1
|
Yin H, Zheng Y, Chen M, Ding M, Zhang L, Wang R, Wang C, Jia J, Liu X. Pesticide avermectin B1a exerts cytotoxicity by blocking the interaction between mini-chromosome maintenance 6 protein (MCM6) and chromatin licensing and DNA replication factor 1 (CDT1). ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 364:125377. [PMID: 39579921 DOI: 10.1016/j.envpol.2024.125377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 11/06/2024] [Accepted: 11/21/2024] [Indexed: 11/25/2024]
Abstract
Avermectin B1a, a widely used pesticide, has recently raised safety concerns since it possesses potential cytotoxicity toward mammalian cells. Nevertheless, the exact mechanisms that underlie the cytotoxicity induced by avermectin B1a remain elusive. The loading of the mini-chromosome maintenance 6 protein (MCM6) onto chromatin at replication origins by chromatin licensing and DNA replication factor 1 (CDT1) is an essential step for licensing DNA for replication. Here, we first report that avermectin B1a occupies the CDT1-binding domain (CBD) of MCM6 to block the interaction between MCM6 and CDT1 and thus inhibits the licensing for DNA replication. Avermectin B1a inhibits the proliferation with IC50 being 15.1 μM and induces cell cycle arrest at the G0/G1 phase in MEF cells. Moreover, abnormal replication licensing induced by avermectin B1a causes replication stress and DNA double strand breaks, which in turn leads to apoptosis in MEF cells. Further molecular docking uncovers that four residues Glu763, Ile760, Arg771, and Glu774 are vital for the formation of hydrogen bonds in avermectin B1a-CBD interaction. Furthermore, the upregulation of MCM6 or/and CDT1 reverses the avermectin B1a-induced decrease in cell viability and normalizes the cell cycle, indicating that the blockage of MCM6-CDT1 interaction is one of the mechanisms underlying avermectin B1a-induced cytotoxicity. This study not only provides new insights into the mechanism of avermectin B1a-induced cytotoxicity but also offers a useful molecular tool for the investigation of MCM6-CDT1 interaction.
Collapse
Affiliation(s)
- Hao Yin
- Level Three Laboratory of Traditional Chinese Medicine Preparation of National Administration of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China; Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China; Laboratory for Molecular Identification and Biological Evaluation of Chinese Herbal Pieces, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China
| | - Yaoyao Zheng
- Level Three Laboratory of Traditional Chinese Medicine Preparation of National Administration of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China; Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China; Laboratory for Molecular Identification and Biological Evaluation of Chinese Herbal Pieces, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China
| | - Menghan Chen
- Level Three Laboratory of Traditional Chinese Medicine Preparation of National Administration of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China; Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China; Laboratory for Molecular Identification and Biological Evaluation of Chinese Herbal Pieces, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China
| | - Ming Ding
- Level Three Laboratory of Traditional Chinese Medicine Preparation of National Administration of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China; Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China; Laboratory for Molecular Identification and Biological Evaluation of Chinese Herbal Pieces, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China
| | - Lin Zhang
- Level Three Laboratory of Traditional Chinese Medicine Preparation of National Administration of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China
| | - Roujia Wang
- Level Three Laboratory of Traditional Chinese Medicine Preparation of National Administration of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China
| | - Chunyu Wang
- Level Three Laboratory of Traditional Chinese Medicine Preparation of National Administration of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China
| | - Jing Jia
- Laboratory for Molecular Identification and Biological Evaluation of Chinese Herbal Pieces, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China.
| | - Xiaoqian Liu
- Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China.
