1
|
Raza W, Meena A, Luqman S. THF induces apoptosis by downregulating initiation, promotion, and progression phase biomarkers in skin and lung carcinoma. J Biochem Mol Toxicol 2024; 38:e23838. [PMID: 39243196 DOI: 10.1002/jbt.23838] [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: 05/06/2024] [Revised: 07/18/2024] [Accepted: 08/23/2024] [Indexed: 09/09/2024]
Abstract
3,5,7-Trihydroxy-2-phenylchromen-4-one (THF) possesses a diverse range of pharmacological activities. Evidence suggests that THF exerts anticancer activity by distinct mechanisms of action. This study explores the anticancer potential of THF in human lung (A549) and skin (A431) cancer cells by employing different antiproliferative assays. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, neutral red uptake, sulphorhodamine B, and cell motility assays were used to confirm the anticancer potential of THF. Cell target-based and quantitative reverse transcription polymerase chain reaction (qRT-PCR) assays were used to explore the effect of THF on the initiation, promotion and progression phase biomarkers of carcinogenesis. THF suppresses the activity of lipoxygenase-5 up to ~40% in both A549 and A431 cells and up to ~50% hyaluronidase activity in A549 cells. qRT-PCR assay reveals that THF inhibits the activity of phosphatidyl inositol-3 kinase/protein kinase B/mammalian target of rapamycin in both cell lines, which is responsible for the initiation of cancer. It also arrests the G2/M phase of the cell cycle in A431 cells and increases the sub-diploid population in both A549 and A431 cell lines which leads to cell death. Annexin V-FITC assay confirmed that THF induces apoptosis and necrosis in A431 and A549 cell lines. Further investigation revealed that THF not only enhances reactive oxygen species production but also modulates mitochondrial membrane potential in both cell lines. It significantly inhibits S-180 tumour formation at 5 and 10 mg/kg bw, i.p. dose. An acute skin toxicity study on mice showed that erythema and edema scores are within the acceptable range, besides acceptable drug-likeness properties and non-toxic effects on human erythrocytes. Conclusively, THF showed potent anticancer activity on skin and lung carcinoma cell lines, suppressed the level of the biomarkers and inhibited tumour growth in mice.
Collapse
Affiliation(s)
- Waseem Raza
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Jawaharlal Nehru University, New Delhi, India
| | - Abha Meena
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Suaib Luqman
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
2
|
Yuan Z, Wang Y, Yang Y, Qin X. POLR1D silencing suppresses lung cancer cells proliferation and migration via inhibition of PI3K-Akt pathway. J Cardiothorac Surg 2024; 19:322. [PMID: 38844975 PMCID: PMC11155163 DOI: 10.1186/s13019-024-02791-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 05/18/2024] [Indexed: 06/10/2024] Open
Abstract
AIM The most common type of cancer that leads to death worldwide is lung cancer. Despite significant surgery and chemotherapy improvements, lung cancer patient's survival rate is still poor. The RNA polymerase I subunit D (POLR1D) gene can induce various cancers. A current study reported that POLR1D plays a vital role in cancer prognosis. However, its biological function in the development of lung cancer remains unclear. METHODS Reverse transcription PCR (RT-PCR) measured the relative POLR1D protein expression level in lung cancer cell lines. Lung cancer cell proliferation, migration, and invasion were analyzed by performing cell counting kit-8 (CCK-8), and transwell. The phosphatidylinositol 3-kinase/serine-threonine kinase (PI3K/AKT) signaling pathway-related protein expressions were examined by Western blotting assay. RESULTS POLR1D protein expression was elevated in lung cancer. Lung cancer cell loss-of-function tests showed that POLR1D silencing could attenuate cell viability both in SK-MES-1 and in H2170 cells. Furthermore, silencing POLR1D inhibited SK-MES-1 and H2170 cells proliferation, migration, and invasion. Moreover, SK-MES-1 and H2170 cells' migration and invasion capacity were potentially suppressed by the knockdown of POLR1D. The progression of multiple cancers has been implicated in the PI3K/AKT pathway. Here, we observed that POLR1D silencing suppressed lung cancer progression by inhibition of the PI3K-Akt pathway. CONCLUSIONS The study speculated that POLR1D might provide a new potential therapeutic possibility for treating lung cancer patients via targeting PI3K/AKT.
