1
|
Ibrahim A, Mohamady Farouk Abdalsalam N, Liang Z, Kashaf Tariq H, Li R, O Afolabi L, Rabiu L, Chen X, Xu S, Xu Z, Wan X, Yan D. MDSC checkpoint blockade therapy: a new breakthrough point overcoming immunosuppression in cancer immunotherapy. Cancer Gene Ther 2025; 32:371-392. [PMID: 40140724 PMCID: PMC11976280 DOI: 10.1038/s41417-025-00886-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/25/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025]
Abstract
Despite the success of cancer immunotherapy in treating hematologic malignancies, their efficacy in solid tumors remains limited due to the immunosuppressive tumor microenvironment (TME), which is mainly formed by myeloid-derived suppressor cells (MDSCs). MDSCs not only exert potent immunosuppressive effects that hinder the success of immune checkpoint inhibitors (ICIs) and adaptive cellular therapies, but they also promote tumor advancement through non-immunological pathways, including promoting angiogenesis, driving epithelial-mesenchymal transition (EMT), and contributing to the establishment of pre-metastatic environments. While targeting MDSCs alone or in combination with conventional therapies has shown limited success, emerging evidence suggests that MDSC checkpoint blockade in combination with other immunotherapies holds great promise in overcoming both immunological and non-immunological barriers. In this review, we discussed the dual roles of MDSCs, with a particular emphasis on their underexplored checkpoints blockade strategies. We discussed the rationale behind combination strategies, their potential advantages in overcoming MDSC-mediated immunosuppression, and the challenges associated with their development. Additionally, we highlight future research directions aimed at optimizing combination immunotherapies to enhance cancer therapeutic effectiveness, particularly in solid tumor therapies where MDSCs are highly prevalent.
Collapse
Affiliation(s)
- Abdulrahman Ibrahim
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
- University of Chinese Academy of Sciences, 100864, Beijing, China
| | - Nada Mohamady Farouk Abdalsalam
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
- University of Chinese Academy of Sciences, 100864, Beijing, China
| | - Zihao Liang
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Hafiza Kashaf Tariq
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
- University of Chinese Academy of Sciences, 100864, Beijing, China
| | - Rong Li
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Lukman O Afolabi
- Department of Pediatrics, Indiana University School of Medicine, 1234 Notre Dame Ave, South Bend, IN, 46617, USA
| | - Lawan Rabiu
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
- University of Chinese Academy of Sciences, 100864, Beijing, China
| | - Xuechen Chen
- College of Pharmacy, Jinan University, 511436, Guangzhou, China.
| | - Shu Xu
- Cancer Center, Shenzhen Guangming District People's Hospital, 518106, Shenzhen, China
| | - Zhiming Xu
- Cancer Center, Shenzhen Guangming District People's Hospital, 518106, Shenzhen, China.
| | - Xiaochun Wan
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China.
- University of Chinese Academy of Sciences, 100864, Beijing, China.
| | - Dehong Yan
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China.
- University of Chinese Academy of Sciences, 100864, Beijing, China.
| |
Collapse
|
2
|
Zhu L, Zhang X, Fu X, Li Z, Sun Z, Wu J, Wang X, Wang F, Li X, Niu S, Ding M, Yang Z, Yang W, Yin M, Zhang L, Zhang M. Retraction Note: TIPE2 suppresses progression and tumorigenesis of esophageal carcinoma via inhibition of the Wnt/β-catenin pathway. J Transl Med 2024; 22:977. [PMID: 39468634 PMCID: PMC11520825 DOI: 10.1186/s12967-024-05795-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024] Open
Affiliation(s)
- Linan Zhu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, 450052, Zhengzhou, Henan, People's Republic of China
| | - Xudong Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, 450052, Zhengzhou, Henan, People's Republic of China
| | - Xiaorui Fu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, 450052, Zhengzhou, Henan, People's Republic of China
| | - Zhaoming Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, 450052, Zhengzhou, Henan, People's Republic of China
| | - Zhenchang Sun
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, 450052, Zhengzhou, Henan, People's Republic of China
| | - Jingjing Wu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, 450052, Zhengzhou, Henan, People's Republic of China
| | - Xinhua Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, 450052, Zhengzhou, Henan, People's Republic of China
| | - Feng Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, 450052, Zhengzhou, Henan, People's Republic of China
| | - Xiangke Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, 450052, Zhengzhou, Henan, People's Republic of China
| | - Songtao Niu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, 450052, Zhengzhou, Henan, People's Republic of China
| | - Mengjie Ding
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, 450052, Zhengzhou, Henan, People's Republic of China
| | - Zhenzhen Yang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, 450052, Zhengzhou, Henan, People's Republic of China
| | - Wanqiu Yang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, 450052, Zhengzhou, Henan, People's Republic of China
| | - Meifeng Yin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, 450052, Zhengzhou, Henan, People's Republic of China
| | - Lei Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, 450052, Zhengzhou, Henan, People's Republic of China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, 450052, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
3
|
Tang Z, Zhang D, Yao C, Jiang M, Wang C, Chen Z, Yu H, Xue C, Liu Y, Shi Y, Zhang L, Wang X, Wei Z. TIPE2 inhibits the migration and invasion of epithelial ovarian cancer cells by targeting Smad2 to reverse TGF-β1-induced EMT. FASEB J 2024; 38:e70045. [PMID: 39259551 DOI: 10.1096/fj.202401427r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/19/2024] [Accepted: 08/29/2024] [Indexed: 09/13/2024]
Abstract
Epithelial ovarian cancer is the deadliest gynecologic malignancy, characterized by high metastasis. Transforming growth factor-β1 (TGF-β1) drives epithelial- mesenchymal transformation (EMT), a key process in tumor metastasis. Tumor necrosis factor-α-induced protein 8 (TNFAIP8)-like 2 (TIPE2) acts as a negative regulator of innate and adaptive immunity and involves in various cancers. However, its relationship with TGF-β1 in ovarian cancer and its role in reversing TGF-β1-induced EMT remain unclear. This study examined TIPE2 mRNA and protein expression using quantitative RT-PCR (qRT-PCR), western blot and immunohistochemistry. The effects of TIPE2 overexpression and knockdown on the proliferation, migration and invasion of epithelial ovarian cancer cells were assessed through 5-ethynyl-2-deoxyuridine, colony-forming, transwell migration and invasion assays. The relationship between TIPE2 and TGF-β1 was investigated using qRT-PCR and enzyme-linked immunosorbent assay, while the interaction between TIPE2 and Smad2 was identified via co-immunoprecipitation. The results revealed that TIPE2 protein was significantly down-regulated in epithelial ovarian cancer tissues and correlated with the pathological type of tumor, patients' age, tumor differentiation degree and FIGO stage. TIPE2 and TGF-β1 appeared to play an opposite role to each other during the progression of human ovarian cancer cells. Furthermore, TIPE2 inhibited the metastasis and EMT of ovarian cancer cells by combining with Smad2 in vitro or in an intraperitoneal metastasis model. Consequently, these findings suggest that TIPE2 plays a crucial inhibitory role in ovarian cancer metastasis by modulating the TGF-β1/Smad2/EMT signaling pathway and may serve as a potential target for ovarian cancer, providing important direction for future diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Zhongyun Tang
- Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
- Department of Gynecology and Obstetrics, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Derui Zhang
- Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
- Department of Gynecology and Obstetrics, Jinan Third People's Hospital, Jinan, Shandong, P.R. China
| | - Chenchen Yao
- Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
- Department of Gynecology and Obstetrics, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Mengmeng Jiang
- Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
- Department of Gynecology and Obstetrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Chongli Wang
- Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
- Department of Gynecology and Obstetrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Zhen Chen
- Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
- Department of Gynecology and Obstetrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Huayun Yu
- Department of Gynecology and Obstetrics, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Chenyue Xue
- Department of Gynecology and Obstetrics, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Yuqiu Liu
- Department of Gynecology and Obstetrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Yongyu Shi
- Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Lining Zhang
- Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Xiaoyan Wang
- Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Zengtao Wei
- Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
- Department of Gynecology and Obstetrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| |
Collapse
|
4
|
Xu L, Pan F, Guo Z. TIPE2: A Candidate for Targeting Antitumor Immunotherapy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:755-763. [PMID: 38377476 DOI: 10.4049/jimmunol.2300433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 12/18/2023] [Indexed: 02/22/2024]
Abstract
TNF-α-induced protein 8-like 2 (TIPE2 or TNFAIP8L2) is a recently discovered negative regulator of innate and adaptive immunity. TIPE2 is expressed in a wide range of tissues, both immune and nonimmune, and is implicated in the maintenance of immune homeostasis within the immune system. Furthermore, TIPE2 has been shown to play a pivotal role in the regulation of inflammation and the development of tumor. This review focuses on the structural characteristics, expression patterns, and functional roles of TIPE proteins, with a particular emphasis on the role and underlying mechanisms of TIPE2 in immune regulation and its involvement in different diseases. However, the current body of evidence is still limited in providing a comprehensive understanding of the complex role of TIPE2 in the human body, warranting further investigation to elucidate the possible mechanisms and functions of TIPE2 in diverse disease contexts.
Collapse
Affiliation(s)
- Luxia Xu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Feiyan Pan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zhigang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
5
|
Zhou Y, Xu L, Wang J, Ge B, Wang Q, Wang T, Liu C, Wei B, Wang Q, Gao Y. LRFN2 binding to NMDAR inhibits the progress of ESCC via regulating the Wnt/β-Catenin and NF-κB signaling pathway. Cancer Sci 2022; 113:3566-3578. [PMID: 35879265 PMCID: PMC9530863 DOI: 10.1111/cas.15510] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/12/2022] [Accepted: 07/19/2022] [Indexed: 11/29/2022] Open
Abstract
As a neuronal transmembrane protein, leucine-rich repeat and fibronectin type-III domain-containing protein 2 (LRFN2) can recruit and combine with N-methyl-D-aspartic acid receptors (NMDARs) to promote nerve growth. Genetic studies suggest that mutations in LRFN2 are associated with various cancers. However, the role and mechanism of LRFN2 in the progression of esophageal squamous cell carcinoma (ESCC) have not been elucidated. In this study, we demonstrated that LRFN2 was significantly downregulated in ESCC tissues by qRT-PCR and immunohistochemistry. Low LRFN2 expression was an adverse prognostic factor in patients with ESCC. Overexpression of LRFN2 effectively suppressed the proliferation, migration, invasion and epithelial-to-mesenchymal transition in vitro and tumor growth in vivo. Bioinformatics analysis indicated that Wnt/β-catenin signaling regulation was one of the most potential mechanisms and studies confirmed that overexpression of LFRN2 obviously downregulated the expression of β-catenin, c-Myc and cyclin D1 in ESCC cells and tumor tissues. Further studies revealed that LRFN2 plays anti-ESCC role by binding with NMDAR-GRIN2B and this effect can be weakened by NR2B-selective NMDA antagonist-NMDA-IN-1. Moreover, the bioinformatics analysis showed that the interaction of GRIN2B and GSK3β affects the NF-κB pathway, which was demonstrated by western blot experiments. Collectively, our results indicate that LRFN2 binding to NMDARs inhibits the progression of ESCC by regulating the Wnt/β-catenin and NF-κB pathway, which provides a new therapeutic target for improving the prognosis of patients with ESCC.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Medical Oncology, Cancer Center, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, China
| | - Lijuan Xu
- Department of Medical Oncology, Cancer Center, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, China
| | - Jiru Wang
- Department of Medical Oncology, Cancer Center, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, China
| | - Beibei Ge
- Department of Medical Oncology, Cancer Center, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, China
| | - Qiuzi Wang
- Department of Medical Oncology, Cancer Center, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, China
| | - Tao Wang
- Department of Medical Oncology, Cancer Center, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, China
| | - Chang Liu
- Department of Medical Oncology, Cancer Center, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, China
| | - Bin Wei
- Department of Medical Oncology, Cancer Center, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, China
| | - Qilong Wang
- Department of Central Laboratory, Cancer Center, The Affiliated Huaian No.1 People's Hospital, Nanjing Medical University, China
| | - Yong Gao
- Department of Medical Oncology, Cancer Center, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, China
| |
Collapse
|
6
|
The Function Mechanism of the Current Situation of Chinese Cultural Integration and Environment on the Development of Chinese Enterprises. JOURNAL OF ENVIRONMENTAL AND PUBLIC HEALTH 2022; 2022:4393337. [PMID: 35755885 PMCID: PMC9232369 DOI: 10.1155/2022/4393337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 05/26/2022] [Indexed: 12/04/2022]
Abstract
China's economy is booming and many Chinese enterprises are growing rapidly. The report card handed over by Chinese enterprises through practical actions proves that Chinese outstanding entrepreneurs have successful modern management experience. These Chinese entrepreneurs have experienced the test of practice, and the experience gained has Chinese characteristics and is worth promoting. Entrepreneurs carry out more business management practices, which also provides valuable reference resources for the study of Chinese management thought. At the same time, The Times is also calling for more Chinese entrepreneurs to form their own management ideas. The influence of the current situation of the Chinese cultural integration environment on the development of Chinese enterprises is mainly that a good cultural integration environment will promote the development of enterprises and provide preconditions for the development of Chinese enterprises. This paper studies the current situation of Chinese culture integration environment of Chinese entrepreneur management thought formation process; the main purpose is, through the analysis of Chinese outstanding entrepreneur management thought formation process, to reveal the Chinese culture integration management environment formation law and essential characteristics, in order to enrich the management cognitive research and Chinese entrepreneurs management thought research, publicity, and promotion of Chinese outstanding entrepreneurs form management thought and also to provide a reference for more practitioners.
