51
|
Luo H, Vong CT, Chen H, Gao Y, Lyu P, Qiu L, Zhao M, Liu Q, Cheng Z, Zou J, Yao P, Gao C, Wei J, Ung COL, Wang S, Zhong Z, Wang Y. Naturally occurring anti-cancer compounds: shining from Chinese herbal medicine. Chin Med 2019; 14:48. [PMID: 31719837 PMCID: PMC6836491 DOI: 10.1186/s13020-019-0270-9] [Citation(s) in RCA: 331] [Impact Index Per Article: 55.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/23/2019] [Indexed: 12/24/2022] Open
Abstract
Numerous natural products originated from Chinese herbal medicine exhibit anti-cancer activities, including anti-proliferative, pro-apoptotic, anti-metastatic, anti-angiogenic effects, as well as regulate autophagy, reverse multidrug resistance, balance immunity, and enhance chemotherapy in vitro and in vivo. To provide new insights into the critical path ahead, we systemically reviewed the most recent advances (reported since 2011) on the key compounds with anti-cancer effects derived from Chinese herbal medicine (curcumin, epigallocatechin gallate, berberine, artemisinin, ginsenoside Rg3, ursolic acid, silibinin, emodin, triptolide, cucurbitacin B, tanshinone I, oridonin, shikonin, gambogic acid, artesunate, wogonin, β-elemene, and cepharanthine) in scientific databases (PubMed, Web of Science, Medline, Scopus, and Clinical Trials). With a broader perspective, we focused on their recently discovered and/or investigated pharmacological effects, novel mechanism of action, relevant clinical studies, and their innovative applications in combined therapy and immunomodulation. In addition, the present review has extended to describe other promising compounds including dihydroartemisinin, ginsenoside Rh2, compound K, cucurbitacins D, E, I, tanshinone IIA and cryptotanshinone in view of their potentials in cancer therapy. Up to now, the evidence about the immunomodulatory effects and clinical trials of natural anti-cancer compounds from Chinese herbal medicine is very limited, and further research is needed to monitor their immunoregulatory effects and explore their mechanisms of action as modulators of immune checkpoints.
Collapse
Affiliation(s)
- Hua Luo
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Chi Teng Vong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Hanbin Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yan Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peng Lyu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Ling Qiu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Mingming Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Qiao Liu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zehua Cheng
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jian Zou
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peifen Yao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Caifang Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jinchao Wei
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Carolina Oi Lam Ung
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Shengpeng Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zhangfeng Zhong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yitao Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| |
Collapse
|
52
|
Novel role of sex-determining region Y-box 7 (SOX7) in tumor biology and cardiovascular developmental biology. Semin Cancer Biol 2019; 67:49-56. [PMID: 31473269 DOI: 10.1016/j.semcancer.2019.08.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 08/19/2019] [Accepted: 08/28/2019] [Indexed: 02/06/2023]
Abstract
The sex-determining region Y-box 7 (Sox7) is an important member of the SOX F family, which is characterized by a high-mobility-group DNA-binding domain. Previous studies have demonstrated the role of SOX7 in cardiovascular development. SOX7 expression could be detected in normal adult tissues. Furthermore, the expression levels of SOX7 were different in different tumors. Most studies showed the downregulation of SOX7 in tumors, while some studies reported its upregulation in tumors. In this review, we first summarized the upstream regulators (including transcription factors, microRNAs (miRNAs), long noncoding RNAs (lncRNAs) and some exogenous regulators) and downstream molecules (including factors in the Wnt/β-catenin signaling pathway and some other signaling pathways) of SOX7. Then, the roles of SOX7 in multiple tumors were presented. Finally, the significance of divergent SOX7 expression during cardiovascular development was briefly discussed. The information compiled in this study characterized SOX7 during tumorigenesis and cardiovascular development, which should facilitate the design of future research and promote SOX7 as a therapeutic target.