Collapse
Affiliation(s)
- Zhize Yuan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, No. 507, Zhengmin Road, Yangpu District, Shanghai, 200433, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, China
| | - Yichao Wang
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong Yang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, No. 507, Zhengmin Road, Yangpu District, Shanghai, 200433, China
| | - Xiong Qin
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, No. 507, Zhengmin Road, Yangpu District, Shanghai, 200433, China.
| |
Collapse
|
3
|
Zhu M, Chu Z, Dai X, Pan F, Luo Y, Feng X, Hu Y, Wang H, Liu Y. Effect of Celastrus Orbiculatus Extract on proliferation and apoptosis of human Burkitt lymphoma cells. Front Pharmacol 2024; 15:1361371. [PMID: 38633608 PMCID: PMC11021594 DOI: 10.3389/fphar.2024.1361371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 03/19/2024] [Indexed: 04/19/2024] Open
Abstract
The lymphoma incidence rate is on the rise, with invasive forms particularly prone to relapse following conventional treatment, posing a significant threat to human life and wellbeing. Numerous studies have shown that traditional Chinese botanical drug medicine offers promising therapeutic benefits for various malignancies, with previous experimental findings indicating that Celastrus orbiculatus extract effectively combats digestive tract tumors. However, its impact on lymphoma remains unexplored. This study aims to investigate the impact and underlying mechanisms of COE on the proliferation and apoptosis of Burkitt lymphoma cells. We diluted COE in RPMI-1640 medium to create various working concentrations and introduced it to human Burkitt lymphoma Raji and Ramos cells. To evaluate cell viability, we used the CCK-8 assay, and we observed morphological changes using HE staining. We also conducted Annexin V-PI and JC-1 staining experiments to assess apoptosis. By combining the cell cycle experiment with the EDU assay, we gained insights into the effects of COE on DNA replication in lymphoma cells. Using Western blotting, we detected alterations in apoptosis-related proteins. In vivo experiments revealed that following COE intervention, tumor volume decreased, survival time was prolonged, spleen size reduced, and the expression of tumor apoptosis-related proteins changed. Our findings indicate that COE effectively inhibits lymphoma cell proliferation and promotes apoptosis by regulating these apoptosis-related proteins.
Collapse
Affiliation(s)
- Miao Zhu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou, China
- Clinical Medical College of Yangzhou University, Yangzhou, China
| | - Zewen Chu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou, China
| | - Xiaojun Dai
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou, China
- Traditional Chinese Medicine Hospital of Yangzhou, Yangzhou, China
| | - Fan Pan
- Clinical Medical College of Yangzhou University, Yangzhou, China
| | - Yuanyuan Luo
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou, China
| | - Xingyi Feng
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou, China
| | - Yaqi Hu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou, China
| | - Haibo Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou, China
| | - Yanqing Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou, China
| |
Collapse
|
4
|
Feng X, Yin Z, Ou S, Chu Z, Feng J, Luo Y, Hu Y, Liu Y, Jiang W, Wang X, Wang H. The anti-tumor effects of Celastrus orbiculatus Thunb. and its monomer composition: A review. JOURNAL OF ETHNOPHARMACOLOGY 2023; 310:116363. [PMID: 36948266 DOI: 10.1016/j.jep.2023.116363] [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: 12/26/2022] [Revised: 03/04/2023] [Accepted: 03/04/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Celastrus orbiculatus Thunb. has been included in "The Plant List" (http://www. theplantlist.org) and is the most widely researched species in its genus. It is called Nanshe Teng in China. Celastrus orbiculatus Thunb. is a plant of Euonymus and it's medicinal part is the vine and stem. It is also called Alias Dragon grass, Yellow Yine, etc. It has good anti-tumor, anti-inflammatory and other effects. More and more studies have shown that Celastrus orbiculatus Thunb. has a significant therapeutic effect on a variety of malignant tumors. The research on Celastrus orbiculatus Thunb. has a good application prospect for the development of anti-tumor drugs. However, no systematic reports on Celastrus orbiculatus Thunb. have been published before. AIM OF THE REVIEW This paper summarizes the metabolic products for anti-tumor and the mechanism for anti-tumor of Celastrus orbiculatus Thunb. to provide