Collapse
|
7
|
Zhang H, Han WJ, Zhang ZL. The Importance of Tumor Necrosis Factor-α-Induced Protein-8 Like-2 in the Pathogenesis of Cervical Cancer and Preeclampsia via Regulation of Cell Invasion. TOHOKU J EXP MED 2022; 257:181-191. [PMID: 35418534 DOI: 10.1620/tjem.2022.j026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Hong Zhang
- Department of Obstetrics, Yantaishan Hospital
| | - Wen-Jun Han
- Department of Gynecology, Qingdao Women and Children's Hospital
| | - Zhi-Lei Zhang
- Department of Gynecology, Qingdao Women and Children's Hospital
| |
Collapse
|
8
|
Jiang Y, Zhang Z, Wang X, Feng Z, Hong B, Yu D, Wang Y. A Novel Prognostic Factor TIPE2 in Bladder Cancer. Pathol Oncol Res 2022; 28:1610282. [PMID: 35388275 PMCID: PMC8978781 DOI: 10.3389/pore.2022.1610282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/01/2022] [Indexed: 11/13/2022]
Abstract
Objective: We sought to identify tumor necrosis factor (TNF)-alpha-induced protein 8-like 2 (TIPE2/TNFAIP8L2) expression in bladder cancer and its relationship to clinicopathological findings and prognosis. Methods: Immunohistochemical (IHC) staining for TIPE2 was performed on 110 archived radical cystectomy specimens. Ten high-power fields were randomly selected from each specimen to observe and record the percentage of immunoreactive cells of TIPE2 in tumor cells (grade 0–4) and the corresponding immunostaining intensity (grade 0–3). The expression score of TIPE2 was obtained by multiplying the results of the above two scores, which ranged from 0 to 12 points. The cut-off point of the sum of the scores were defined as follows: 0–3 scores were defined as negative expression (-); >3 scores were classified as positive expression, < 7, low expression, ≥7, high expression. Results: In 110 cases, TIPE2 was stained in various degrees in bladder cancer tissues, and expressed in both nucleus and cytoplasm. 4.5% (5/110) showed negative expression, 40.9% (45/110) showed low expression, and 54.5% (60/110) showed high expression. TIPE2 expression was negatively correlated with lymph node metastasis (p = 0.004) and disease progression (p = 0.021). Survival curves were plotted to show that patients with high TIPE2 expression had a progression-free survival curve above those with negative/low TIPE2 expression (p = 0.027). In multivariate Cox proportional hazard regression analysis, TIPE2 was a protective factor for progression-free survival in bladder urothelial carcinoma (p = 0.031), pT stage (p = 0.016) was a risk factor for progression-free survival, and age was a risk factor for overall survival (p = 0.020). Conclusion: TIPE2 may be a new biomarker to predict the disease progression and prognosis of patients with urothelial carcinoma of the bladder.
Collapse
Affiliation(s)
- Yu Jiang
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhiqiang Zhang
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xian Wang
- Department of Pathology, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhenzhong Feng
- Department of Pathology, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Bo Hong
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Dexin Yu
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yi Wang
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Provincial Institute of Translational Medicine, Hefei, China
| |
Collapse
|
9
|
Yang X, Zhang M, Su T, Tang S, Wang Y, Liu H, Wang P, Wang J, Pan X. TIPE2 Inhibits Migration and Promotes Apoptosis as a Tumor Suppressor in Hypopharyngeal Carcinoma. Curr Protein Pept Sci 2022; 23:424-436. [PMID: 35894468 DOI: 10.2174/1389203723666220727090317] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/23/2022] [Accepted: 05/23/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Hypopharyngeal squamous cell carcinoma (HSCC) is a common malignant cancer characterized by high metastasis and infiltration. The development of new approaches for the early diagnosis and identification of new therapeutic targets is essential. TIPE2 is well known as a tumor suppressor and related to a favorable prognosis of HSCC. However, its underlying mechanism remains unclear. METHODS AND MATERIALS TIPE2 expression was determined by immunohistochemistry and RT-qPCR. A TIPE2 overexpression stable cell line was generated by lentivirus infection. TIPE2 and other related protein levels were detected by western blotting. The cell cycle and apoptosis were performed by flow cytometric analysis. Cell proliferation was measured with a Cell Counting Kit-8 (CCK-8) assay, and the activity of caspase-3 and caspase-7 was assessed by Caspase-Glo® 3/7 Assay. All data were analyzed with SPSS 25 and GraphPad Prism 8.0. RESULTS TIPE2 expression was significantly down-regulated in HSCC. Low TIPE2 expression may be associated with poor prognosis in HSCC. TIPE2 overexpression markedly inhibited tumor cell migration. Moreover, TIPE2 decreased cell proliferation but promoted apoptosis. TIPE2 suppressed tumor growth by activating Epithelial-Mesenchymal Transition (EMT) and the extrinsic apoptosis pathway. CONCLUSION TIPE2 inhibited tumor progression by suppressing cell migration but promoting apoptosis. TIPE2 can be a new therapeutic target in HSCC.
