growth. Interestingly, Foxp3 inhibition enhanced PPARγ expression and activity. In addition, Foxp3 inhibition downregulated NF-κB subunit p65 and cyclin D1 but upregulated caspase-3 levels. These molecular changes are in line with Foxp3 shRNA-mediated alteration of cell functions. Collectively, our study demonstrates that thyroid cancer cells express a high level of functional Foxp3 and that the inhibition of the Foxp3 suppresses the proliferation and migration but promotes apoptosis, suggesting that targeting Foxp3 in thyroid cancer cells may offer a novel therapeutic option for thyroid cancer.
Collapse
Affiliation(s)
- Ryan Chu
- Department of Otorhinolaryngology, Head and Neck Surgery, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| | - Shirley Y W Liu
- Department of Surgery, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| | - Alexander C Vlantis
- Department of Otorhinolaryngology, Head and Neck Surgery, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| | - C Andrew van Hasselt
- Department of Otorhinolaryngology, Head and Neck Surgery, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| | - Enders K W Ng
- Department of Surgery, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| | - Michael Dahua Fan
- Department of Otorhinolaryngology, Head and Neck Surgery, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| | - Siu Kwan Ng
- Department of Otorhinolaryngology, Head and Neck Surgery, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| | - Amy B W Chan
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| | - Jing Du
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Wei Wei
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Xiaoling Liu
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Zhimin Liu
- Department of Biochemistry and Molecular Biology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - George G Chen
- Department of Surgery, Prince of Wales Hospital, Shatin, NT, Hong Kong, China.
| |
Collapse
|
59
|
Tan B, Anaka M, Deb S, Freyer C, Ebert LM, Chueh AC, Al-Obaidi S, Behren A, Jayachandran A, Cebon J, Chen W, Mariadason JM. FOXP3 over-expression inhibits melanoma tumorigenesis via effects on proliferation and apoptosis. Oncotarget 2014; 5:264-76. [PMID: 24406338 PMCID: PMC3960207 DOI: 10.18632/oncotarget.1600] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The Forkhead box P3 (FOXP3) transcription factor is the key driver of regulatory T cell (Treg cells) differentiation and immunosuppressive function. In addition, FOXP3 has been reported to be expressed in many tumors, including melanoma. However, its role in tumorigenesis is conflicting, with both tumor suppressive and tumor promoting functions described. The aim of the current study was to characterize the expression and function of FOXP3 in melanoma. FOXP3 expression was detected by immunohistochemistry (IHC) in 12% (18/146) of stage III and IV melanomas. However expression was confined to fewer than 1% of cells in these tumors. Stable over-expression of FOXP3 in the SK-MEL-28 melanoma cell line reduced cell proliferation and clonogenicity in vitro, and reduced xenograft growth in vivo. FOXP3 over-expression also increased pigmentation and the rate of apoptosis of SK-MEL-28 cells. Based on its infrequent expression in human melanoma, and its growth inhibitory and pro-apoptotic effect in over-expressing melanoma cells, we conclude that FOXP3 is not likely to be a key tumor suppressor or promoter in melanoma.
Collapse
Affiliation(s)
- BeeShin Tan
- Ludwig Institute for Cancer Research Ltd. Melbourne-Austin Branch, Heidelberg, Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Hermans C, Anz D, Engel J, Kirchner T, Endres S, Mayr D. Analysis of FoxP3+ T-regulatory cells and CD8+ T-cells in ovarian carcinoma: location and tumor infiltration patterns are key prognostic markers. PLoS One 2014; 9:e111757. [PMID: 25365237 PMCID: PMC4218808 DOI: 10.1371/journal.pone.0111757] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 09/30/2014] [Indexed: 01/05/2023] Open
Abstract
Purpose Tumor infiltrating CD4+CD25+FoxP3+ regulatory immune cells (Treg) have been associated with impaired anti- tumor immune response and unfavorable prognosis for patients affected by ovarian carcinoma, whereas CD8+ T-cells have been found to positively influence survival rates in a large panel of solid tumors. Recently, density, location and tumor infiltration patterns of the respective immune cell subtypes have been identified as key prognostic factors for different types of tumors. Patients and Methods We stained 210 human ovarian carcinoma samples immunhistochemically for FoxP3 and CD8 to identify the impact different immune cell patterns have on generally accepted prognostic variables as well as on overall survival. Results We found that FoxP3+ cells located within lymphoid aggregates surrounding the tumor were strongly associated with reduced survival time (P = 0.007). Central accumulation of CD8+ effector cells within the tumor bed shows a positive effect on survival (P = 0,001). Conclusion The distribution pattern of immune cells within the tumor environment strongly influences prognosis and overall survival time of patients with ovarian carcinoma.
Collapse
Affiliation(s)
- Cecilia Hermans
- Department of Pathology, Ludwig-Maximilians-University, Munich, Germany
- * E-mail:
| | - David Anz
- Center of Integrated Protein Science Munich (CIPS-M) Division of Clinical Pharmacology, Ludwig-Maximilians-University, Munich, Germany
| | - Jutta Engel
- Tumorregister Munich, Institute of Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-University, Munich, Germany
| | - Thomas Kirchner
- Department of Pathology, Ludwig-Maximilians-University, Munich, Germany
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) Division of Clinical Pharmacology, Ludwig-Maximilians-University, Munich, Germany
| | - Doris Mayr
- Department of Pathology, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
61
|
Chemopreventive effect of flavonoids from Ougan (Citrus reticulata cv. Suavissima) fruit against cancer cell proliferation and migration. J Funct Foods 2014. [DOI: 10.1016/j.jff.2014.08.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
62
|
Douglass S, Meeson AP, Overbeck-Zubrzycka D, Brain JG, Bennett MR, Lamb CA, Lennard TWJ, Browell D, Ali S, Kirby JA. Breast cancer metastasis: demonstration that FOXP3 regulates CXCR4 expression and the response to CXCL12. J Pathol 2014; 234:74-85. [PMID: 24870556 DOI: 10.1002/path.4381] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 04/29/2014] [Accepted: 05/22/2014] [Indexed: 01/20/2023]
Abstract
The X-linked transcription factor FOXP3 is expressed by epithelial cells of organs including the breast, where it is considered a tumour suppressor. The chemokine receptor CXCR4 also regulates the development of breast cancer by stimulating cell migration towards CXCL12-expressing sites of metastatic spread. During activation, human T cells show reciprocal regulation of FOXP3 and CXCR4. This study was designed to examine the role FOXP3 plays in metastatic breast cancer, with a particular focus on its potential to regulate CXCR4. Human breast cancer samples showed significantly decreased FOXP3 protein expression but an increased number of CXCR4 transcripts. In comparison with normal primary breast epithelial cells, FOXP3 was down-regulated at both transcript and protein levels in the breast cancer cell lines MCF-7 and MDA-MB-231. In the invasive MDA-MB-231 cells, the remaining FOXP3 was located predominately within the cytoplasm. Following stable FOXP3 overexpression in MDA-MB-231 cells, significant decreases were observed in the expression of ErbB2/HER2, SKP2, c-MYC, and CXCR4. In contrast, an increase in p21 expression led to inhibition of cell proliferation, with a greater proportion in the G1 phase of the cell cycle suggesting the induction of senescence. Specific knockdown of FOXP3 in normal human breast epithelial cells with siRNA significantly increased ErbB2/HER2, SKP2, c-MYC, and CXCR4, and decreased p21 expression. These cells also showed a significantly increased chemotactic response towards CXCL12, consistent with a role for FOXP3 in the regulation of cell migration. Results from this study are consistent with FOXP3 functioning as an important tumour suppressor in breast cancer. Indeed, the potential functions of FOXP3 in breast epithelium can now be extended to include regulation of CXCR4 expression and response to the pro-metastatic chemokine CXCL12.