reference for further development and research. MATERIALS AND METHODS The relevant information on Celastrus orbiculatus Thunb. was collected from the scientific databases including PubMed, CNKI, ScienceDirect, Wiley, Springer, Web of Science, Google Scholar, Baidu Scholar, Pharmacopoeia of the People's Republic of China and Flora Republicae Popularis Sinicae, etc. RESULTS: At present, more than 200 compounds have been identified from Celastrus orbiculatus Thunb., including terpenoids, flavonoids, phenylpropanoids, polyketides and benzene derivatives, etc. Pharmacological studies have shown that Celastrus orbiculatus Thunb. has a variety effects of inhibiting tumor cell proliferation, inducing tumor cell apoptosis, inhibiting tumor cells invasion, metastasis and angiogenesis, reversing multi-drug resistance, and also collaborativing Micro RNA to inhibit tumor growth, etc. It has a significant effect on gastric cancer, liver cancer, lung cancer, etc. The extracts of Celastrus orbiculatus Thunb. have been widely used in experiments, and the toxic and side effects are small. CONCLUSIONS Celastrus orbiculatus Thunb. is rich in chemical constituents, diverse in pharmacological activities and abundant in resources, which is widely used in clinics from traditional to modern. However, there is no systematic report on the chemical compounds and anti-tumor effects of Celastrus orbiculatus Thunb. We organize and summarize it to provide reference for further development and research.
Collapse
Affiliation(s)
- Xinyi Feng
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China.
| | - Zixin Yin
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China.
| | - Shiya Ou
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China.
| | - Zewen Chu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China.
| | - Jun Feng
- Department of Oncology, Gaoyou Hospital of Traditional Chinese Medicine, Yangzhou, 225600, PR China.
| | - Yuanyuan Luo
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China.
| | - Yaqi Hu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China.
| | - Yanqing Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China.
| | - Wei Jiang
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China; College of Environmental Science & Engineering, Yangzhou University, Yangzhou, Jiangsu, 225127, China.
| | - Xiaoqing Wang
- Department of Oncology, Gaoyou Hospital of Traditional Chinese Medicine, Yangzhou, 225600, PR China.
| | - Haibo Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China.
| |
Collapse
|
5
|
Jin C, Wang T, Yang Y, Zhou P, Li J, Wu W, Lv X, Ma G, Wang A. Rational targeting of autophagy in colorectal cancer therapy: From molecular interactions to pharmacological compounds. ENVIRONMENTAL RESEARCH 2023; 227:115721. [PMID: 36965788 DOI: 10.1016/j.envres.2023.115721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/13/2023] [Accepted: 03/18/2023] [Indexed: 05/08/2023]
Abstract
The abnormal progression of tumors has been a problem for treatment of cancer and therapeutic should be directed towards targeting main mechanisms involved in tumorigenesis in tumors. The genomic mutations can result in changes in biological mechanisms in human cancers. Colorectal cancer is one of the most malignant tumors of gastrointestinal tract and its treatment has been faced some difficulties due to development of resistance in tumor cells and also, their malignant behavior. Hence, new therapeutic modalities for colorectal cancer are being investigated. Autophagy is a "self-digestion" mechanism that is responsible for homeostasis preserving in cells and its aberrant activation/inhibition can lead to tumorigenesis. The current review focuses on the role of autophagy mechanism in colorectal cancer. Autophagy may be associated with increase/decrease in progression of colorectal cancer due to mutual function of this molecular mechanism. Pro-survival autophagy inhibits apoptosis to increase proliferation and survival rate of colorectal tumor cells and it is also involved in cancer metastasis maybe due to EMT induction. In contrast, pro-death autophagy decreases growth and invasion of colorectal tumor cells. The status of autophagy (upregulation and down-regulation) is a determining factor for therapy response in colorectal tumor cells. Therefore, targeting autophagy can increase sensitivity of colorectal tumor cells to chemotherapy and radiotherapy. Interestingly, nanoparticles can be employed for targeting autophagy in cancer therapy and they can both induce/suppress autophagy in tumor cells. Furthermore, autophagy modulators can be embedded in nanostructures in improving tumor suppression and providing cancer immunotherapy.