| |
Collapse
|
2
|
Li Y, Wu Z, Ding T, Zhang W, Guo H, Huang F. Comprehensive bioinformatics analysis and cell line experiments revealed the important role of CDCA3 in sarcoma. Heliyon 2024; 10:e32785. [PMID: 39035484 PMCID: PMC11259814 DOI: 10.1016/j.heliyon.2024.e32785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 07/23/2024] Open
Abstract
Background Sarcoma mainly originate from bone and soft tissue and are highly aggressive malignant tumors. Cell division cycle-related protein 3 (CDCA3) is a protein involved in the regulation of the cell cycle, which is highly expressed in a variety of malignant tumors. However, its role in sarcoma remains unclear. This study aims to investigate the function and potential mechanism of CDCA3 in sarcoma and to elucidate its importance in sarcoma. Methods We first studied the expression and prognosis of CDCA family members in sarcoma by Oncomine and the Gene Expression Profiling Interactive Analysis (GEPIA). The role of CDCA3 protein in sarcoma was further analyzed by the Cancer Genome Atlas Program (TCGA), the Cancer Cell Lineage Encyclopedia (CCLE), and Linke-dOmics. In addition, immunohistochemistry and Western blot were used to verify the expression of CDCA3 protein in clinical samples as well as sarcoma cell lines (U2OS, SAOS2, MG63, and HOS). Subsequently, in vitro experiments (cloning and scratching experiments) were performed using sh-NC as well as sh-CDCA3 group cells to reveal the biological functions of CDCA3. Results We found that the CDCA family (CDCA3, CDCA4, and CDCA8) is highly expressed in sarcoma, and the expression level of CDCA3, CDCA4, and CDCA8 negatively correlates with the prognosis of sarcoma patients. CDCA3 mRNA was highly expressed in pan-cancer by CCLE and TCGA database analysis. KEGG analysis showed that CDCA3 was mainly enriched in the cell cycle signaling pathway (It promoted the transition of the cell cycle from the G0/G1 phase to the S phase). In the level of immune infiltration, CDCA3 was negatively correlated with pDC cells, CD8+T cells, and cytotoxic cells. Finally, patients with high CDCA3 expression in sarcoma were analyzed for resistance to NU7441 and others, while sensitive to Fulvestrant and Dihydrorotenone. Furthermore, we demonstrated high expression of CDCA3 protein in sarcoma tissues and cell lines by immunohistochemistry and Western blot experiments. Cloning, EDU, scratching, and migration experiments showed that the knockdown of CDCA3 inhibited the Proliferation and progression of sarcoma cells. Conclusion These results suggest for the first time that knockdown of CDCA3 may inhibit sarcoma progression. CDCA3 may be an effective target for the treatment of sarcoma.
Collapse
Affiliation(s)
- Yang Li
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Zhiwei Wu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Tao Ding
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Wenbiao Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Hongjuan Guo
- School of Life Sciences, Anhui Medical University, Hefei 230032, China
| | - Fei Huang
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| |
Collapse
|
3
|
Zhu Y, Xu Y, Dai Y, Zhang G, Ji C, Zhang Q, Zhao M. Comparing the enantioselective toxicity on cell cycle and apoptosis of DL-glufosinate and L-glufosinate to SH-SY5Y cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 895:165106. [PMID: 37356769 DOI: 10.1016/j.scitotenv.2023.165106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 06/27/2023]
Abstract
Glufosinate (Glu), a broad-spectrum and highly effective non-selective herbicide, behaves in typical chiral features to target organisms. However, the information on the enantioselective toxicity of DL-Glu and L-Glu against non-target organisms is still limited especially at environmental concentrations. In this study, we investigated the potential mechanism accounting for the enantioselective cytotoxicity of Glu based on cell cycle and apoptosis. Results showed that DL-Glu and L-Glu had no suppression on cell viability at 10-5 M, however, SH-SY5Y cells were significantly arrested at G1/G0 phase after L-Glu exposure compared with DL-Glu. The apoptosis assay exhibited an increase in late apoptosis cells and a decrease in viable cells for DL-Glu and L-Glu treatment. The bioinformatics analysis demonstrated that alterations in transcription translation and signal transduction including "calcium signaling pathway", "Wnt signaling pathway", "FoxO signaling pathway" were the possible pathways responsible for Glu-induced enantioselectivity in cell cycle and apoptosis. Interestingly, the Gene Set Enrichment Analysis (GSEA) also revealed the probable association between DL-Glu exposure and degenerative diseases. These findings serve as a reminder that caution should be exercised not only when using pesticide racemates but also when promoting or applying single- or enriched-isomer pesticides.
Collapse
Affiliation(s)
- Yingying Zhu
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou 310014, PR China; College of Life Science, Taizhou University, Taizhou 318000, PR China
| | - Yongan Xu
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, PR China
| | - Yaoyao Dai
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Guizhen Zhang
- College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua 321004, PR China
| | - Chenyang Ji
- Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Interdisciplinary Research Academy, Zhejiang Shuren University, Hangzhou 310015, PR China.
| | - Quan Zhang
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Meirong Zhao
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou 310014, PR China
| |
Collapse
|
4
|
New screening system using Twist1 promoter activity identifies dihydrorotenone as a potent drug targeting cancer-associated fibroblasts. Sci Rep 2020; 10:7058. [PMID: 32341496 PMCID: PMC7184745 DOI: 10.1038/s41598-020-63996-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 03/24/2020] [Indexed: 12/27/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) are the most abundant stromal cells in tumor microenvironments. These cells strongly support tumor progression and are considered to be potent therapeutic targets. Therefore, drugs targeting CAFs have been developed, but most of them have failed in clinical trials. The discovery of additional drugs to inactivate or eliminate CAFs is thus essential. In this study, we developed a high-throughput screening system to find anti-CAF drugs using reporter cells that express Twist1 promoter-GFP. This screening system uses the activity of the Twist1 promoter as an indicator of CAF activation because Twist1 is known to be a central player in CAF activation. Using this screening system, we found that dihydrorotenone (DHR), an inhibitor of electron transfer chain complex 1 in mitochondria, can effectively deactivate CAFs. DHR-treated CAFs exhibited reduced expression of CAF-enriched markers, decreased capability of collagen gel contraction, and impaired ability to engage in tumor-promoting activities, such as facilitating the proliferation and colonization of cancer cells. Furthermore, conditioned media from DHR-treated CAFs attenuated tumor progression in mice grafted with MNK28 cells. In conclusion, DHR can be considered as a candidate drug targeting CAFs.