Collapse
|
53
|
Cao M, Fang Y, Jia W, Wang Y, Sun J, Tao D. Emodin relieves hypoxia-triggered injury via elevation of microRNA-25 in PC-12 cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2678-2687. [PMID: 31257935 DOI: 10.1080/21691401.2019.1633339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Emodin (EMO) possesses extensive pharmacological activities, which has been proven to exert the protective impact in diverse nervous system diseases. Nonetheless, whether EMO emerges a neuro-protective activity in hypoxic-evoked ischemic brain injury is still further probed. The intention of the research is to disclose whether EMO emerges neuro-protective activity in hypoxic-evoked ischemic brain injury. PC-12 received hypoxia administration, and then cell viability, apoptosis and autophagy were estimated. After EMO disposition, the above-involved cellular processes were evaluated again. MiR-25 functions in EMO-affected cells were also estimated. The interrelation between miR-25 and neurofilament light-chain polypeptide gene (NEFL) and the conceivable roles of NEFL in hypoxia-disposed cells were investigated. The latent mechanism was uncovered by mTOR and Notch pathways determination. Hypoxia triumphantly triggered apoptosis and autophagy, but EMO repressed these functions in PC-12 cells. Increased miR-25 was induced by EMO, and inhibited miR-25 abated the impacts of EMO on hypoxia-disposed PC-12 cells. NEFL as a neoteric target gene of miR-25 was predicated, and overexpressed NEFL annulled the functions of EMO in hypoxia-injured cells. EMO activated mTOR and Notch pathways through repressing NEFL. The investigations corroborated that EMO weakened hypoxia-triggered injury via elevating miR-25 by targeting NEFL in PC-12 cells.
Collapse
Affiliation(s)
- Mingjuan Cao
- a Department of Rehabilitation, Zibo Integrated Traditional Chinese and Western Medicine Hospital , Zibo , PR China
| | - Yuqing Fang
- b Department of Neurology, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University , Jinan , PR China
| | - Wei Jia
- c Department of Gastroenterology, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University , Jinan , PR China
| | - Yao Wang
- d Department of Neurology, The First Affiliated Hospital of Dalian Medical University , Dalian , PR China
| | - Jingyi Sun
- d Department of Neurology, The First Affiliated Hospital of Dalian Medical University , Dalian , PR China
| | - Dingbo Tao
- d Department of Neurology, The First Affiliated Hospital of Dalian Medical University , Dalian , PR China
| |
Collapse
|
54
|
Tan B, Huang Y, Lan L, Zhang B, Ye L, Yan W, Wang F, Lin N. Bruceine D induces apoptosis in human non-small cell lung cancer cells through regulating JNK pathway. Biomed Pharmacother 2019; 117:109089. [PMID: 31226632 DOI: 10.1016/j.biopha.2019.109089] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 05/23/2019] [Accepted: 06/04/2019] [Indexed: 12/24/2022] Open
Abstract
Bruceine D (BD) is the quassinoids isolated from the traditional Chinese herbal medicine Brucea javanica's fruit, which exhibits anti-cancer activity. Here, we demonstrated that BD inhibited human non-small cell lung cancer (NSCLC) cell lines in vitro that were attributed to the induction of cell apoptosis. Human NSCLC H460 and A549 cell lines were treated with BD, and cell viability was conducted with CCK-8 assay. Cell clone formation was observed by clone formation assay. Cell apoptosis was measured using DAPI staining and flow cytometry. Protein levels was analyzed by western blot. The results showed BD inhibited the cell viability of H460 and A549 cells in a dose-dependent manner with IC50 values of 0.5 and 0.6 μmol/L, respectively, at 48 h of treatment. Treatment with BD (0.125-1.0 μmol/L) dose-dependently promoted chromatin condensation, Annexin V-positive cell population and caspase-dependent apoptosis in H460 and A549 cells. Mechanistically, BD stimulated the phosphorylation of JNK. Furthermore, the anti-cancer effects of BD were alleviated effectively by a specific JNK inhibitor SP600125 in NSCLC cells. In conclusion, the results demonstrated that BD exerted anti-cancer activity against NSCLC cells through JNK activation, which suggests its potent usefulness for prevention and treatment of NSCLC.
Collapse
Affiliation(s)
- Biqin Tan
- Department of Clinical Pharmacology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Yuyu Huang
- Department of Clinical Pharmacology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Lihua Lan
- Institute of Agricultural Bio-Environmental Engineering, College of Biosystem Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Bo Zhang
- Department of Clinical Pharmacology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Lijun Ye
- Department of Clinical Pharmacology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Wei Yan
- Department of Clinical Pharmacology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Fei Wang
- Department of Clinical Pharmacology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China.
| | - Nengming Lin
- Department of Clinical Pharmacology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China.