Collapse
Affiliation(s)
- Canhui Jin
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China
| | - Tianbao Wang
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China
| | - Yanhui Yang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, 471003, China
| | - Pin Zhou
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China
| | - Juncheng Li
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China
| | - Wenhao Wu
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China
| | - Xin Lv
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China
| | - Guoqing Ma
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China
| | - Aihong Wang
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China.
| |
Collapse
|
6
|
Pan L, Cheng Y, Yang W, Wu X, Zhu H, Hu M, Zhang Y, Zhang M. Nintedanib Ameliorates Bleomycin-Induced Pulmonary Fibrosis, Inflammation, Apoptosis, and Oxidative Stress by Modulating PI3K/Akt/mTOR Pathway in Mice. Inflammation 2023:10.1007/s10753-023-01825-2. [PMID: 37160579 PMCID: PMC10359208 DOI: 10.1007/s10753-023-01825-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/11/2023] [Accepted: 04/23/2023] [Indexed: 05/11/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) seriously threatens human life and health, and no curative therapy is available at present. Nintedanib is the first agent approved by the US Food and Drug Administration (FDA) in order to treat IPF; however, its mechanism of inhibition of IPF is still elusive. According to recent studies, nintedanib is a potent inhibitor. It can antagonize platelet-derived growth factor (PDGF), basic fibroblast growth factor (b-FGF), vascular endothelial growth factor (VEGF), etc., to inhibit pulmonary fibrosis. Whether there are other signaling pathways involved in IPF remains unknown. This study focused on investigating the therapeutic efficacy of nintedanib in bleomycin-mediated pulmonary fibrosis (PF) mice through PI3K/Akt/mTOR pathway. Following the induction of pulmonary fibrosis in C57 mice through bleomycin (BLM) administration, the mice were randomized into five groups: (1) the normal control group, (2) the BLM model control group, (3) the low-dose Nintedanib administration model group, (4) the medium-dose nintedanib administration model group, and (5) the high-dose nintedanib administration model group. For lung tissues, morphological changes were found by HE staining and Masson staining, ELISA method was used to detect inflammatory factors, alkaline water method to estimate collagen content, and western blotting for protein levels. TUNEL staining and immunofluorescence methods were used to analyze the effect of nintedanib on lung tissue and the impacts and underlying mechanisms of bleomycin-induced pulmonary fibrosis. After 28 days, bleomycin-treated mice developed significant pulmonary fibrosis. Relative to bleomycin-treated mice, nintedanib-treated mice had markedly reduced degrees of PF. In addition, nintedanib showed lung-protective effects by up-regulating antioxidant levels, down-regulating inflammatory protein expression, and reducing collagen accumulation. We demonstrated that nintedanib ameliorated bleomycin-induced lung injury by inhibiting the P13K/Akt/mTOR pathway as well as apoptosis. In addition, significant improvement in pulmonary fibrosis was seen after nintedanib (30/60/120 mg/kg body weight/day) treatment through a dose-dependent way. Histopathological results further corroborated the effect of nintedanib treatment on remarkably attenuating bleomycin-mediated mouse lung injury. According to our findings, nintedanib restores the antioxidant system, suppresses pro-inflammatory factors, and inhibits apoptosis. Nintedanib can reduce bleomycin-induced inflammation by downregulating PI3K/Akt/mTOR pathway, PF, and oxidative stress (OS).
Collapse
Affiliation(s)
- Lin Pan
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Yiju Cheng
- Department of Respiratory and Critical Care Medicine, Guiyang First People's Hospital, Guiyang, 550004, China.