Collapse
|
5
|
Zhang L, Zhang J, Zhou H, Dai T, Guo F, Xu S, Chen Y. MicroRNA‐425‐5p promotes breast cancer cell growth by inducing PI3K/AKT signaling. Kaohsiung J Med Sci 2020; 36:250-256. [PMID: 31688991 DOI: 10.1002/kjm2.12148] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 10/13/2019] [Indexed: 12/18/2022] Open
Affiliation(s)
- Li‐Feng Zhang
- Department of General SurgeryThe First Affiliated Hospital of Soochow University Suzhou Jiangsu China
| | - Ji‐Gang Zhang
- Department of Emergency SurgeryThe First Affiliated Hospital of Soochow University Suzhou Jiangsu China
| | - Hao Zhou
- Department of General SurgeryThe First Affiliated Hospital of Soochow University Suzhou Jiangsu China
| | - Tian‐Tian Dai
- Department of General SurgeryThe First Affiliated Hospital of Soochow University Suzhou Jiangsu China
| | - Feng‐Bao Guo
- Department of Emergency SurgeryThe First Affiliated Hospital of Soochow University Suzhou Jiangsu China
| | - Shao‐Yong Xu
- Department of General SurgeryPeople's Hospital of Shiqian County Tongren Guizhou China
| | - Yan Chen
- Department of General SurgeryThe First Affiliated Hospital of Soochow University Suzhou Jiangsu China
| |
Collapse
|
6
|
Xu X, Li S, Cui X, Han K, Wang J, Hou X, Cui L, He S, Xiao J, Yang Y. Inhibition of Ubiquitin Specific Protease 1 Sensitizes Colorectal Cancer Cells to DNA-Damaging Chemotherapeutics. Front Oncol 2019; 9:1406. [PMID: 31921663 PMCID: PMC6930197 DOI: 10.3389/fonc.2019.01406] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 11/27/2019] [Indexed: 12/31/2022] Open
Abstract
Mutations and altered expression of deubiquitinating enzymes (DUBs) have been found associated with many human diseases including cancers. In this study, Ubiquitin specific protease 1 (USP1) expression was found significantly increased in some colorectal cancers (CRC). The elevated USP1 level was associated with short overall survival of patients and with advanced stages of cancers. In cultured CRC cells, knockdown of USP1 induced growth arrest at G2/M of cell cycle and reduced the expression of anti-apoptotic proteins Bcl-2 and Mcl-1. Its knockdown also led to reduction of DNA-repair related substrates FANCD2 and ID1. Further investigations found that small molecular inhibitor of USP1 ML323 sensitized CRC cells to DNA-targeting chemotherapeutics, including doxorubicin, TOPI/II inhibitors, and PARP inhibitor, but not to 5-Fu. These results indicate that USP1 plays a critical in colorectal cancer cell survival and is a promising target for anti-colorectal cancer chemotherapy. Targeting USP1 may represent an effective strategy to regulate the DNA-repairing system.