Collapse
Affiliation(s)
- Xiaoqi Yang
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
- NHC Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, Shandong 250012, China
| | - Minfa Zhang
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
- NHC Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, Shandong 250012, China
| | - Tongdong Su
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
- NHC Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, Shandong 250012, China
| | - Shuangmei Tang
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
- NHC Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, Shandong 250012, China
| | - Yin Wang
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
- NHC Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, Shandong 250012, China
| | - Heng Liu
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
- NHC Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, Shandong 250012, China
| | - Pin Wang
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
- NHC Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, Shandong 250012, China
| | - Juan Wang
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
- NHC Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, Shandong 250012, China
| | - Xinliang Pan
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
- NHC Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, Shandong 250012, China
| |
Collapse
|
10
|
Feng F, Liu C, Bian H, Cai W, Zhou Y, Zhou L, Zhuang Z. TIPE2 Suppresses Malignancy of Pancreatic Cancer Through Inhibiting TGFβ1 Mediated Signaling Pathway. Front Oncol 2021; 11:680985. [PMID: 34249724 PMCID: PMC8260882 DOI: 10.3389/fonc.2021.680985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/01/2021] [Indexed: 12/04/2022] Open
Abstract
Pancreatic cancer is one of the major reasons of cancer-associated deaths due to poor diagnosis, high metastasis and drug resistance. Therefore, it is important to understand the cellular and molecular mechanisms of pancreatic cancer to identify new targets for the treatment. TIPE2 is an essential regulator of tumor apoptosis, inflammation and immune homeostasis. However, the role of TIPE2 is still not fully understood in pancreatic cancer. In this study, we found the expression of TIPE2 was decreased in pancreatic cancer tissues compare to paracancerous tissues, which was negatively correlated with tumor size in patients. Overexpression of TIPE2 significantly decreased cell proliferation, metastasis and increased apoptotic events in pancreatic cancer cell lines. Moreover, the results obtained from real time PCR and western blot revealed that TIPE2 was also involved in inhibiting MMPs and N-Cadherin expression while increasing Bax expression in pancreatic cancer cells. Similarly, TIPE2 could inhibit tumor growth in vivo, decrease the expression of Ki-67 and N-Cadherin, and increase the expression of Bax by IHC analysis in tumor tissues isolated from tumor-bearing mice. Mechanistic studies exhibited that TIPE2 might suppress pancreatic cancer development through inhibiting PI3K/AKT and Raf/MEK/ERK signaling pathways triggered by TGFβ1. Moreover, the tumor-infiltrating lymphocytes from tumor-bearing mice were analyzed by flow cytometry, and showed that TIPE2 could promote T cell activation to exert an anti-tumor effect possibly through activation of DCs in a TGFβ1 dependent manner. In general, we described the multiple regulatory mechanisms of TIPE2 in pancreatic tumorigenesis and tumor microenvironment, which suggested TIPE2 may act as a potential therapeutic target in pancreatic cancer.
Collapse
Affiliation(s)
- Fang Feng
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Medical College, Soochow University, Suzhou, China.,Department of Oncology, Suzhou Ninth People's Hospital, Soochow University, Suzhou, China
| | - Chunliang Liu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Huahui Bian
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Medical College, Soochow University, Suzhou, China
| | - Wei Cai
- Department of Oncology, Suzhou Ninth People's Hospital, Soochow University, Suzhou, China
| | - Ying Zhou
- Department of Oncology, Suzhou Ninth People's Hospital, Soochow University, Suzhou, China
| | - Li Zhou
- Department of Oncology, Suzhou Ninth People's Hospital, Soochow University, Suzhou, China
| | - Zhixiang Zhuang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Medical College, Soochow University, Suzhou, China
| |
Collapse
|
11
|
Xu Y, Wang N, Liu R, Lv H, Li Z, Zhang F, Gai C, Tian Z. Epigenetic Study of Esophageal Carcinoma Based on Methylation, Gene Integration and Weighted Correlation Network Analysis. Onco Targets Ther 2021; 14:3133-3149. [PMID: 34012270 PMCID: PMC8128498 DOI: 10.2147/ott.s298620] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 04/13/2021] [Indexed: 12/16/2022] Open
Abstract
Purpose Esophageal carcinoma is a common and highly metastatic malignant tumor of the digestive tract. The aim of the present study was to identify potential molecular markers of esophageal carcinoma that may help its diagnosis and treatment. Materials and Methods First, mRNA and DNA methylation data were downloaded from The Cancer Genome Atlas (TCGA) database for the identification of differentially expressed genes (DEGs) and DNA methylation analysis. Secondly, Weighted Gene Co-Expression Network Analysis (WGCNA) was used to identify important modules and hub genes. In addition, correlation analysis between DNA methylation genes and DEGs was performed. Thirdly, the GSE45670 dataset was used to validate the expression of the diagnostic and survival ability analysis of genes in TCGA data. Finally, reverse transcription-quantitative PCR and immunohistochemical analysis of genes were performed. Results A total of 2408 DEGs and 5134 differentially methylated sites were obtained. In the WGCNA analysis, the royal blue module was found to be the optimal module. In addition, hub genes in the module, including ESRRG, MFSD4, CCKBR, ATP4B, ESRRB, ATP4A, CCKAR and B3GAT1, were also differentially methylated genes and DEGs. It was found that CCKAR, MFSD4 and ESRRG may be diagnostic gene biomarkers for esophageal carcinoma. In addition, the high expression of MFSD4 was significantly correlated with patient survival. Immunohistochemistry analysis results showed that the gene expression levels of ATP4B, B3GAT1, CCKBR and ESRRG were decreased in esophageal carcinoma tissues, which was in line with the bioinformatics results. Conclusion Therefore, these identified molecular markers may be helpful in the diagnosis and treatment of esophageal carcinoma.