- Guizhou Medical University, Guiyang, 550004, China.
| | - Wenting Yang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
| | - Xiao Wu
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Honglan Zhu
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Meigui Hu
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Yuquan Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Menglin Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| |
Collapse
|
7
|
Li P, Yang Y, Yang X, Wang Y, Chou CK, Jiang M, Zheng J, Chen F, Chen X. TNFR2 deficiency impairs the growth of mouse colon cancer. Int J Biol Sci 2023; 19:1024-1035. [PMID: 36923938 PMCID: PMC10008691 DOI: 10.7150/ijbs.72606] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 09/06/2022] [Indexed: 02/04/2023] Open
Abstract
Objective: Tumor necrosis factor (TNF) receptor type II (TNFR2) is expressed by a wide spectrum of tumor cells including colon cancer, non-Hodgkin lymphoma, myeloma, renal carcinoma and ovarian cancer, and its exact role remains to be fully understood. In this study, we examined the effect of genetic ablation of TNFR2 on in vitro and in vivo growth of mouse MC38 and CT26 colon cancer cells. Methods: CRISPR/Cas9 technology was used to knockout TNFR2 on mouse MC38 and CT26 colon cancer cells. In vitro growth and colony formation of wild-type (W.T.) and TNFR2 deficiency of MC38 and CT26 cells, as well as the potential mechanism, was studied. The growth of W.T. and TNFR2 deficient MC38 and CT26 tumors in mice and intratumoral CD8 CTLs were also examined. Results: TNFR2 deficiency impaired in vitro proliferation and colony formation of cancer cells. This was associated with the inhibition of protein kinase B (AKT) phosphorylation and enhanced autophagy-induced cell death. Moreover, deficiency of TNFR2 also markedly impaired in vivo growth of MC38 or CT26 in the syngeneic C57BL/6 mice or BALB/c mice, respectively, accompanied by the decrease in soluble TNFR2 levels in the circulation and the increase in the number of tumor-infiltrating IFNγ+ CD8 cells. Conclusion: TNFR2 plays a role in the growth of mouse colon cancers. Our study provides further experimental evidence to support the development of TNFR2 antagonistic agents in the treatment of cancer.
Collapse
Affiliation(s)
- Ping Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Macau SAR, 999078, P.R. China
| | - Yang Yang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Macau SAR, 999078, P.R. China
| | - Xinyu Yang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Macau SAR, 999078, P.R. China
| | - Yifei Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Macau SAR, 999078, P.R. China
| | - Chon-Kit Chou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Macau SAR, 999078, P.R. China
| | - Mengmeng Jiang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Macau SAR, 999078, P.R. China
| | - Jingbin Zheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Macau SAR, 999078, P.R. China
| | - Fengyang Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Macau SAR, 999078, P.R. China
| | - Xin Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Macau SAR, 999078, P.R. China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macau SAR, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China
- ✉ Corresponding author: Xin Chen, E-mail:
| |
Collapse
|
8
|
Sharma P, Mondal H, Mondal S, Majumder R. Recent updates on the role of phytochemicals in the treatment of glioblastoma multiforme. J Cancer Res Ther 2023; 19:S513-S522. [PMID: 38384013 DOI: 10.4103/jcrt.jcrt_1241_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/07/2022] [Indexed: 02/23/2024]
Abstract
ABSTRACTS Glioblastoma multiforme (GBM) is a malignant type of glioma. This malignant brain tumor is a devastating disease and is often fatal. The spectrum of illness and poor prognosis associated with brain tumors extract a terrible toll on patients and their families. The inoperability of these tumors and resistance to radiation and chemotherapy contribute to the fatal outcome of this disease. Thus, scientists are hunting for the new drug candidate and safer chemoprevention, especially the phytochemicals that possess potent anti-tumor properties. We have summarized the cellular and biochemical impacts of different phytochemicals that can successfully encounter GBM via induction of apoptosis and active interference in different cell and molecular pathways associated with GBM in brain tumors. The in silico predictive model determining the blood-brain barrier permeability of the compound and their potential druggability are discussed in the review.