Collapse
Affiliation(s)
- Xin Xu
- Center for Systems Medicine, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
| | - Shaoyan Li
- Center for Systems Medicine, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China.,School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ximao Cui
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Colorectal Cancer Research Center, Shanghai, China
| | - Kunkun Han
- The Asclepius Technology Company Group and Asclepius Cancer Research Center, Suzhou, China
| | - Jun Wang
- The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaodan Hou
- Center for Systems Medicine, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
| | - Long Cui
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Colorectal Cancer Research Center, Shanghai, China
| | - Songbing He
- The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiecheng Xiao
- The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yili Yang
- Center for Systems Medicine, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China.,State Key Laboratory of Innovative Natural Medicine and TCM Injections, Qingfeng Pharmaceutical, Ganzhou, China
| |
Collapse
|
7
|
Huang Z, Ge H, Yang C, Cai Y, Chen Z, Tian W, Tao J. MicroRNA‐26a‐5p inhibits breast cancer cell growth by suppressing RNF6 expression. Kaohsiung J Med Sci 2019; 35:467-473. [PMID: 31063232 DOI: 10.1002/kjm2.12085] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/22/2019] [Indexed: 02/06/2023] Open
Affiliation(s)
- Zi‐Ming Huang
- Department of Emergency SurgeryThe Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University Huai'an Jiangsu China
| | - Heng‐Fa Ge
- Department of Emergency SurgeryThe Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University Huai'an Jiangsu China
| | - Chen‐Chen Yang
- Department of Emergency SurgeryThe Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University Huai'an Jiangsu China
| | - Yong Cai
- Department of Emergency SurgeryThe Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University Huai'an Jiangsu China
| | - Zhen Chen
- Department of Emergency SurgeryThe Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University Huai'an Jiangsu China
| | - Wen‐Ze Tian
- Department of Cardio‐Thoracic SurgeryThe Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University Huai'an Jiangsu China
| | - Jia‐Li Tao
- Department of EmergencyThe Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University Huai'an Jiangsu China
| |
Collapse
|
8
|
Shen WM, Yin JN, Xu RJ, Xu DF, Zheng SY. Ubiquitin specific peptidase 49 inhibits non-small cell lung cancer cell growth by suppressing PI3K/AKT signaling. Kaohsiung J Med Sci 2019; 35:401-407. [PMID: 31001918 DOI: 10.1002/kjm2.12073] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 04/01/2019] [Indexed: 12/16/2022] Open
Abstract
Ubiquitin specific peptidase 49 (USP49) has been reported as a tumor suppressor in several tumors, but its function and molecular mechanism in non-small cell lung cancer (NSCLC) are still unknown. In this study, USP49 was found downregulated in NSCLC primary tissues and cell lines, and high USP49 predicted a positive index for the overall survival of NSCLC patients. Overexpression of USP49 downregulated the expression levels of Cyclin D1, and upregulated p53 expression. Further flow cytometry analysis showed that overexpressed USP49 induced cell cycle arrest at G0/G1 phase. As a result, overexpression of USP49 significantly inhibited cell growth of NSCLC cells. In mechanism, overexpression of USP49 inhibited PI3K/AKT signaling, but knockdown of USP49 enhanced this signaling. Further studies indicated that USP49 deubiquitinated PTEN and stabilized PTEN protein, which suggested that USP49 inhibited PI3K/AKT signaling by stabilizing PTEN in NSCLC cells. In conclusion, we demonstrated that USP49 was functional in NSCLC cells, and inhibited NSCLC cell growth by suppressing PI3K/AKT signaling, suggesting that USP49 could be as a novel target for NSCLC therapy.
Collapse
Affiliation(s)
- Wen-Ming Shen
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Department of Emergency Surgery, The Affiliated Wujin People's Hospital of Jiangsu University, Changzhou, Jiangsu, China
| | - Jin-Nan Yin
- Department of Emergency Surgery, The Affiliated Wujin People's Hospital of Jiangsu University, Changzhou, Jiangsu, China
| | - Rui-Jun Xu
- Department of Endocrinology, The Affiliated Wujin People's Hospital of Jiangsu University, Changzhou, Jiangsu, China
| | - Da-Fu Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Department of Thoracic Surgery, Huai'an First People's Hospital, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Shi-Ying Zheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
9
|
Huang ZM, Li PL, Yang P, Hou XD, Yang YL, Xu X, Xu F. Overexpression of CMTM7 inhibits cell growth and migration in liver cancer. Kaohsiung J Med Sci 2019; 35:332-340. [PMID: 30903681 DOI: 10.1002/kjm2.12058] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 02/25/2019] [Indexed: 12/27/2022] Open
Abstract
Chemokine-like factor (CKLF)-like, MAL and related proteins for vesicle trafficking and membrane link (MARVEL) transmembrane domain-containing family proteins (CMTMs) have significant roles in the immune system, in male reproduction, as well as in tumorigenesis. Previous studies have shown that CMTM family member 7 (CMTM7) was broadly expressed in various normal tissues, but not in lung, gastric, esophageal, pancreas, and cervix cancers. To explore its relationship with liver cancer, we examined the expression of CMTM7 in liver cancers and its correlation with clinical and pathological conditions. We found that CMTM7 expression was markedly reduced in liver cancer tissues, and negatively correlated with TNM staging and tumor metastasis. In vitro studies showed that enforced expression of CMTM7 inhibited the cell growth and migration of liver cancer cells. Further analysis revealed that CMTM7 suppressed AKT signaling and induced cell cycle arrest at the G0/G1 phase in the liver cancer cells, likely as the consequent of decreased levels of cyclin D1, cyclin-dependent kinase 4 (CDK4), and CDK6, and increased p27 expression. Thus, CMTM7 functions as a tumor suppressor in liver cancer through suppressing cell cycle progression.