Collapse
Affiliation(s)
| | - Na Wang
- Department of Cancer Institute
| | - Rongfeng Liu
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, People's Republic of China
| | | | | | | | | | | |
Collapse
|
12
|
Gu Z, Cui X, Sun P, Wang X. Regulatory Roles of Tumor Necrosis Factor-α-Induced Protein 8 Like-Protein 2 in Inflammation, Immunity and Cancers: A Review. Cancer Manag Res 2020; 12:12735-12746. [PMID: 33364825 PMCID: PMC7751774 DOI: 10.2147/cmar.s283877] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/15/2020] [Indexed: 12/12/2022] Open
Abstract
Tumor necrosis factor-alpha (TNF-α)-induced protein 8 (TNFAIP8/TIPE) family, including TNFAIP8 (TIPE), TNFAIP8 like-protein 1 (TNFAIP8L1/TIPE1), TNFAIP8 like-protein 2 (TNFAIP8L2/TIPE2), and TNFAIP8 like-protein 3 (TNFAIP8L3/TIPE3), plays a vital role in regulating inflammatory responses, immune homeostasis, and cancer development. Over the last decade, studies have shown that TIPE2 protein is differentially expressed in diverse cells and tissues. The dysregulation of TIPE2 protein can lead to dysregulation of inflammatory responses and immune homeostasis, and change the basic characteristics of cancers. In consideration of the immeasurable values of TIPE2 in diagnosis, treatment, and prognosis of various human diseases, this review will focus on the expression pattern, structure, and regulatory roles of TIPE2 in inflammation, immunity, and cancers.
Collapse
Affiliation(s)
- Zhengzhong Gu
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| | - Xiaohan Cui
- Nanjing Medical University, Nanjing, Jiangsu Province, People's Republic of China
| | - Pengda Sun
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| | - Xudong Wang
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| |
Collapse
|
13
|
Huang H, Fan X, Zhang X, Xie Y, Ji Z. LncRNA CARLo-7 facilitates proliferation, migration, invasion, and EMT of bladder cancer cells by regulating Wnt/β-catenin and JAK2/STAT3 signaling pathways. Transl Androl Urol 2020; 9:2251-2261. [PMID: 33209690 PMCID: PMC7658127 DOI: 10.21037/tau-20-1293] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Background Aberrant expression of long noncoding RNAs (lncRNAs) has been found to enroll in the initiation and progression of bladder cancer (BC). Earlier results show cancer-associated region long noncoding RNA-7 (CARLo-7) can be a prognostic marker for BC, but its biological function and the underlying mechanism is still to be discovered. Our study aims to explore the effects of CARLo-7 on the initiation and progression of BC and the potential mechanisms. Methods The expression of CARLo-7 in BC tissues and cell lines was determined by quantitative real-time polymerase chain reaction (qRT-PCR). T24 and HT1197 cells were transfected with CARLo-7 expression vector or sh-CARLo-7, then cell viability assay, BrdU assay, flow cytometry, Transwell cell migration, and invasion assay, and western blot were conducted to evaluate cell proliferation, apoptosis, invasion, migration, and epithelial-mesenchymal transition (EMT). Results CARLo-7 was dramatically upregulated in BC tissues and cell lines. Silencing CARLo-7 by sh-CARLo-7 significantly suppressed proliferation and induced apoptosis of BC cells, while enforced CARLo-7 expression promoted cell proliferation. Meanwhile, silencing CARLo-7 attenuated migration, invasion, and EMT of BC cells, while CARLo-7 overexpression had the contrary effects. The β-catenin, p-JAK2 and p-STAT3 levels were decreased by CARLo-7 knockdown, while activation of Wnt/β-catenin or JAK2/STAT3 pathways abolished the effects of CARLo-7 knockdown on cell proliferation and migration. Conclusions Collectively, CARLo-7 plays a critical role in regulating BC development by regulating cell proliferation, migration, invasion, and EMT through Wnt/β-catenin and JAK2/STAT3 signaling. Therefore, CARLo-7 might be a promising therapeutic target for BC.
Collapse
Affiliation(s)
- Houfeng Huang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinrong Fan
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuebin Zhang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Xie
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhigang Ji
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
14
|
Xu Z, Lu W, Miao Y, Li H, Xie X, Zhang F. mRNA profiling reveals the potential mechanism of TIPE2 in attenuating cognitive deficits in APP/PS1 mice. Int Immunopharmacol 2020; 87:106792. [DOI: 10.1016/j.intimp.2020.106792] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 06/17/2020] [Accepted: 07/06/2020] [Indexed: 01/08/2023]
|
15
|
Yang S, Li X, Shen W, Hu H, Li C, Han G. MicroRNA-140 Represses Esophageal Cancer Progression via Targeting ZEB2 to Regulate Wnt/β-Catenin Pathway. J Surg Res 2020; 257:267-277. [PMID: 32862055 DOI: 10.1016/j.jss.2020.07.074] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 06/11/2020] [Accepted: 07/11/2020] [Indexed: 01/18/2023]
Abstract
BACKGROUND MicroRNAs have been reported to play regulatory functions in various cancers, including esophageal cancer. The aim of this study was to investigate the effects of miR-140 on the progression of esophageal cancer and the underlying regulatory mechanism. METHODS The levels of miR-140 and zinc finger E-box-binding homeobox 2 (ZEB2) messenger RNA in esophageal cancer tissues and cell lines were measured by quantitative real-time polymerase chain reaction. The protein levels of ZEB2, β-catenin, c-Myc, and cyclinD1 were determined by Western blot. Cell proliferation and apoptosis were determined by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide assay and flow cytometry, respectively. Cell migration and invasion were assessed by transwell assay. In addition, the relationship between miR-140 and ZEB2 was predicted by TargetScan online database and confirmed by dual-luciferase reporter assay. The tumor xenograft model was used to verify the role of miR-140 in esophageal cancer progression in vivo. RESULTS The expression of miR-140 was downregulated whereas ZEB2 expression was upregulated in esophageal cancer tissues compared with paracancerous normal tissues. Functionally, both miR-140 overexpression and ZEB2 knockdown inhibited proliferation, migration, and invasion and induced apoptosis in esophageal cancer cells. ZEB2 overexpression reversed the effects of miR-140 on proliferation, apoptosis, migration, and invasion of esophageal cancer cells. Mechanistically, ZEB2 was identified as a target of miR-140. Furthermore, miR-140 suppressed Wnt/β-catenin pathway by regulating ZEB2 expression in esophageal cancer cells. MiR-140 inhibited tumor growth of esophageal cancer through repressing ZEB2 expression in vivo. CONCLUSIONS Our results demonstrated that miR-140 inhibited esophageal cancer development by targeting ZEB2 through inactivating Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Song Yang
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, China
| | - Xiangyi Li
- Department of Endocrinology, Taizhou People's Hospital, Taizhou, Jiangsu, China
| | - Wenhao Shen
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, China
| | - Haitao Hu
- Clinical Laboratory, Taizhou People's Hospital, Taizhou, Jiangsu, China
| | - Chen Li
- Department of Stomatology, Taizhou People's Hospital, Taizhou, Jiangsu, China
| | - Gaohua Han
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, China.