Collapse
Affiliation(s)
- Stephen Douglass
- Applied Immunobiology Research Group, Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Zhuo C, Li Z, Xu Y, Wang Y, Li Q, Peng J, Zheng H, Wu P, Li B, Cai S. Higher FOXP3-TSDR demethylation rates in adjacent normal tissues in patients with colon cancer were associated with worse survival. Mol Cancer 2014; 13:153. [PMID: 24938080 PMCID: PMC4074420 DOI: 10.1186/1476-4598-13-153] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Accepted: 06/11/2014] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The influence of natural regulatory T cells (nTregs) on the patients with colon cancer is unclear. Demethylated status of the Treg-specific demethylated region (TSDR) of the FOXP3 gene was reported to be a potential biomarker for the identification of nTregs. METHODS The demethylation rate of the TSDR (TSDR-DMR) was calculated by using methylation-specific quantitative polymerase chain reaction (MS-qPCR) assay. The expression of TSDR-DMR and FOXP3 mRNA was investigated in various colorectal cancer cell lines. A total of 130 colon carcinoma samples were utilized to study the DMR at tumor sites (DMRT) and adjacent normal tissue (DMRN). The correlations between DMRs and clinicopathological variables of patients with colon cancer were studied. RESULTS The TSDR-DMRs varied dramatically among nTregs (97.920 ± 0.466%) and iTregs (3.917 ± 0.750%). Significantly, DMRT (3.296 ± 0.213%) was higher than DMRN (1.605 ± 0.146%) (n = 130, p = 0.000). Higher DMRN levels were found in female patients (p = 0.001) and those with distant metastases (p = 0.017), and were also associated with worse recurrence-free survival in non-stage IV patients (low vs. high, p = 0.022). However, further Cox multivariate analysis revealed that the FOXP3-TSDR status does not have prognostic value. CONCLUSION MS-qPCR assays of FOXP3-TSDR can efficiently distinguish nTregs from non-nTregs. Abnormal recruitment of nTregs occurs in the local tumor microenvironment. Infiltration of tissue-resident nTregs may have a negative role in anti-tumor effects in patients with colon cancer; however, this role is limited and complicated.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Bin Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, No, 270 Dong-an Road, Shanghai 20032, People's Republic of China.
| | | |
Collapse
|
64
|
Gan F, Chen X, Liao SF, Lv C, Ren F, Ye G, Pan C, Huang D, Shi J, Shi X, Zhou H, Huang K. Selenium-enriched probiotics improve antioxidant status, immune function, and selenoprotein gene expression of piglets raised under high ambient temperature. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:4502-4508. [PMID: 24814575 DOI: 10.1021/jf501065d] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
This research was conducted to evaluate the effects of selenium-enriched probiotics (SP) on growth performance, antioxidant status, immune function, and selenoprotein gene expression of piglets under natural high ambient temperature in summer. Forty-eight crossbred weanling piglets randomly allocated to four groups were fed for 42 days ad libitum a basal diet without (Con, 0.16 mg Se/kg) and with supplementation of probiotics (P, 0.16 mg Se/kg), sodium selenite (SS, 0.46 mg Se/kg), and SP (0.46 mg Se/kg). From each group, three piglets were randomly selected for blood collection on days 0, 14, 28, and 42 and tissue collection on day 42. The SP improved growth performance of piglets. Both SS and SP increased blood glutathione peroxidase activity and tissue thioredoxin reductase 1 mRNA expression, with SP being higher than SS. All P, SS, and SP supplementation increased the superoxide dismutase activity (40.1, 53.0, and 64.5%), glutathione content (84.6, 104, and 165%), TCR-induced T lymphocyte proliferation (20.8, 26.4, and 50.0%), and IL-2 concentration (24.9, 27.2, and 46.2%) and decreased malondialdehyde content (25.1, 26.3, and 49.3%), respectively. The greatest effects of SP supplementation suggest that SP may serve as a better feed additive than P or SS for piglets under high-temperature environments.
Collapse
Affiliation(s)
- Fang Gan
- Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University , Nanjing, Jiangsu 210095, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Luo F, Fu Y, Xiang Y, Yan S, Hu G, Huang X, Huang G, Sun C, Li X, Chen K. Identification and quantification of gallotannins in mango (Mangifera indica L.) kernel and peel and their antiproliferative activities. J Funct Foods 2014. [DOI: 10.1016/j.jff.2014.03.030] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
66
|
High FoxP3 expression in tumour cells predicts better survival in gastric cancer and its role in tumour microenvironment. Br J Cancer 2014; 110:1552-60. [PMID: 24548868 PMCID: PMC3960619 DOI: 10.1038/bjc.2014.47] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Revised: 12/31/2013] [Accepted: 01/08/2014] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Forkhead Box P3 (FoxP3) is thought to be a key transcription factor in regulatory T cells (Tregs), and recent data indicate that it is expressed in several tumour cells. However, its precise roles in gastric cancer (GC) and the underlying mechanisms regulating the interaction between GC cells and lymphocytes remain unclear. METHODS FoxP3 expression was examined in tumour cells and Tregs in 150 cases of gastric precancer and cancer, and their prognostic significances were evaluated, respectively, using a tissue microarray containing 135 GC patient samples with a mean 102-month follow-up. FoxP3 involvement in the tumour cells-lymphocytes interaction and its gene function were further investigated. RESULTS strong cytoplasmic staining of FoxP3 was observed in GC cells. FoxP3 protein expression in tumour cells predicts a good prognosis, whereas high-density Treg predicts a poor prognosis. Moreover, FoxP3 expression in GC cells increased after coculture with peripheral blood mononuclear cells through coculture systems. Upregulation of FoxP3 inhibited tumour growth in tumour-bearing nude mice. CONCLUSIONS High FoxP3 expression in tumour cells predicts better survival in GC, possibility in relation to interaction between tumour cells and lymphocytes in microenvironment. Interfering with FoxP3 expression may open a new therapeutic strategy against tumour progression.