Collapse
Affiliation(s)
- Zi-Ming Huang
- Department of Emergency Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Department of Emergency Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Peng-Ling Li
- Department of Respiratory Care, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Peng Yang
- Department of Emergency Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiao-Dan Hou
- Suzhou Institute of Systems Medicine, Center for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, Jiangsu, China
| | - Yi-Li Yang
- Suzhou Institute of Systems Medicine, Center for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, Jiangsu, China
| | - Xin Xu
- Suzhou Institute of Systems Medicine, Center for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, Jiangsu, China
| | - Feng Xu
- Department of Emergency Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
10
|
Xu YY, Song YQ, Huang ZM, Zhang HB, Chen M. MicroRNA-26a inhibits multiple myeloma cell growth by suppressing cyclin-dependent kinase 6 expression. Kaohsiung J Med Sci 2019; 35:277-283. [PMID: 30897301 DOI: 10.1002/kjm2.12057] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 02/25/2019] [Indexed: 12/28/2022] Open
Abstract
MicroRNA-26a (miR-26a) has been reported to be involved in the tumorigenesis of several tumors, but its biological function and molecular mechanism in multiple myeloma (MM) are still unknown. In this study, we found that overexpression of miR-26a obviously inhibited MM cell growth, and delayed tumor growth in xenografts. Further studies showed that overexpression of miR-26a induced cell cycle arrest at G0/G1 phase in MM cells. MiR-26a mimic down-regulated the expression levels of CDK6 and E2F1, but up-regulated p53 and p21 expression. In contrast, overexpression of CDK6 decreased the effect of miR-26a mimic on MM cell survival. Moreover, miR-26a targeted CDK6 mRNA and thus suppressed CDK6 protein expression. Overexpression of miR-26a also enhanced the cytotoxic action of doxorubicin against MM. These results demonstrated that miR-26a was involved in the development of MM through regulating CDK6 signaling pathway, and indicated that miR-26a could be as a novel target for anti-tumor therapy in clinic as a single strategy or in combination with other anti-tumor drugs in MM.
Collapse
Affiliation(s)
- Ying-Ying Xu
- Department of Radiation Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China.,Department of Radiotherapy, The Affiliated Huai'an NO. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Ya-Qi Song
- Department of Radiotherapy, The Affiliated Huai'an NO. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Zi-Ming Huang
- Department of Emergency Medicine, The Affiliated Huai'an NO.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Hai-Bing Zhang
- Department of Radiotherapy, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Ming Chen
- Department of Radiation Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
11
|
Xu D, Tian W, Jiang C, Huang Z, Zheng S. The anthelmintic agent oxfendazole inhibits cell growth in non‑small cell lung cancer by suppressing c‑Src activation. Mol Med Rep 2019; 19:2921-2926. [PMID: 30720086 DOI: 10.3892/mmr.2019.9897] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 12/04/2018] [Indexed: 11/05/2022] Open
Abstract
The c‑Src protein family of tyrosine kinases are important in the tumorigenesis of many types of tumors, and may be a potential target for antitumor drug discovery. In the present study, immunoblotting was performed to analyze protein expression, CCK‑8 assay was carried out to assess cell viability and cell cycle was analyzed using a flow cytometer. The anthelmintic agent oxfendazole was observed to be a novel c‑Src inhibitor that blocked the activation of c‑Src. Oxfendazole also suppressed the cell growth of non‑small cell lung cancer (NSCLC) cells, and overexpression of c‑Src decreased the cytotoxicity of oxfendazole against NSCLC cells. In addition, oxfendazole induced cell cycle arrest at the G0/G1 phase, and downregulated the protein levels of Cyclin‑dependent kinase (CDK)‑4, CDK6, retinoblastoma protein and E2 transcription factor 1, and upregulated the expression levels of p53 and p21 in NSCLC cells. Furthermore, oxfendazole enhanced the cytotoxicity of cisplatin against NSCLC cells. These results demonstrated that oxfendazole exerted its antitumor activity by suppressing c‑Src signaling, and it was also indicated that the anthelmintic agent oxfendazole may be effective for anti‑NSCLC therapy in the clinic as a single agent or in combination with other antitumor drugs.