| |
Collapse
|
16
|
Zhao LL. TIPE2 suppresses progression and tumorigenesis of the oral tongue squamous cell carcinoma by regulating FoxP3 + regulatory T cells. J Bioenerg Biomembr 2020; 52:279-289. [PMID: 32594290 DOI: 10.1007/s10863-020-09840-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/01/2020] [Indexed: 12/21/2022]
Abstract
To discover the effect of tumor necrosis factor-alpha-induced protein 8-like 2 (TIPE2) on the oral tongue squamous cell carcinoma (OTSCC) via affecting FoxP3+ regulatory T (Treg) cells. Immunohistochemistry was conducted to examine TIPE2 and FoxP3 expressions in OTSCC tumor tissues and corresponding oral mucosa. Tca8113 cells were transfected with TIPE2/control lentiviral activation particles followed by the detection with qRT-PCR, Western blotting, MTT assay, Wound healing, Transwell assay and Annexin V-FITC/PI staining. In vivo experiment was carried out on the nude mice xenografts of OTSCC with TIPE2 overexpression to observe the tumor volume and survival, and the CD4+ T cell subgroups were detected by flow cytometry. TIPE2 was lower in the OTSCC tissues than the corresponding oral mucosa, which was correlated with T stage, N stage, TNM stage, and differentiation of patients. Patients with TIPE2-positive expression had worse prognosis and lower expression of FoxP3+ Treg cells than the negative ones. Furthermore, TIPE2 overexpression curbed proliferation, invasion and migration of Tca8113 cells, while cell apoptosis was increased. Besides, TIPE2 suppressed the tumor growth and extended the survival of OTSCC mice, with the decreased proportion of FoxP3+ Treg cells in the spleen and tumor-infiltrated lymphocytes (TILs). The clinical results showed the down-regulation of TIPE2 in OTSCC tissues. Meanwhile TIPE2 overexpression affected OTSCC cells biological behavior in vitro, as well as exhibited strong tumor-growth suppressive effects in vivo, which may be a potential therapeutic target in OTSCC via regulating FoxP3+ Treg cells.
Collapse
Affiliation(s)
- Li-Li Zhao
- Department of Stomatology, Caoxian people's Hospital, Heze City, 274400, Shandong Province, China.
| |
Collapse
|
17
|
Tang DR, Li CL, Xu KP, Wu QQ, Chen QY, Lv JJ, Ji J, Zang B, Chen C, Gu B, Zhao JQ. Pigment Epithelium-Derived Factor Promotes the Growth and Migration of Human Esophageal Squamous Cell Carcinoma. Front Oncol 2020; 9:1520. [PMID: 32010619 PMCID: PMC6978803 DOI: 10.3389/fonc.2019.01520] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 12/17/2019] [Indexed: 11/13/2022] Open
Abstract
Pigment epithelium-derived factor (PEDF) is an oncogene found in various types of cancers. However, how PEDF affects the development of human esophageal squamous cell carcinoma (ESCC) is unknown. This study investigates the role of PEDF in ESCC cell proliferation, migration, and cell cycle both in vitro and in vivo. The PEDF expression was examined in patient tumor samples and ESCC cell lines. Short hairpin RNA technology was used to inhibit the PEDF expression in ESCC EC9706 and KYSE150 cells. In vitro cell proliferation and migration assays were performed. The effects of PEDF on tumor growth and progression were examined in vivo in murine subcutaneous xenograft tumor models. It was found that PEDF was overexpressed in esophageal cancer cells and patient tumor tissues compared to normal control samples. PEDF enhanced cell cycle progression and inhibited cell apoptosis. Knock down of PEDF inhibited esophageal cell proliferation and migration in vitro. Moreover, Inhibition of PEDF significantly reduced tumor growth and tumor size in vivo. These results indicate that PEDF induce tumorigenesis in ESCC and can be a potential therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- De-Rong Tang
- Department of Thoracic Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Cheng-Lin Li
- Department of Thoracic Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Ke-Ping Xu
- Department of Thoracic Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Qing-Quan Wu
- Department of Thoracic Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Qi-You Chen
- Department of Thoracic Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Jun-Jie Lv
- Department of Thoracic Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Jian Ji
- Department of Thoracic Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Bao Zang
- Department of Thoracic Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Chen Chen
- Department of Thoracic Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Biao Gu
- Department of Thoracic Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Jian-Qiang Zhao
- Department of Thoracic Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| |
Collapse
|
18
|
Bie YN, Gu P, Chen YT, Zhou XX, Tian YG, Yang Q, Li HY, Lin X, Guan YH, Lin TY, Lu X, Shen HF, Fang TX, Liu YM, Xiao D, Gu WW. TZAP plays an inhibitory role in the self-renewal of porcine mesenchymal stromal cells and is implicated the regulation of premature senescence via the p53 pathway. J Transl Med 2019; 17:72. [PMID: 30845965 PMCID: PMC6404308 DOI: 10.1186/s12967-019-1820-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Accepted: 02/21/2019] [Indexed: 01/01/2023] Open
Abstract