Collapse
|
67
|
Yang L, Su T, Lv D, Xie F, Liu W, Cao J, Sheikh IA, Qin X, Li L, Chen L. ERK1/2 mediates lung adenocarcinoma cell proliferation and autophagy induced by apelin-13. Acta Biochim Biophys Sin (Shanghai) 2014; 46:100-11. [PMID: 24374773 DOI: 10.1093/abbs/gmt140] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The aim of this study was to investigate the role of apelin in the cell proliferation and autophagy of lung adenocarcinoma. The over-expression of APJ in lung adenocarcinoma was detected by immunohistochemistry, while plasma apelin level in lung cancer patients was measured by enzyme-linked immunosorbent assay. Our findings revealed that apelin-13 significantly increased the phosphorylation of ERK1/2, the expression of cyclin D1, microtubule-associated protein 1 light chain 3A/B (LC3A/B), and beclin1, and confirmed that apelin-13 promoted A549 cell proliferation and induced A549 cell autophagy via ERK1/2 signaling. Moreover, there are pores on the surface of human lung adenocarcinoma cell line A549 and apelin-13 causes cell surface smooth and glossy as observed under atomic force microscopy. These results suggested that ERK1/2 signaling pathway mediates apelin-13-induced lung adenocarcinoma cell proliferation and autophagy. Under our experimental condition, autophagy associated with 3-methyladenine was not involved in cell proliferation.
Collapse
Affiliation(s)
- Li Yang
- Learning Key Laboratory for Pharmaco-proteomics, Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Liu T, Song YN, Shi QY, Liu Y, Bai XN, Pang D. Study of circulating antibodies against CD25 and FOXP3 in breast cancer. Tumour Biol 2013; 35:3779-83. [PMID: 24347486 DOI: 10.1007/s13277-013-1500-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 11/29/2013] [Indexed: 01/14/2023] Open
Abstract
Our recent work suggests that circulating levels of anti-CD25 and anti-FOXP3 antibodies were significantly increased in patients with either lung cancer or esophageal cancer. To confirm if these two autoantibodies are specific for certain types of malignant tumors, the present work was thus undertaken to examine an alteration of anti-CD25 and anti-FOXP3 IgG levels in breast cancer. A total of 152 patients with breast cancer and 112 control subjects were recruited in this study. The levels of circulating anti-CD25 and anti-FOXP3 IgG antibodies were tested using an in-house enzyme-linked immunosorbent assay (ELISA). Student's t test showed no significant differences in the levels of either anti-CD25 IgG or anti-FOXP3 IgG between patients with breast cancer and control subjects, although patients at stage I had increased levels of anti-CD25 IgG compared with control subjects (t = 2.11, P = 0.037); there was no significant association of the anti-FOXP3 IgG levels with stages of breast cancer. In conclusion, circulating IgG autoantibody to CD25 instead of FOXP3 may be a potential biomarker for early diagnosis of breast cancer but further investigation remains needed to replicate this initial finding.
Collapse
Affiliation(s)
- Tong Liu
- Department of Breast Surgery, The Third Affiliated Hospital of Harbin Medical University, Harbin, 150040, China
| | | | | | | | | | | |
Collapse
|
69
|
Wang J, Yang ZR, Dong WG, Zhang JX, Guo XF, Song J, Qiu S. Cooperative inhibitory effect of sinomenine combined with 5-fluorouracil on esophageal carcinoma. World J Gastroenterol 2013; 19:8292-8300. [PMID: 24363520 PMCID: PMC3857452 DOI: 10.3748/wjg.v19.i45.8292] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 10/04/2013] [Accepted: 10/14/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the inhibitory effects of sinomenine (SIN) combined with 5-fluorouracil (5-FU) on esophageal carcinoma in vitro and in vivo.
METHODS: Esophageal carcinoma (Eca-109) cells were cultured in DMEM. The single or combined growth inhibition effects of SIN and 5-FU on the Eca-109 cells were examined by measuring the absorbance of CCK-8 dye in living cells. Hoechst 33258 staining and an Annexin V/PI apoptosis kit were used to detect the percentage of cells undergoing apoptosis. Western blotting was used to investigate the essential mechanism underlying SIN and 5-FU-induced apoptosis. SIN at 25 mg/kg and 5-FU at 12 mg/kg every 3 d, either combined or alone, was injected into nude mice and tumor growth inhibition and side effects of the drug treatment were observed.
RESULTS: SIN and 5-FU, both in combination and individually, significantly inhibited the proliferation of Eca-109 cells and induced obvious apoptosis. Furthermore, the combined effects were greater than those of the individual agents (P < 0.05). Annexin V/PI staining and Hoechst 33258 staining both indicated that the percentage of apoptotic cells induced by SIN and 5-FU combined or alone were significantly different from the control (P < 0.05). The up-regulation of Bax and down-regulation of Bcl-2 showed that the essential mechanism of apoptosis induced by SIN and 5-FU occurs via the mitochondrial pathway. SIN and 5-FU alone significantly inhibited the growth of tumor xenografts in vivo, and the combined inhibition rate was even higher (P < 0.05). During the course of chemotherapy, no obvious side effects were observed in the liver or kidneys.
CONCLUSION: The combined effects of SIN and 5-FU on esophageal carcinoma were superior to those of the individual compounds, and the drug combination did not increase the side effects of chemotherapy.
Collapse
|
70
|
Nakahira K, Morita A, Kim NS, Yanagihara I. Phosphorylation of FOXP3 by LCK downregulates MMP9 expression and represses cell invasion. PLoS One 2013; 8:e77099. [PMID: 24155921 PMCID: PMC3796550 DOI: 10.1371/journal.pone.0077099] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 08/30/2013] [Indexed: 12/31/2022] Open
Abstract
Forkhead Box P3 (FOXP3) is a member of the forkhead/winged helix family of the transcription factors and plays an important role not only as a master gene in T-regulatory cells, but also as a tumor suppressor. In this study, we identified lymphocyte-specific protein tyrosine kinase (LCK), which correlates with cancer malignancy, as a binding partner of FOXP3. FOXP3 downregulated LCK-induced MMP9, SKP2, and VEGF-A expression. We observed that LCK phosphorylated Tyr-342 of FOXP3 by immunoprecipitation and in vitro kinase assay, and the replacement of Tyr-342 with phenylalanine (Y342F) abolished the ability to suppress MMP9 expression. Although FOXP3 decreased the invasive ability induced by LCK in MCF-7 cells, Y342F mutation in FOXP3 diminished this suppressive effect. Thus we demonstrate for the first time that LCK upregulates FOXP3 by tyrosine phosphorylation, resulting in decreased MMP9, SKP2, and VEGF-A expression, and suppressed cellular invasion. We consider that further clarification of transcriptional mechanism of FOXP3 may facilitate the development of novel therapeutic approaches to suppress cancer malignancy.