Collapse
Affiliation(s)
- Dafu Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Wenze Tian
- Department of Thoracic Surgery, Huai'an First People's Hospital, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Chao Jiang
- Department of Oncology, Huai'an First People's Hospital, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Ziming Huang
- Department of Emergency Surgery, Huai'an First People's Hospital, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Shiying Zheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
12
|
Liu Y, Wang X, Zeng S, Zhang X, Zhao J, Zhang X, Chen X, Yang W, Yang Y, Dong Z, Zhu J, Xu X, Tian F. The natural polyphenol curcumin induces apoptosis by suppressing STAT3 signaling in esophageal squamous cell carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:303. [PMID: 30518397 PMCID: PMC6280482 DOI: 10.1186/s13046-018-0959-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 11/13/2018] [Indexed: 12/12/2022]
Abstract
Background We and others have previously shown that the STAT3 signaling pathway is activated in some esophageal squamous cell carcinoma (ESCC) cells and is required for the survival and growth of these primary ESCC-derived xenografts. It has also been shown that the natural polyphenol curcumin is an effective anti-tumor agent. Methods Luciferase assay and immunoblotting were performed to examine whether curcumin suppressed STAT3 signaling. CCK-8 assay and xenografts were utilized for analyzing ESCC cell growth in culture and mice. Soft agar assay was carried out to determine the colony formation ability of ESCC cells in the presence or absence of curcumin. Cell death and cell cycle were assessed by In CELL Analyzer 2000. Immunohistochemistry and TUNEL assay were used for detecting apoptosis in ESCC tisuses. Molecular docking was performed to evaluate the interaction of curcumin with JAK2. JAK2 activity was assessed using an in vitro cell-free system. HE staining was used to evaluate the ESCC tissues. Results The natural polyphenol curcumin inhibited STAT3 phosphorylation rapidly and blocked STAT3-mediated signaling in ESCC cells. It also induced growth arrest and apoptosis in cultured ESCC cells, which were attenuated by enforced expression of STAT3. Furthermore, curcumin preferentially blocked the growth of primary ESCC-derived xenografts that harbored activated STAT3. Conclusions Curcumin is able to exert anti-tumor action through inhibiting the STAT3 signaling pathway. Giving its wide use in traditional medicines with low toxicity and few adverse reactions, it is conceivable that curcumin might be further explored as a unique STAT3 inhibitor for anti-cancer therapies.
Collapse
Affiliation(s)
- Ying Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China.,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, 450001, People's Republic of China.,Clinical Research Center, People's Hospital of Zhengzhou, Zhengzhou, Henan, 450001, People's Republic of China
| | - Xinhua Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China
| | - Shuang Zeng
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China.,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, 450001, People's Republic of China
| | - Xiane Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China.,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, 450001, People's Republic of China
| | - Jimin Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China.,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, 450001, People's Republic of China
| | - Xiaoyan Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China.,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, 450001, People's Republic of China
| | - Xinhuan Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China.,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, 450001, People's Republic of China
| | - Wanjing Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China.,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, 450001, People's Republic of China
| | - Yili Yang
- Suzhou Institute of Systems Medicine, Center for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, Jiangsu, 215123, People's Republic of China
| | - Ziming Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China.,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, 450001, People's Republic of China
| | - Jingyu Zhu
- School of Medicine and Pharmaceutics, Jiangnan University, Wuxi, Jiangsu, 214000, People's Republic of China
| | - Xin Xu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China. .,Suzhou Institute of Systems Medicine, Center for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, Jiangsu, 215123, People's Republic of China.
| | - Fang Tian
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China. .,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, 450001, People's Republic of China.
| |
Collapse
|
13
|
Huang Z, Cai Y, Yang C, Chen Z, Sun H, Xu Y, Chen W, Xu D, Tian W, Wang H. Knockdown of RNF6 inhibits gastric cancer cell growth by suppressing STAT3 signaling. Onco Targets Ther 2018; 11:6579-6587. [PMID: 30323630 PMCID: PMC6178940 DOI: 10.2147/ott.s174846] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background and objective RNF6, an E3 ligase, has been reported to play an important role in the tumorigenesis in several tissues, but its role in gastric cancer is still unknown. In this study, we aimed to investigate the biological function and molecular mechanisms of RNF6 in gastric cancer. Materials and methods The expression levels of RNF6 were detected by quantitative real-time PCR (qRT-PCR) and immunoblotting in gastric cancer tissues and cell lines. Cell Counting Kit-8 assay was performed to evaluate cell proliferation. Cell apoptosis was analyzed by flow cytometer and immunoblotting. Luciferase assay, immunoblotting and qRT-PCR were performed to explore the activation of STAT3. Immunoprecipitation was performed to evaluate the ubiquitination of SHP-1. Results In this study, RNF6 was found to be upregulated in both primary tissues and cell lines of gastric cancer. Knockdown or overexpression of RNF6 inhibited or promoted cell growth of gastric cancer cells. Knockdown of RNF6 also induced the cleavage of PARP and promoted cell apoptosis in gastric cancer cells. In addition, knockdown of RNF6 also increased the cytotoxicity of doxorubicin against gastric cancer. Moreover, knockdown of RNF6 inhibited STAT3-derived luciferase activity and downregulated the phosphorylation of STAT3, but upregulated the protein level of SHP-1. Knockdown of RNF6 downregulated the expression of MCL1 and XIAP, which are target genes of STAT3. Further studies showed that RNF6 regulated the stability of SHP-1 by inducing its polyubiquitination. Conclusion These results demonstrated that RNF6 was highly expressed in gastric cancer and regulated the growth of gastric cancer cells by affecting SHP-1/STAT3 signaling, which suggested that RNF6 could be a novel target for gastric cancer therapy.