Background Mesenchymal stromal cells (MSCs) were originally characterized by the ability to differentiate into different mesenchymal lineages in vitro, and their immunomodulatory and trophic functions have recently aroused significant interest in the application of MSCs in cell-based regenerative medicine. However, a major problem in clinical practice is the replicative senescence of MSCs, which limits the cell proliferation potential of MSCs after large-scale expansion. Telomeric zinc finger-associated protein (TZAP), a novel specific telomere-binding protein, was recently found to stimulate telomere trimming and prevent excessive telomere elongation. The aim of this study was to elucidate the role of TZAP in regulating MSCs senescence, differentiation and proliferation. Method Primary porcine mesenchymal stromal cells (pMSCs) were isolated from the bone marrow of Tibet minipigs by a noninvasive method in combination with frequent medium changes (FMCs). The deterioration of the pMSCs’ proliferation capacity and their resultant entry into senescence were analyzed by using CCK8 and EdU incorporation assays, SA-β-gal staining and comparisons of the expression levels of cellular senescence markers (p16INK14 and p21) in pMSC cell lines with TZAP overexpression or knockout. The effects of TZAP overexpression or knockout on the differentiation potential of pMSCs were assessed by alizarin red S staining after osteogenic induction or by oil red O staining after adipogenic induction. The effect of TZAP overexpression and the involvement of the p53 signaling pathway were evaluated by detecting changes in ARF, MDM2, P53 and P21 protein levels in pMSCs. Results TZAP levels were significantly elevated in late-passage pMSCs compared to those in early-passage pMSCs. We also observed significantly increased levels of the senescence markers p16INK4A and p21. Overexpression of TZAP reduced the differentiation potential of the cells, leading to premature senescence in early-passage pMSCs, while knockout of TZAP led to the opposite phenotype in late-passage pMSCs. Furthermore, overexpression of TZAP activated the P53 pathway (ARF-MDM2-P53-P21WAF/CDKN1A) in vitro. TZAP also downregulated the expression levels of PPARγ and Cebpα, two key modulators of adipogenesis. Conclusions This study demonstrates that the level of TZAP is closely related to differentiation potential in pMSCs and affects cellular senescence outcomes via the p53 pathway. Therefore, attenuation of intracellular TZAP levels could be a new strategy for improving the efficiency of pMSCs in cell therapy and tissue engineering applications. Electronic supplementary material The online version of this article (10.1186/s12967-019-1820-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ya-Nan Bie
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou, China.,Songshan Lake Pearl Laboratory Animal Sci & Tech. Co., Ltd., Dongguan, China
| | - Peng Gu
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou, China.,Songshan Lake Pearl Laboratory Animal Sci & Tech. Co., Ltd., Dongguan, China
| | - Yu-Ting Chen
- Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xiao-Xu Zhou
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou, China.,Songshan Lake Pearl Laboratory Animal Sci & Tech. Co., Ltd., Dongguan, China
| | - Yu-Guang Tian
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou, China
| | - Qin Yang
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hai-Yan Li
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou, China
| | - Xia Lin
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou, China
| | - Yan-Hong Guan
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou, China
| | - Tao-Yan Lin
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou, China
| | - Xun Lu
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou, China
| | - Hong-Fen Shen
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou, China
| | - Ting-Xiao Fang
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou, China
| | - Yu-Min Liu
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou, China
| | - Dong Xiao
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou, China.
| | - Wei-Wang Gu
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou, China. .,Songshan Lake Pearl Laboratory Animal Sci & Tech. Co., Ltd., Dongguan, China.
| |
Collapse
|
19
|
Wu DD, Liu SY, Gao YR, Lu D, Hong Y, Chen YG, Dong PZ, Wang DY, Li T, Li HM, Ren ZG, Guo JC, He F, Ren XQ, Sun SY, Duan SF, Ji XY. Tumour necrosis factor-α-induced protein 8-like 2 is a novel regulator of proliferation, migration, and invasion in human rectal adenocarcinoma cells. J Cell Mol Med 2019; 23:1698-1713. [PMID: 30637920 PMCID: PMC6378198 DOI: 10.1111/jcmm.14065] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/07/2018] [Accepted: 11/08/2018] [Indexed: 12/11/2022] Open
Abstract
Tumour necrosis factor‐α‐induced protein 8‐like 2 (TIPE2) is a tumour suppressor in many types of cancer. However, the mechanism of action of TIPE2 on the growth of rectal adenocarcinoma is unknown. Our results showed that the expression levels of TIPE2 in human rectal adenocarcinoma tissues were higher than those in adjacent non‐tumour tissues. Overexpression of TIPE2 reduced the proliferation, migration, and invasion of human rectal adenocarcinoma cells and down‐regulation of TIPE2 showed reverse effects. TIPE2 overexpression increased apoptosis through down‐regulating the expression levels of Wnt3a, phospho (p)‐β‐Catenin, and p‐glycogen synthase kinase‐3β in rectal adenocarcinoma cells, however, TIPE2 knockdown exhibited reverse trends. TIPE2 overexpression decreased autophagy by reducing the expression levels of p‐Smad2, p‐Smad3, and transforming growth factor‐beta (TGF‐β) in rectal adenocarcinoma cells, however, TIPE2 knockdown showed opposite effects. Furthermore, TIPE2 overexpression reduced the growth of xenografted human rectal adenocarcinoma, whereas TIPE2 knockdown promoted the growth of rectal adenocarcinoma tumours by modulating angiogenesis. In conclusion, TIPE2 could regulate the proliferation, migration, and invasion of human rectal adenocarcinoma cells through Wnt/β‐Catenin and TGF‐β/Smad2/3 signalling pathways. TIPE2 is a potential therapeutic target for the treatment of rectal adenocarcinoma.