Collapse
Affiliation(s)
- Kumiko Nakahira
- Department of Developmental Medicine, Research Institute, Osaka Medical Center for Maternal and Child Health, Izumi, Osaka, Japan
| | - Akihiro Morita
- Department of Developmental Medicine, Research Institute, Osaka Medical Center for Maternal and Child Health, Izumi, Osaka, Japan
| | - Nam-Soon Kim
- Biomedical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon, Korea
| | - Itaru Yanagihara
- Department of Developmental Medicine, Research Institute, Osaka Medical Center for Maternal and Child Health, Izumi, Osaka, Japan
- * E-mail:
| |
Collapse
|
71
|
Wang Z, Han J, Cui Y, Zhou X, Fan K. miRNA-21 inhibition enhances RANTES and IP-10 release in MCF-7 via PIAS3 and STAT3 signalling and causes increased lymphocyte migration. Biochem Biophys Res Commun 2013; 439:384-9. [PMID: 23998932 DOI: 10.1016/j.bbrc.2013.08.072] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 08/21/2013] [Indexed: 12/14/2022]
Abstract
MicroRNAs (miRNAs) are a class of small endogenous gene regulators that have been implicated in various developmental and pathological processes. However, the precise identities and functions of miRNAs involved in antitumor immunity are not yet well understood. miRNA-21 is an oncogenic miRNA that can be detected in various tumours. In this study, we report that a miRNA-21 inhibitor enhances the release of chemoattractants RANTES and IP-10 in the MCF-7 breast cancer cell line and results in increased lymphocyte migration. Thus, miRNA-21 is a potential therapeutic target for cancer immunotherapy. We further demonstrated that PIAS3, a protein inhibitor of activated STAT3, is a target of miRNA-21 in MCF-7. Thus, miRNA-21 is a novel miRNA regulating immune cell recruitment, which acts at least in part via its inhibition of PIAS3 expression and oncogenic STAT3 signalling in tumour cells.
Collapse
Affiliation(s)
- Zhiyu Wang
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan 250012, Shandong, PR China; Shandong Medicinal Biotechnology Center, Key Laboratory for Biotech-Drugs, Ministry of Health, Shandong Academy of Medical Sciences, Jinan 250062, Shandong, PR China
| | | | | | | | | |
Collapse
|
72
|
Abstract
Forkhead box (FOX) proteins are multifaceted transcription factors that are responsible for fine-tuning the spatial and temporal expression of a broad range of genes both during development and in adult tissues. This function is engrained in their ability to integrate a multitude of cellular and environmental signals and to act with remarkable fidelity. Several key members of the FOXA, FOXC, FOXM, FOXO and FOXP subfamilies are strongly implicated in cancer, driving initiation, maintenance, progression and drug resistance. The functional complexities of FOX proteins are coming to light and have established these transcription factors as possible therapeutic targets and putative biomarkers for specific cancers.
Collapse
Affiliation(s)
- Eric W-F Lam
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK.
| | | | | | | |
Collapse
|
73
|
Abstract
The transcription factor FOXP3 is widely known for its role in the development and function of immunoregulatory T cells. However, it has been discovered recently that FOXP3 is also expressed in epithelial cells of the normal human breast, ovary and prostate. Aggressive cancer of these epithelial tissues often correlates with abnormal expression of FOXP3, which can be either absent or underexpressed at transcript or protein levels. It is becoming clear that this failure of normal FOXP3 expression can result in dysregulation of the expression of a range of oncogenes which have been implicated in the development and metastasis of cancer. Recent evidence suggests that FOXP3 might also regulate chemokine receptor expression, providing a possible explanation for the chemokine-driven, tissue-specific spread that is characteristic of many cancers. This review first summarises the general structure, function and properties of FOXP3. This is followed by an analysis of the tumour-suppressive properties of this transcription factor, with particular reference to the development and chemokine-mediated spread of human breast cancer. A final section focuses on potential applications of this new knowledge for therapeutic intervention.
Collapse
|
74
|
Abstract
In this review, we introduce the IPEX syndrome and its relationship with germline mutations of the FOXP3 gene. We then describe the multiple functional roles of FOXP3 in regulatory T cells and epithelial cells as well as in IPEX syndrome and tumor progression. Potential mechanisms of FOXP3 inactivation and transcriptional regulation are discussed with recent advances. Finally, we point out current issues and a potential FOXP3-mediated therapeutic strategy as well as the reactivation of FOXP3 in patients with IPEX syndrome and cancer.
Collapse
Affiliation(s)
- Runhua Liu
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | - Silin Li
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | - Wei-Hsiung Yang
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia 31404, USA
| | | |
Collapse
|
75
|
Takenaka M, Seki N, Toh U, Hattori S, Kawahara A, Yamaguchi T, Koura K, Takahashi R, Otsuka H, Takahashi H, Iwakuma N, Nakagawa S, Fujii T, Sasada T, Yamaguchi R, Yano H, Shirouzu K, Kage M. FOXP3 expression in tumor cells and tumor-infiltrating lymphocytes is associated with breast cancer prognosis. Mol Clin Oncol 2013; 1:625-632. [PMID: 24649219 PMCID: PMC3915667 DOI: 10.3892/mco.2013.107] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 04/05/2013] [Indexed: 12/04/2022] Open
Abstract
The forkhead box protein 3 (FOXP3) transcription factor is highly expressed in tumor cells as well as in regulatory T cells (Tregs). It plays a tumor-enhancing role in Tregs and suppresses carcinogenesis as a potent repressor of several oncogenes. The clinical prognostic value of FOXP3 expression has not yet been elucidated. In this study, immunohistochemistry was used to investigate the prognostic significance of FOXP3 expression in tumor cells and tumor-infiltrating lymphocytes (TILs) in breast cancer patients. Of the 100 tumor specimens obtained from primary invasive breast carcinoma, 63 and 57% were evaluated as FOXP3+ tumor cells and as being highly infiltrated by FOXP3+ lymphocytes, respectively. Although FOXP3 expression in tumor cells was of no prognostic significance, FOXP3+ lymphocytes were significantly associated with poor overall survival (OS) (n=98, log-rank test P=0.008). FOXP3 exhibited a heterogeneous subcellular localization in tumor cells (cytoplasm, 31%; nucleus, 26%; both, 6%) and, although cytoplasmic FOXP3 was associated with poor OS (P= 0.058), nuclear FOXP3 demonstrated a significant association with improved OS (P=0.016). Furthermore, when patients were grouped according to their expression of tumor cytoplasmic FOXP3 and lymphocyte FOXP3, there were notable differences in the Kaplan-Meier curves for OS (P<0.001), with a high infiltration of FOXP3+ lymphocytes accompanied by a cytoplasmic FOXP3+ tumor being the most detrimental phenotype. These findings indicated that FOXP3 expression in lymphocytes as well as in tumor cells may be a prognostic marker for breast cancer. FOXP3 in tumor cells may have distinct biological activities and prognostic values according to its localization, which may help establish appropriate cancer treatments.