| |
Collapse
|
55
|
Qu J, Liu B, Li B, Du G, Li Y, Wang J, He L, Wan X. TRIB3 suppresses proliferation and invasion and promotes apoptosis of endometrial cancer cells by regulating the AKT signaling pathway. Onco Targets Ther 2019; 12:2235-2245. [PMID: 30988628 PMCID: PMC6441550 DOI: 10.2147/ott.s189001] [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] [Indexed: 12/11/2022] Open
Abstract
Objective The aim of this study was to examine the effect of TRIB3 on proliferation, apoptosis, and migration of endometrial cancer (EC) cells and explore the relationship between TRIB3 and AKT signaling pathway in EC progression. Methods Immunohistochemical analysis was performed to measure the expression level of TRIB3 in normal endometrium tissues and EC tissues. Overexpression and shRNA knockdown techniques were applied by transfecting EC cells (ISK and AN3CA), and the effect of TRIB3 on EC cell biological behaviors was evaluated. Cell Counting Kit-8 and colony formation assays were utilized to investigate EC cell proliferation ability, and flow cytometry was performed to assess the apoptosis of EC cells. Moreover, the migration and invasion of EC cells were detected by transwell assay, and the levels of MMP-2 and MMP-9 were measured by ELISA. Additionally, Western blot analysis was carried out to determine the levels of AKT and p-AKT. Results The expression level of TRIB3 was higher in EC than normal endometrium tissues, and its overexpression promoted apoptosis and suppressed proliferation of EC cells. Furthermore, TRIB3 retarded the migration and invasion of EC cells and decreased the levels of MMP-2 and MMP-9. Conversely, TRIB3 inhibition enhanced the expression levels of MMP-2 and MMP-9, and proliferation and migration of EC cells but suppressed their apoptosis. Similarly, TRIB3 overexpression reduced while its knockdown increased the level of p-AKT. Conclusion TRIB3 inhibited proliferation and migration and promoted apoptosis of EC cells probably through regulating AKT signaling pathway.
Collapse
Affiliation(s)
- Junjie Qu
- Department of Gynaecology, Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai 201204, China,
| | - Binya Liu
- Department of Gynaecology, Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai 201204, China,
| | - Bilan Li
- Department of Gynaecology, Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai 201204, China,
| | - Guiqiang Du
- Department of Gynaecology, Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai 201204, China,
| | - Yiran Li
- Department of Gynaecology, Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai 201204, China,
| | - Jingyun Wang
- Department of Gynaecology, Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai 201204, China,
| | - Laman He
- Department of Gynaecology, Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai 201204, China,
| | - Xiaoping Wan
- Department of Gynaecology, Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai 201204, China,
| |
Collapse
|
56
|
Dong X, Ni B, Fu J, Yin X, You L, Leng X, Liang X, Ni J. Emodin induces apoptosis in human hepatocellular carcinoma HepaRG cells via the mitochondrial caspase‑dependent pathway. Oncol Rep 2018; 40:1985-1993. [PMID: 30106438 PMCID: PMC6111625 DOI: 10.3892/or.2018.6620] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 02/12/2018] [Indexed: 12/17/2022] Open
Abstract
Emodin-induced hepatotoxicity in vivo and in vitro has been gaining increasing attention. However, the exact molecular pathways underlying these effects remain poorly clarified. The aim of the present study was to evaluate the cytotoxic effect of emodin on HepaRG cells and to define the underlying mechanism. The results demonstrated that emodin evidently inhibited HepaRG cell growth in a dose- and time-dependent manner by blocking cell cycle progression in the S and G2/M phase and by inducing apoptosis. Emodin treatment also resulted in generation of reactive oxygen species (ROS), which abrogated mitochondrial membrane potential (MMP). The above effects were all suppressed by antioxidants, such as N-acetylcysteine (NAC). Further studies by western blot analysis howed that emodin upregulated p53, p21, Bax, cyclin E, cleaved caspase-3, 8 and 9, and cleaved poly(ADP-ribose)polymerase (PARP). However, the protein expression of Bcl-2, cyclin A and CDK2 was downregulated. Taken together, our results suggest that emodin induces apoptosis via the mitochondrial apoptosis pathway through cell cycle arrest and ROS generation in HepaRG cells.
Collapse
Affiliation(s)
- Xiaoxv Dong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, P.R. China
| | - Boran Ni
- School of Basic Medical Science, Beijing University of Chinese Medicine, Beijing 100102
| | - Jing Fu
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010
| | - Xingbin Yin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, P.R. China
| | - Longtai You
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, P.R. China
| | - Xin Leng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, P.R. China
| | - Xiao Liang
- Shanghai Binuo Medical Instrument Co., Ltd., Shanghai 200000, P.R. China
| | - Jian Ni
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, P.R. China
| |
Collapse
|
57
|
Ding CZ, Guo XF, Wang GL, Wang HT, Xu GH, Liu YY, Wu ZJ, Chen YH, Wang J, Wang WG. High glucose contributes to the proliferation and migration of non-small cell lung cancer cells via GAS5-TRIB3 axis. Biosci Rep 2018; 38:BSR20171014. [PMID: 29367413 PMCID: PMC5857909 DOI: 10.1042/bsr20171014] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 11/29/2017] [Accepted: 01/23/2018] [Indexed: 01/01/2023] Open
Abstract
Despite the growing number of studies exhibited an association of diabetes mellitus (DM) and lung cancer progression, the concrete mechanism of DM aggravating lung cancer has not been elucidated. This study was to investigate whether and how high glucose (HG) contribute to the proliferation and migration of non-small cell lung cancer (NSCLC) cells in vitro. In the present study, we confirmed that HG promoted the proliferation and migration of NSCLC cells, and also induced an anti-apoptosis effect on NSCLC cells. Moreover, HG inhibited the expression of GAS5 in NSCLC cells but elevated the protein level of TRIB3. GAS5 overexpression promoted the degradation of TRIB3 protein by ubiquitination and inhibited the HG induced-proliferation, anti-apoptosis and migration of NSCLC cells. Importantly, TRIB3 overexpression reversed the effects of GAS5 on the HG-treated NSCLC cells. Taken together, down-regulated GAS5 by HG significantly enhanced the proliferation, anti-apoptosis and migration in NSCLC cells through TRIB3, thus promoting the carcinogenesis of NSCLC.