Collapse
Affiliation(s)
- Dong-Dong Wu
- School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, China.,Joint National Laboratory for Antibody Drug Engineering, Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Shi-Yu Liu
- School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, China.,Joint National Laboratory for Antibody Drug Engineering, Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Ying-Ran Gao
- School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, China.,Joint National Laboratory for Antibody Drug Engineering, Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Dan Lu
- School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, China.,Joint National Laboratory for Antibody Drug Engineering, Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Ya Hong
- School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, China.,Joint National Laboratory for Antibody Drug Engineering, Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Ya-Ge Chen
- School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, China.,Joint National Laboratory for Antibody Drug Engineering, Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Peng-Zhen Dong
- School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, China.,Joint National Laboratory for Antibody Drug Engineering, Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Da-Yong Wang
- Joint National Laboratory for Antibody Drug Engineering, Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China.,The First Affiliated Hospital of Henan University, Kaifeng, China
| | - Tao Li
- School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, China.,Joint National Laboratory for Antibody Drug Engineering, Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Hui-Min Li
- School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, China.,Joint National Laboratory for Antibody Drug Engineering, Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Zhi-Guang Ren
- School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, China.,Joint National Laboratory for Antibody Drug Engineering, Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Jian-Cheng Guo
- Center for Precision Medicine, Zhengzhou University, Zhengzhou, China
| | - Fei He
- Huaihe Hospital of Henan University, Kaifeng, China
| | - Xue-Qun Ren
- Huaihe Hospital of Henan University, Kaifeng, China
| | - Shi-Yong Sun
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia
| | - Shao-Feng Duan
- Joint National Laboratory for Antibody Drug Engineering, Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China.,Institute for Innovative Drug Design and Evaluation, Henan University School of Pharmacy, Kaifeng, China
| | - Xin-Ying Ji
- School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, China.,Joint National Laboratory for Antibody Drug Engineering, Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| |
Collapse
|
20
|
Gao S, Lu A, Amra S, Guo P, Huard J. TIPE2 gene transfer with adeno-associated virus 9 ameliorates dystrophic pathology in mdx mice. Hum Mol Genet 2019; 28:1608-1619. [DOI: 10.1093/hmg/ddz001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 12/12/2018] [Accepted: 12/31/2018] [Indexed: 12/23/2022] Open
Affiliation(s)
- Shanshan Gao
- Department of Orthopedic Surgery, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Aiping Lu
- Department of Orthopedic Surgery, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
- Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO, USA
| | - Sarah Amra
- Department of Orthopedic Surgery, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Ping Guo
- Department of Orthopedic Surgery, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
- Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO, USA
| | - Johnny Huard
- Department of Orthopedic Surgery, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
- Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO, USA
| |
Collapse
|
21
|
Zhao W, Yang S, Chen J, Zhao J, Dong J. Forced overexpression of FBP1 inhibits proliferation and metastasis in cholangiocarcinoma cells via Wnt/β-catenin pathway. Life Sci 2018; 210:224-234. [PMID: 30193944 DOI: 10.1016/j.lfs.2018.09.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 08/30/2018] [Accepted: 09/03/2018] [Indexed: 02/07/2023]
Abstract
AIM To investigate the effect of fructose-1,6-bisphosphatase 1 (FBP1) on the malignant phenotypes of cholangiocarcinoma (CCA) cells, and to explore the underlying mechanism. MAIN METHODS The expression of FBP1 in clinical CCA tissues was detected by real-time PCR, Western blot and immunohistochemistry staining. FBP1 was overexpressed by transfection of a forced expression plasmid. MTT, plate colony formation assay, Hoechst staining, flow cytometry, Western blot, wound healing, transwell assays and xenograft were performed to detect the growth, proliferation, cell cycle, apoptosis, migration, invasion and tumorigenesis in RBE and HCCC-9801 cells. In addition, the Wnt/β-catenin signaling was detected. KEY FINDINGS FBP1 was downregulated in clinical CCA specimens and cell lines, compared to paired para-carcinoma tissues or normal cholangetic epithelial cells. Gain-of-function experiments demonstrated that the forced expression of FBP1 inhibited the proliferation, colony formation, and blocked cell cycle of RBE and HCCC-9801 cells. Apoptosis of CCA cells was significantly enhanced by FBP1 overexpression, evidenced by upregulation of cleaved caspase-3, cleaved PARP and Bax levels, while downregulation of Bcl-2 level. Moreover, overexpression of FBP1 decreased the migratory and invasive ability in RBE and HCCC-9801 cells. However, FBP1-induced phenotypic changes were eliminated by overexpression of β-catenin. Finally, the forced overexpression of FBP1 inhibited tumorigenesis in vivo. SIGNIFICANCE Our findings demonstrate that FBP1 is downregulated in CCA tissues and cell lines, and the overexpression of FBP1 inhibits the proliferation, migration, invasion and tumorigenesis of CCA cells partly via inactivation of Wnt/β-catenin pathway. FBP1 may be a novel early diagnosis marker and therapeutic target for CCA.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China; Department of Hepatopancreatobiliary Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| | - Shizhong Yang
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, Beijing 102218, People's Republic of China
| | - Jianfeng Chen
- Department of Hepatobiliary Surgery, 401 Hospital of Chinese PLA, Qingdao 266071, People's Republic of China
| | - Jing Zhao
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| | - Jiahong Dong
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China; Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, Beijing 102218, People's Republic of China.
| |
Collapse
|
22
|
Park B, Lim JW, Kim H. Lycopene treatment inhibits activation of Jak1/Stat3 and Wnt/β-catenin signaling and attenuates hyperproliferation in gastric epithelial cells. Nutr Res 2018; 70:70-81. [PMID: 30098838 DOI: 10.1016/j.nutres.2018.07.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 07/05/2018] [Accepted: 07/19/2018] [Indexed: 12/19/2022]
Abstract
Helicobacter pylori (H pylori) colonizes the human stomach and increases the risk of gastric diseases including gastric cancer. H pylori increases reactive oxygen species (ROS), which activate Janus-activator kinase 1 (Jak1)/signal transducers and activators of transcription 3 (Stat3) in gastric epithelial cells. ROS mediate hyperproliferation, a hallmark of carcinogenesis, by activating Wnt/β-catenin signaling in various cells. Lycopene is a potent antioxidant exhibiting anticancer effects. We hypothesized that lycopene may inhibit H pylori-induced hyperproliferation by suppressing ROS-mediated activation of Jak1/Stat3 and Wnt/β-catenin signaling, and β-catenin target gene expression in gastric epithelial cells. We determined cell viability, ROS levels, and the protein levels of phospho- and total Jak1/Stat3, Wnt/β-catenin signaling molecules, Wnt-1, lipoprotein-related protein 5, and β-catenin target oncogenes (c-Myc and cyclin E) in H pylori-infected gastric epithelial AGS cells. The Jak1/Stat3 inhibitor AG490 served as the control treatment. The significance of the differences among groups was calculated using the 1-way analysis of variance followed by Newman-Keuls post hoc tests. The results show that lycopene reduced ROS levels and inhibited Jak1/Stat3 activation, alteration of Wnt/β-catenin multiprotein complex molecules, expression of c-Myc and cyclin E, and cell proliferation in H pylori-infected AGS cells. AG490 similarly inhibited H pylori-induced cell proliferation, alteration of Wnt/β-catenin multiprotein complex molecules, and oncogene expression. H pylori increased the levels of Wnt-1 and its receptor lipoprotein-related protein 5; this increase was inhibited by either lycopene or AG490 in AGS cells. In conclusion, lycopene inhibits ROS-mediated activation of Jak1/Stat3 and Wnt/β-catenin signaling and, thus, oncogene expression in relation to hyperproliferation in H pylori-infected gastric epithelial cells. Lycopene might be a potential and promising nutrient for preventing H pylori-associated gastric diseases including gastric cancer.
Collapse
Affiliation(s)
- Bohye Park
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul 03722, Korea.
| | - Joo Weon Lim
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul 03722, Korea.
| | - Hyeyoung Kim
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul 03722, Korea.
| |
Collapse
|