Collapse
Affiliation(s)
- Miki Takenaka
- Departments of Pathology, School of Medicine, Kurume University, Kurume, Fukuoka 830-0011, Japan ; ; Surgery, School of Medicine, Kurume University, Kurume, Fukuoka 830-0011, Japan
| | - Naoko Seki
- Research Center for Innovative Cancer Therapy, School of Medicine, Kurume University, Kurume, Fukuoka 830-0011, Japan
| | - Uhi Toh
- Surgery, School of Medicine, Kurume University, Kurume, Fukuoka 830-0011, Japan
| | - Satoshi Hattori
- Biostatistical Center, Kurume University, Kurume, Fukuoka 830-0011, Japan
| | - Akihiko Kawahara
- Department of Diagnostic Pathology, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Tomohiko Yamaguchi
- Department of Diagnostic Pathology, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Keiko Koura
- Departments of Pathology, School of Medicine, Kurume University, Kurume, Fukuoka 830-0011, Japan ; ; Surgery, School of Medicine, Kurume University, Kurume, Fukuoka 830-0011, Japan
| | - Ryuji Takahashi
- Surgery, School of Medicine, Kurume University, Kurume, Fukuoka 830-0011, Japan
| | - Hiroko Otsuka
- Surgery, School of Medicine, Kurume University, Kurume, Fukuoka 830-0011, Japan
| | - Hiroki Takahashi
- Surgery, School of Medicine, Kurume University, Kurume, Fukuoka 830-0011, Japan
| | - Nobutaka Iwakuma
- Surgery, School of Medicine, Kurume University, Kurume, Fukuoka 830-0011, Japan
| | - Shino Nakagawa
- Surgery, School of Medicine, Kurume University, Kurume, Fukuoka 830-0011, Japan
| | - Teruhiko Fujii
- Surgery, School of Medicine, Kurume University, Kurume, Fukuoka 830-0011, Japan ; ; Multidisciplinary Treatment Center, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Tetsuro Sasada
- Department of Immunology, School of Medicine, Kurume University, Kurume, Fukuoka 830-0011, Japan
| | - Rin Yamaguchi
- Departments of Pathology, School of Medicine, Kurume University, Kurume, Fukuoka 830-0011, Japan
| | - Hirohisa Yano
- Departments of Pathology, School of Medicine, Kurume University, Kurume, Fukuoka 830-0011, Japan
| | - Kazuo Shirouzu
- Surgery, School of Medicine, Kurume University, Kurume, Fukuoka 830-0011, Japan
| | - Masayoshi Kage
- Research Center for Innovative Cancer Therapy, School of Medicine, Kurume University, Kurume, Fukuoka 830-0011, Japan ; ; Department of Diagnostic Pathology, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| |
Collapse
|
76
|
Fu HY, Li C, Yang W, Gai XD, Jia T, Lei YM, Li Y. FOXP3 and TLR4 protein expression are correlated in non-small cell lung cancer: implications for tumor progression and escape. Acta Histochem 2013; 115:151-7. [PMID: 22749378 DOI: 10.1016/j.acthis.2012.06.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 06/04/2012] [Accepted: 06/05/2012] [Indexed: 12/24/2022]
Abstract
NSCLC (non-small cell lung cancer) is the most common type of lung cancer and usually has poor prognosis. FOXP3 in regulatory T cells (Tregs) and toll-like receptor 4 (TLR4) on some tumor cells are known to be important for tumor escape and clinical tumor formation. Since FOXP3 was found recently in some tumor cells, we speculated if lung tumor cells express FOXP3 and then mimic Tregs to promote tumor escape. As TLR4 induces activation of Tregs, we also hypothesized that FOXP3 and TLR4 may have a correlation in NSCLC progression. The expression levels of FOXP3 and TLR4 protein were detected using immunohistochemistry in 53 postoperative specimens of NSCLC patients and in 15 normal lung tissues from excisions of benign lesion. The relationship between protein expression levels and clinical pathology parameters, as well as the relationship between the expression of FOXP3 and TLR4, were analyzed. FOXP3 and TLR4 expression in NSCLC were significantly elevated as compared to normal lung tissue. FOXP3 expression was closely related with lymph node metastasis and TNM staging, whereas TLR4 expression was closely related with tumor differentiation. The Spearman correlation coefficient indicated a significant positive correlation between FOXP3 and TLR4 expression. These results indicate that FOXP3 and TLR4 may coordinate to play a role in tumor escape and subsequent tumor progression.
Collapse
Affiliation(s)
- Hai-ying Fu
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun, Jilin 130021, PR China
| | | | | | | | | | | | | |
Collapse
|
77
|
Katoh H, Zheng P, Liu Y. FOXP3: genetic and epigenetic implications for autoimmunity. J Autoimmun 2013; 41:72-8. [PMID: 23313429 DOI: 10.1016/j.jaut.2012.12.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 12/16/2012] [Indexed: 12/12/2022]
Abstract
FOXP3 plays an essential role in the maintenance of self-tolerance and, thus, in preventing autoimmune diseases. Inactivating mutations of FOXP3 cause immunodysregulation, polyendocrinopathy, and enteropathy, X-linked syndrome. FOXP3-expressing regulatory T cells attenuate autoimmunity as well as immunity against cancer and infection. More recent studies demonstrated that FOXP3 is an epithelial cell-intrinsic tumor suppressor for breast, prostate, ovary and other cancers. Corresponding to its broad function, FOXP3 regulates a broad spectrum of target genes. While it is now well established that FOXP3 binds to and regulates thousands of target genes in mouse and human genomes, the fundamental mechanisms of its broad impact on gene expression remain to be established. FOXP3 is known to both activate and repress target genes by epigenetically regulating histone modifications of target promoters. In this review, we first focus on germline mutations found in the FOXP3 gene among IPEX patients, then outline possible molecular mechanisms by which FOXP3 epigenetically regulates its targets. Finally, we discuss clinical implications of the function of FOXP3 as an epigenetic modifier. Accumulating results reveal an intriguing functional convergence between FOXP3 and inhibitors of histone deacetylases. The essential epigenetic function of FOXP3 provides a foundation for experimental therapies against autoimmune diseases.