Collapse
Affiliation(s)
- Ziming Huang
- Department of Emergency Surgery, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China,
| | - Yong Cai
- Department of Emergency Surgery, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China,
| | - Chenchen Yang
- Department of Emergency Surgery, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China,
| | - Zhen Chen
- Department of Emergency Surgery, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China,
| | - Hong Sun
- Department of Emergency Surgery, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China,
| | - Yingying Xu
- Department of Radiotherapy, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China
| | - Wei Chen
- Department of Respiratory Care, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China
| | - Dafu Xu
- Department of Cardiothoracic Surgery, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China
| | - Wenze Tian
- Department of Cardiothoracic Surgery, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China
| | - Haixiao Wang
- Department of General Surgery, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China,
| |
Collapse
|
14
|
Xu X, Han K, Zhu J, Mao H, Lin X, Zhang Z, Cao B, Zeng Y, Mao X. An inhibitor of cholesterol absorption displays anti-myeloma activity by targeting the JAK2-STAT3 signaling pathway. Oncotarget 2018; 7:75539-75550. [PMID: 27705908 PMCID: PMC5342759 DOI: 10.18632/oncotarget.12265] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 09/16/2016] [Indexed: 01/08/2023] Open
Abstract
The activated JAK2-STAT3 signaling pathway is a high risk factor for multiple myeloma (MM), a fatal malignancy of plasma cells. In the present study, SC09, a potential inhibitor of cholesterol absorption, was identified in a STAT3-targeted drug screen. SC09 suppressed the activation of STAT3 in a time-course and concentration-dependent manner but did not affect its family members STAT1 and STAT5. SC09 inhibited STAT3 transcriptional activity and downregulated the expression of STAT3-regulated genes. Further studies showed that SC09 selectively inhibited JAK2 activation but not other kinases including c-Src, ERK, p38 and mTOR that are all associated with STAT3 activation. Moreover, SC09 obviously induced MM cell death in vitro and delayed MM tumor growth in vivo. SC09-induced MM cell death was dependent on the endogenous STAT3 status, and this effect could be attenuated by enforced expression of STAT3. All the results collectively indicated that SC09 blocks the JAK2-STAT3 signaling thus displaying anti-MM activity. Given its well tolerance and anti-MM potency, SC09 is credited for further investigation as a promising drug for MM treatment.
Collapse
Affiliation(s)
- Xin Xu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Kunkun Han
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jingyu Zhu
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, China
| | - Hongwu Mao
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Xu Lin
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Zubin Zhang
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Biyin Cao
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yuanying Zeng
- Department of Oncology, Suzhou Municipal Hospital East Campus, Suzhou, China
| | - Xinliang Mao
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| |
Collapse
|
15
|
Wang H, Guo Q, Yang P, Long G. Restoration of microRNA-212 causes a G0/G1 cell cycle arrest and apoptosis in adult T-cell leukemia/lymphoma cells by repressing CCND3 expression. J Investig Med 2016; 65:82-87. [DOI: 10.1136/jim-2016-000233] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2016] [Indexed: 12/22/2022]
Abstract
Adult T-cell leukemia/lymphoma (ATL) is a highly aggressive T-cell malignancy. This study was designed to explore the expression and functional significance of microRNA (miR)-212 in ATL. The expression of miR-212 in human ATL tissues and cell lines were investigated. Gain-of-function experiments were carried out to determine the roles of miR-212 in cell proliferation, tumorigenesis, cell cycle progression, and apoptosis. We also identified and functionally characterized the target genes of miR-212 in ATL cells. Compared with normal lymph node biopsies, lymphoma samples from ATL patients displayed underexpression of miR-212 (p=0.0032). Consistently, miR-212 was downregulated in human ATL cell lines, compared with normal T lymphocytes. Restoration of miR-212 significantly (p<0.05) inhibited ATL cell proliferation and tumorigenesis in mice. Overexpression of miR-212 led to an accumulation of G0/G1-phase cells and a concomitant reduction of S-phase cells. Moreover, enforced expression of miR-212-induced significant apoptosis in ATL cells. CCND3, which encodes a cell cycle regulator cyclin D3, was identified as a direct target of miR-212 in ATL cells. Rescue experiments with a miR-212-resistant variant of CCND3 demonstrated that overexpression of CCND3 restored cell-cycle progression and attenuated apoptotic response in miR-212-overexpressing ATL cells. Taken together, miR-212 exerts growth-suppressive effects in ATL cells largely by targeting CCND3 and may have therapeutic potential in ATL.