Collapse
Affiliation(s)
- Cheng-Zhi Ding
- Department of Thoracic Oncology, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Xu-Feng Guo
- Department of Thoracic Surgery, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Guo-Lei Wang
- Department of Thoracic Oncology, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Hong-Tao Wang
- Department of Thoracic Oncology, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Guang-Hui Xu
- Department of Thoracic Oncology, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Yuan-Yuan Liu
- Department of Thoracic Surgery, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Zhen-Jiang Wu
- Department of Thoracic Oncology, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Yu-Hang Chen
- Department of Thoracic Oncology, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Jiao Wang
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wen-Guang Wang
- Department of Thoracic Oncology, Henan Provincial Chest Hospital, Zhengzhou, China
| |
Collapse
|
58
|
Qu W, Wang Y, Wu Q, Hao D, Li D. Emodin Impairs Radioresistance of Human Osteosarcoma Cells by Suppressing Sonic Hedgehog Signaling. Med Sci Monit 2017; 23:5767-5773. [PMID: 29203762 PMCID: PMC5719725 DOI: 10.12659/msm.907453] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Material/Methods Results Conclusions
Collapse
Affiliation(s)
- Wei Qu
- Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland).,Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Yufei Wang
- Department of Bone Microsurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Qining Wu
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Dingjun Hao
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Dichen Li
- Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland).,State Key Laboratory for Manufacturing Systems Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| |
Collapse
|
59
|
Wang Y, Luo Q, He X, Wei H, Wang T, Shao J, Jiang X. Emodin Induces Apoptosis of Colon Cancer Cells via Induction of Autophagy in a ROS-Dependent Manner. Oncol Res 2017; 26:889-899. [PMID: 28762328 PMCID: PMC7844792 DOI: 10.3727/096504017x15009419625178] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Recent studies revealed that emodin extracted from Chinese herbs exhibits an anticancer effect on different cancer types, including colon cancer. However, the mechanism is not well understood. In our study, we confirmed that emodin treatment inhibited cell viability and induced apoptosis in colon cancer cells. Further experiments found that emodin was also able to induce autophagy, which is indispensible for apoptosis induced by emodin. More interestingly, emodin treatment also results in mitochondrial dysfunction and ROS accumulation in colon cancer cells. Finally, we stressed that ROS accumulation is essential for autophagy and apoptosis induced by emodin. In conclusion, emodin induces apoptosis in colon cancer cells through induction of autophagy, during which ROS generation is of the essence. Our findings improve understanding of emodin's effect on colon cancer suppression and provide a new theoretical basis for colon cancer therapy.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Department of Biomedical Sciences, Chengdu Medical College, Chengdu, Sichuan, P.R. China
| | - Qin Luo
- Department of Biomedical Sciences, Chengdu Medical College, Chengdu, Sichuan, P.R. China
| | - Xianlu He
- Department of General Surgery, Second Affiliated Hospital of Chengdu Medical College (China National Nuclear Corporation 416 Hospital), Chengdu, Sichuan, P.R. China
| | - He Wei
- Department of Biomedical Sciences, Chengdu Medical College, Chengdu, Sichuan, P.R. China
| | - Ting Wang
- Department of Biomedical Sciences, Chengdu Medical College, Chengdu, Sichuan, P.R. China
| | - Jichun Shao
- Department of Urology, Second Affiliated Hospital of Chengdu Medical College (China National Nuclear Corporation 416 Hospital), Chengdu, Sichuan, P.R. China
| | - Xinni Jiang
- Department of Biomedical Sciences, Chengdu Medical College, Chengdu, Sichuan, P.R. China
| |
Collapse
|