Collapse
Affiliation(s)
- Hiroto Katoh
- Division of Molecular Oncology, Institute for Genetic Medicine, Hokkaido University, Hokkaido, Japan
| | | | | |
Collapse
|
78
|
TNFα induced FOXP3–NFκB interaction dampens the tumor suppressor role of FOXP3 in gastric cancer cells. Biochem Biophys Res Commun 2013. [DOI: 10.1016/j.bbrc.2012.11.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
79
|
Held-Feindt J, Hattermann K, Sebens S, Krautwald S, Mehdorn HM, Mentlein R. The transcription factor Forkhead box P3 (FoxP3) is expressed in glioma cells and associated with increased apoptosis. Exp Cell Res 2012; 319:731-9. [PMID: 23211717 DOI: 10.1016/j.yexcr.2012.11.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 11/19/2012] [Accepted: 11/22/2012] [Indexed: 12/17/2022]
Abstract
The forkhead transcription factor FoxP3 is critically involved in the development and function of regulatory T cells (Tregs) that populate tumors and are considered as powerful parts of their immune evasion. However, also tumor cells are reported to express FoxP3. Since gliomas are particularly immunosuppressive tumors, we investigated the occurrence and possible functions of FoxP3 in these malignant cells. By quantitative RT-PCR, immunohistochemistry and FACS analysis, we detected FoxP3 in glioma cells in situ and in vitro. After exposure of glioma cell lines to chemotherapeutics, expression of FoxP3 was significantly enhanced, and it was dislocated from more nuclear to perinuclear localization. Overexpression of FoxP3 in glioma cell lines considerably favored apoptotic damage of nuclei, DNA fragmentation, increased cleavage of the pro-apoptotic enzyme poly(ADP-ribose) polymerase (PARP) and basal activities of effector caspases-3/7. In FoxP3-transfected cells, apoptotic stimuli like Camptothecin, Temozolomide or tumor necrosis factor-α synergistically enhanced caspases-3/7-activities over controls. Taking together, FoxP3 occurs in glioma cells, is induced by chemotherapeutics, and its expression is correlated with increased apoptosis of glioma cells, especially when propagated by apoptotic stimuli. Thus, FoxP3 is a novel pro-apoptotic transcription factor in gliomas that is critically involved in the action of apoptotic agents.
Collapse
Affiliation(s)
- Janka Held-Feindt
- Department of Neurosurgery, University Medical Center Schleswig-Holstein UKSH, Campus Kiel, 24105 Kiel, Germany
| | | | | | | | | | | |
Collapse
|
80
|
Ma GF, Chen SY, Sun ZR, Miao Q, Liu YM, Zeng XQ, Luo TC, Ma LL, Lian JJ, Song DL. FoxP3 inhibits proliferation and induces apoptosis of gastric cancer cells by activating the apoptotic signaling pathway. Biochem Biophys Res Commun 2012. [PMID: 23201402 DOI: 10.1016/j.bbrc.2012.11.065] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Forkhead Box Protein 3 (FoxP3) was identified as a key transcription factor to the occurring and function of the regulatory T cells (Tregs). However, limited evidence indicated its function in tumor cells. To elucidate the precise roles and underlying molecular mechanism of FoxP3 in gastric cancer (GC), we examined the expression of FoxP3 and the consequences of interfering with FoxP3 gene in human GC cell lines, AGS and MKN45, by multiple cellular and molecular approaches, such as immunofluorescence, gene transfection, CCK-8 assay, clone formation assay, TUNEL assay, Flow cytometry, immunoassay and quantities polymerase chain reaction (PCR). As a result, FoxP3 was expressed both in nucleus and cytoplasm of GC cells. Up-regulation of FoxP3 inhibited cell proliferation and promoted cell apoptosis. Overexpression of FoxP3 increased the protein and mRNA levels of proapoptotic molecules, such as poly ADP-ribose polymerase1 (PARP), caspase-3 and caspase-9, and repressed the expression of antiapoptotic molecules, such as cellular inhibitor of apoptosis-1 (c-IAP1) and the long isoform of B cell leukemia/lymphoma-2 (Bcl-2). Furthermore, silencing of FoxP3 by siRNA in GC cells reduced the expression of proapoptotic genes, such as PARP, caspase-3 and caspase-9. Collectively, our findings identify the novel roles of FoxP3 in inhibiting proliferation and inducing apoptosis in GC cells by regulating apoptotic signaling, which could be a promising therapeutic approach for gastric cancer.
Collapse
Affiliation(s)
- Gui-Fen Ma
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Wang Y, Yang S, Ni Q, He S, Zhao Y, Yuan Q, Li C, Chen H, Zhang L, Zou L, Shen A, Cheng C. Overexpression of forkhead box J2 can decrease the migration of breast cancer cells. J Cell Biochem 2012; 113:2729-37. [PMID: 22441887 DOI: 10.1002/jcb.24146] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The prognosis of breast cancer patients with metastases is generally poor, so it is essential to elucidate related molecules mechanisms. Forkhead Box J2 (FOXJ2) is a member of Forkhead Box transcription factors, many of which have been reported to participate in tumor migration and invasion. In this study, we showed the expression of FOXJ2 was higher in primary breast cancer tissues without lymph nodes metastases than those with, and there was statistical significance between the expression of FXOJ2 and the clinical factors. Hence, we identified a novel function of metastasis, which was not previously known for FOXJ2. Overexpression of FOXJ2 decreased the motility property of highly migrative MDA-MB-231 cells in vitro by wound healing assays and trans-well migration assays, and it was concurrent with the increased expression of epithelial marker E-cadherin and the decreased expression of mesenchymal marker vimentin by Western blot analysis, reverse transcription PCR analysis, and immunofluorescence analysis. Consistent with these observations, the repression of FOXJ2 in weakly metastatic MCF-7 cells remarkably promoted cellular motility. Our study demonstrates that FOXJ2 can inhibit the metastasis of human breast cancer by regulating the EMT key markers E-cadherin and vimentin.