Collapse
|
16
|
Luo X, Peng JM, Su LDI, Wang DY, Yu YJ. Fangchinoline inhibits the proliferation of SPC-A-1 lung cancer cells by blocking cell cycle progression. Exp Ther Med 2015; 11:613-618. [PMID: 26893655 DOI: 10.3892/etm.2015.2915] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 09/24/2015] [Indexed: 01/19/2023] Open
Abstract
Fangchinoline (Fan) is a bioactive compound isolated from the Chinese herb Stephania tetrandra S. Moore (Fen Fang Ji). The aim of the present study was to investigate the effect of Fan on the proliferation of SPC-A-1 lung cancer cells, and to define the associated molecular mechanisms. Following treatment with Fan, Cell Counting Kit-8, phase contrast imaging and Giemsa staining assays were used to detect cell viability; flow cytometry was performed to analyze the cell cycle distribution; and reverse transcription-quantitative polymerase chain reaction and western blot assays were used to investigate changes in the expression levels of cell cycle-associated genes and proteins. In the present study, treatment with Fan markedly inhibited the proliferation of SPC-A-1 lung cancer cells and significantly increased the percentage of cells in the G0/G1 phase of the cell cycle in a dose-dependent manner (P<0.05 for 2.5-5 µm; P<0.01 for 10 µm), whereas the percentage of cells in the S and G2/M phases were significantly reduced following treatment (P<0.05 for 5 µm; P<0.01 for 10 µm). Mechanistically, Fan significantly reduced the mRNA expression levels of cyclin D1, cyclin-dependent kinase 4 (CDK4) and CDK6 (P<0.05 for 2.5-5 µm; P<0.01 for 10 µm), which are key genes in the regulation of the G0/G1 phase of the cell cycle. Furthermore, treatment with Fan also decreased the expression of phosphorylated retinoblastoma (Rb) and E2F transcription factor-1 (E2F-1) proteins (P<0.05 for 5 µm; P<0.01 for 10 µm). In summary, the present study demonstrated that Fan inhibited the proliferation of SPC-A-1 lung cancer cells and induced cell cycle arrest at the G0/G1 phase. These effects may be mediated by the downregulation of cellular CDK4, CDK6 and cyclin D1 levels, thus leading to hypophosphorylation of Rb and subsequent suppression of E2F-1 activity. Therefore, the present results suggest that Fan may be a potential drug candidate for the prevention of lung cancer.
Collapse
Affiliation(s)
- Xue Luo
- Medical College of Yangzhou Polytechnic College, Yangzhou, Jiangsu 225009, P.R. China
| | - Jian-Ming Peng
- Medical College of Yangzhou Polytechnic College, Yangzhou, Jiangsu 225009, P.R. China; Key Laboratory of Pain, Basic Research and Clinical Therapy, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Lan-DI Su
- Medical College of Yangzhou Polytechnic College, Yangzhou, Jiangsu 225009, P.R. China
| | - Dong-Yan Wang
- Medical College of Yangzhou Polytechnic College, Yangzhou, Jiangsu 225009, P.R. China
| | - You-Jiang Yu
- Medical College of Yangzhou Polytechnic College, Yangzhou, Jiangsu 225009, P.R. China
| |
Collapse
|
17
|
Pereira DM, Valentão P, Correia-da-Silva G, Teixeira N, Andrade PB. Translating endoplasmic reticulum biology into the clinic: a role for ER-targeted natural products? Nat Prod Rep 2015; 32:705-22. [PMID: 25703279 DOI: 10.1039/c4np00102h] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
ER stress has been identified as a hallmark, and sometimes trigger, of several pathologies, notably cancer, inflammation and neurodegenerative diseases like Alzheimer's and Parkinson's. Among the molecules described in literature known to affect ER function, the majority are natural products, suggesting that natural molecules may constitute a significant arsenal of chemical entities for modulating this cellular target. In this review, we will start by presenting the current knowledge of ER biology and the hallmarks of ER stress, thus paving the way for presenting the natural products that have been described as being ER modulators, either stress inducers or ER protectors. The chemistry, distribution and mechanism of action of these compounds will be presented and discussed.
Collapse
Affiliation(s)
- David M Pereira
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, no. 228, 4050-313 Porto, Portugal.
| | | | | | | | | |
Collapse
|