Collapse
Affiliation(s)
- Yingying Wang
- Department of Immunology, Medical College, Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Triulzi T, Tagliabue E, Balsari A, Casalini P. FOXP3 expression in tumor cells and implications for cancer progression. J Cell Physiol 2012; 228:30-5. [DOI: 10.1002/jcp.24125] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
83
|
Frattini V, Pisati F, Speranza MC, Poliani PL, Frigé G, Cantini G, Kapetis D, Cominelli M, Rossi A, Finocchiaro G, Pellegatta S. FOXP3, a novel glioblastoma oncosuppressor, affects proliferation and migration. Oncotarget 2012; 3:1146-57. [PMID: 23888189 PMCID: PMC3717952 DOI: 10.18632/oncotarget.644] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 09/20/2012] [Indexed: 12/24/2022] Open
Abstract
The transcription factor FOXP3 plays an essential role in regulatory T cell development and function. In addition, it has recently been identified as a tumor suppressor in different cancers. Here, we report that FOXP3 is expressed in normal brain but strongly down-regulated in glioblastoma (GB) and in corresponding GB stem-like cells growing in culture as neurospheres (GB-NS), as evaluated by real time-PCR and confirmed by immunohistochemistry on an independent set of GB. FOXP3 expression was higher in low-grade gliomas than in GB. Interestingly, we also found that neurosphere generation, a feature present in 58% of the GB that we examined, correlated with lower expression of FOXP3 and shorter patient survival. FOXP3 silencing in one GB-NS expressing measurable levels of the gene caused a significant increase in proliferation and migration as well as highly aggressive growth in xenografts. Conversely, FOXP3 over-expression impaired GB-NS migration and proliferation in vitro. We also demonstrated using ChiP that FOXP3 is a transcriptional regulator of p21 and c-MYC supporting the idea that dysregulated expression of these factors is a major mechanism of tumorigenesis driven by the loss of FOXP3 expression in gliomas. These findings support the assertion that FOXP3 exhibits tumor suppressor activity in glioblastomas.
Collapse
Affiliation(s)
- Véronique Frattini
- Unit of Molecular Neuro-Oncology, Fondazione I.R.C.C.S. Istituto Neurologico C. Besta, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Chemotherapy sorting can be used to identify cancer stem cell populations. Mol Biol Rep 2012; 39:9955-63. [PMID: 22744428 DOI: 10.1007/s11033-012-1864-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 06/14/2012] [Indexed: 12/16/2022]
Abstract
Cancer stem cells (CSCs) of bladder transitional cell cancers (BTCC) had not been identified by the reported common methods. According to the phenomenon that CSCs were resistant to chemotherapy, BTCC cell lines T24 and 5637 were cultured with mitomycin C respectively. Cell inhibition assay revealed an increased population of drug resistant cancer cells with a concentration gradient of mitomycin C. The maximal and minimal cell inhibition rate in cell line T24 was 92.5 % ± 1.0 versus 64.1 % ± 1.4 (P < 0.001), and in cell line 5637 was 90.2 % ± 2.5 versus 55.1 % ± 1.8 (P < 0.001), respectively. There is no significant difference between these two groups. Drug resistant cells just comprised approximately 7.5 % (T24) versus 9.8 % (5637) of the total cells. Compared with control cells, cell cycle analysis demonstrated that more drug resistant cells were at G0G1 phase and fewer were at S phase with the concentration gradient of mitomycin C in both cell lines, which is in accord with the stem cell theory that most stem cells maintain in a quiescent condition. Importantly, we found that embryonic stem cell markers (OCT-4 and NANOG) were highly expressed in both gene and protein level in BTCC cell line T24 and 5637 after 24-h chemotherapy exposure. Interestingly, the drug concentration gradient was in accord with OCT-4 and NANOG expression, suggesting that chemotherapy sorting might be a feasible method for BTCC CSCs identification.
Collapse
|
85
|
Selenium promotes T-cell response to TCR-stimulation and ConA, but not PHA in primary porcine splenocytes. PLoS One 2012; 7:e35375. [PMID: 22530011 PMCID: PMC3328446 DOI: 10.1371/journal.pone.0035375] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 03/15/2012] [Indexed: 11/19/2022] Open
Abstract
There is controversy in the literature over whether the selenium (Se) influences cellular immune responses, and the mechanisms possibly underlying these effects are unclear. In this study, the effects of Se on T-cell proliferation and IL-2 production were studied in primary porcine splenocytes. Splenocytes were treated with different mitogens in the presence of 0.5-4 µmol/L sodium selenite. Se significantly promoted T-cell receptor (TCR) or concanavalin A (ConA)-induced T-cell proliferation and IL-2 production but failed to regulate T-cell response to phytohemagglutinin (PHA). In addition, Se significantly increased the levels of cytosolic glutathione peroxidase (GPx1) and thioredoxin reductase 1 (TR1) mRNA, the activity of GPx1 and the concentration of reduced glutathione (GSH) in the unstimulated, or activated splenocytes. These results indicated that Se improved the redox status in all splenocytes, including unstimulated, TCR, ConA and PHA -stimulated, but only TCR and ConA-induced T-cell activation was affected by the redox status. N-acetylcysteine (NAC), a pharmacological antioxidant, increased T-cell proliferation and IL-2 production by TCR and ConA stimulated splenocytes but had no effect on the response to PHA in primary porcine splenocytes confirming that PHA-induced T-cell activation is insensitive to the redox status. We conclude that Se promotes GPx1 and TR1 expression and increases antioxidative capacity in porcine splenocytes, which enhances TCR or ConA -induced T-cell activation but not PHA-induced T-cell activation. The different susceptibilities to Se between the TCR, ConA and PHA -induced T-cell activation may help to explain the controversy in the literature over whether or not Se boosts immune responses.
Collapse
|
86
|
Abstract
Cancer cells silence autosomal tumor suppressor genes by Knudson's two-hit mechanism in which loss-of-function mutations and then loss of heterozygosity occur at the tumor suppressor gene loci. However, the identification of X-linked tumor suppressor genes has challenged the traditional theory of 'two-hit inactivation' in tumor suppressor genes, introducing the novel concept that a single genetic hit can cause loss of tumor suppressor function. The mechanism through which these genes are silenced in human cancer is unclear, but elucidating the details will greatly enhance our understanding of the pathogenesis of human cancer. Here, we review the identification of X-linked tumor suppressor genes and discuss the potential mechanisms of their inactivation. In addition, we also discuss how the identification of X-linked tumor suppressor genes can potentially lead to new approaches in cancer therapy.
Collapse
Affiliation(s)
- Runhua Liu
- Division of Immunotherapy, Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA
- Department of Genetics, School of Medicine, University of Alabama at Birmingham and Comprehensive Cancer Center, Birmingham, AL, USA
| | - Mandy Kain
- Division of Immunotherapy, Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Lizhong Wang
- Division of Immunotherapy, Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA
- Department of Genetics, School of Medicine, University of Alabama at Birmingham and Comprehensive Cancer Center, Birmingham, AL, USA
| |
Collapse
|
87
|
Tan B, Behren A, Anaka M, Vella L, Cebon J, Mariadason JM, Chen W. FOXP3 is not mutated in human melanoma. Pigment Cell Melanoma Res 2012; 25:398-400. [DOI: 10.1111/j.1755-148x.2012.00993.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
88
|
Seo JM, Park S, Kim JH. Leukotriene B4 receptor-2 promotes invasiveness and metastasis of ovarian cancer cells through signal transducer and activator of transcription 3 (STAT3)-dependent up-regulation of matrix metalloproteinase 2. J Biol Chem 2012; 287:13840-9. [PMID: 22396544 DOI: 10.1074/jbc.m111.317131] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Ovarian cancer is the most lethal gynecologic malignancy in women. Despite the fact that the metastatic spread is associated with the majority of deaths from ovarian cancer, the molecular mechanisms regulating the invasive and metastatic phenotypes of ovarian cancer are poorly understood. In this study, we demonstrated that BLT2, a low affinity leukotriene B(4) receptor, is highly expressed in OVCAR-3 and SKOV-3 human ovarian cancer cells, and that this receptor plays a key role in the invasiveness and metastasis of these cells through activation of STAT3 and consequent up-regulation of matrix metalloproteinase 2 (MMP2). In addition, our results suggest that activation of NAD(P)H oxidase-4 (NOX4) and subsequent reactive oxygen species (ROS) generation lie downstream of BLT2, mediating the stimulation of STAT3-MMP2 cascade in this process. For example, knockdown of BLT2 or NOX4 using each specific siRNA suppressed STAT3 stimulation and MMP2 expression. Similarly, inhibition of STAT3 suppressed the expression of MMP2, thus leading to attenuated invasiveness of these ovarian cancer cells. Finally, the metastasis of SKOV-3 cells in nude mice was markedly suppressed by pharmacological inhibition of BLT2. Together, our results implicate a BLT2-NOX4-ROS-STAT3-MMP2 cascade in the invasiveness and metastasis of ovarian cancer cells.
Collapse
Affiliation(s)
- Ji-Min Seo
- School of Life Sciences and Biotechnology, Korea University, Seoul 136-701, Korea
| | | | | |
Collapse
|
89
|
Wang Z, Fukushima H, Inuzuka H, Wan L, Liu P, Gao D, Sarkar FH, Wei W. Skp2 is a promising therapeutic target in breast cancer. Front Oncol 2012; 1. [PMID: 22279619 PMCID: PMC3263529 DOI: 10.3389/fonc.2011.00057] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Breast cancer is the most common type of cancer among American women, and remains the second leading cause of cancer-related death for female in the United States. It has been known that several signaling pathways and various factors play critical roles in the development and progression of breast cancer, such as estrogen receptor, Notch, PTEN, human epidermal growth factor receptor 2, PI3K/Akt, BRCA1, and BRCA2. Emerging evidence has shown that the F-box protein S-phase kinase associated protein 2 (Skp2) also plays an important role in the pathogenesis of breast cancer. Therefore, in this brief review, we summarize the novel functions of Skp2 in the pathogenesis of breast cancer. Moreover, we provide further evidence regarding the state of our knowledge toward the development of novel Skp2 inhibitors especially natural "chemopreventive agents" as targeted approach for the prevention and/or treatment of breast cancer.
Collapse
Affiliation(s)
- Zhiwei Wang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | |
Collapse
|
90
|
He YH, Zhang HN, Zhang GP, Hou N, Xiao Q, Huang Y, Wu JH, Luo MS, Zhang GS, Yi Q, Chen MS, Luo JD. A physiological concentration of glucocorticoid inhibits the pro-inflammatory cytokine-induced proliferation of adult rat cardiac fibroblasts: Roles of extracellular signal-regulated kinase 1/2 and nuclear factor-κB. Clin Exp Pharmacol Physiol 2011; 38:739-46. [DOI: 10.1111/j.1440-1681.2011.05581.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
91
|
Lin SW, Chang CH, Lin CH. High-throughput Fluorescence Detections in Microfluidic Systems. ACTA ACUST UNITED AC 2011. [DOI: 10.1016/s2211-4254(11)60005-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
92
|
Katoh H, Zheng P, Liu Y. Signalling through FOXP3 as an X-linked tumor suppressor. Int J Biochem Cell Biol 2010; 42:1784-7. [PMID: 20678582 DOI: 10.1016/j.biocel.2010.07.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 06/11/2010] [Accepted: 07/11/2010] [Indexed: 12/27/2022]
Abstract
The FOXP3 (forkhead box P3) gene is a member of forkhead winged helix family transcription factors and functions as both a transcriptional activator and a repressor. FOXP3 dysfunction is responsible for an X-linked autoimmune syndrome: immune dysregulation, polyendopathy, enterophathy, X-linked syndrome. In addition to its role as an essential transcription factor in regulatory T cells, the FOXP3 gene is an epithelial cell-intrinsic tumor suppressor for breast and prostate cancers. We will focus on the FOXP3 signalling pathway in epithelial cells and discuss how genetic and/or epigenetic inactivation of the FOXP3 contributes to the malignant transformation of cells.
Collapse
Affiliation(s)
- Hiroto Katoh
- Division of Immunotherapy, Department of Surgery, University of Michigan School of Medicine and Cancer Center, Ann Arbor, MI 48109, USA
| | | | | |
Collapse
|
93
|
Liu Y, Wang L, Zheng P. X-linked tumor suppressors: perplexing inheritance, a unique therapeutic opportunity. Trends Genet 2010; 26:260-5. [PMID: 20434787 DOI: 10.1016/j.tig.2010.03.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Revised: 03/29/2010] [Accepted: 03/30/2010] [Indexed: 01/03/2023]
Abstract
Unlike autosomal genes, the majority of X-linked genes are subject to dosage compensation. As a result, female tissues comprise cells exclusively expressing X-linked genes from one or other parent. The implication of having only one allele of active X-linked genes in cancer pathogenesis, i.e. somatic single-hit inactivation and dominant inheritance, has not been extensively explored. Recent studies have identified FOXP3 and WTX as two X-linked tumor suppressor genes that are somatically inactivated by single genetic hits. Because the predicted dominant inheritance of cancer risk has not been demonstrated in humans, we will discuss the possible conditions that might prevent such dominant inheritance. We also argue that the existence of a genetically intact allele in cancer cells in women, together with apparent abnormal X inactivation in cancer cells, might provide an opportunity to selectively reactivate tumor suppressor genes for cancer therapy.
Collapse
Affiliation(s)
- Yang Liu
- Divisions of Immunotherapy, Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48105, USA.
| | | | | |
Collapse
|