Collapse
Affiliation(s)
- Pramita Sharma
- Department of Zoology, The University of Burdwan, Burdwan, West Bengal, India
| | - Himel Mondal
- Department of Physiology, All India Institute of Medical Sciences, Deoghar, Jharkhand, India
| | - Shaikat Mondal
- Department of Physiology, Raiganj Government Medical College, Raiganj, West Bengal, India
| | - Rabindranath Majumder
- Centre of Healthcare Science and Technology, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, West Bengal, India
| |
Collapse
|
9
|
Triptonoterpene, a Natural Product from Celastrus orbiculatus Thunb, Has Biological Activity against the Metastasis of Gastric Cancer Cells. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27228005. [PMID: 36432106 PMCID: PMC9693919 DOI: 10.3390/molecules27228005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/15/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022]
Abstract
Cancer is one of the greatest threats to human health. Gastric cancer (GC) is the fifth most common malignant tumor in the world. Invasion and metastasis are the major difficulties in the treatment of GC. Herbal medicines and their extracts have a lengthy history of being used to treat tumors in China. The anti-tumoral effects of the natural products derived from herbs have received a great deal of attention. Our previous studies have shown that the traditional Chinese herb Celastrus orbiculatus Thunb extract (COE) can inhibit the invasion and metastasis of GC cells, but the specific anti-cancer components of COE are still unclear. Dozens of natural products from COE have been isolated and identified by HPLC spectroscopy in our previous experiments. Triptonoterpene is one of the active ingredients in COE. In this study, we focused on revealing whether Triptonoterpene has an excellent anti-GC effect and can be used as an effective component of Celastrus orbiculatus Thunb in the treatment of tumors. We first observed that Triptonoterpene reduces GC cell proliferation through CCK-8 assays and colony formation experiments. The cell adhesion assays have shown that Triptonoterpene inhibits adhesion between cells and the cell matrix during tumor invasion. In addition, the cell migration assay has shown that Triptonoterpene inhibits the invasion and migration of GC cells. The high-connotation cell dynamic tracking experiment has also shown the same results. The effects of Triptonoterpene on epidermal mesenchymal transition (EMT)-related and matrix metalloproteinases (MMPs)-related proteins in gastric cancer cells were detected by Western blots. We found that Triptonoterpene could significantly inhibit the changes in EMT-related and invasion and metastasis-related proteins. Altogether, these results suggest that Triptonoterpene is capable of inhibiting the migration and invasion of GC cells. Triptonoterpene, as a natural product from Celastrus orbiculatus Thunb, has significant anti-gastric cancer effects, and is likely to be one of the major equivalent components of Celastrus orbiculatus Thunb.
Collapse
|
10
|
Gu G, Jiang M, Hu H, Qiao W, Jin H, Hou T, Tao K. Neochamaejasmin B extracted from Stellera chamaejasme L. induces apoptosis through caspase-10-dependent way in insect neuronal cells. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2022; 110:e21892. [PMID: 35478464 DOI: 10.1002/arch.21892] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/09/2022] [Accepted: 03/12/2022] [Indexed: 06/14/2023]
Abstract
To explore the toxicity mechanisms of neochamaejasmin B (NCB) extracted from Stellera chamaejasme L., we first evaluated its cytotoxicity in neuronal cells of Helicoverpa zea (AW1 cells). NCB inhibited cell growth and was cytotoxic to AW1 cells in a dose-dependent manner. Further, transmission electron microscopy (TEM) was used to analyze the microstructure, and typical apoptotic characteristics were observed in AW1 cells treated with NCB. Moreover, the NCB-induced apoptosis was dose dependent. Subsequently, we explored the mechanism of apoptosis. A decline in the mitochondrial membrane potential (MMP) was found. Also, the levels of Bax were increased with increases in drug concentration, but there was no statistical difference in Bcl-2 levels at different NCB doses. Caspase-3 and caspase-10 activity was increased. These findings confirmed that NCB induced apoptosis in AW1 cells through a caspase-10-dependent mechanism. The results provide the basic information needed for understanding the toxicity and mechanisms of action of NCB, which could potentially be used to develop NCB as a new insecticide.
Collapse
Affiliation(s)
- Guirong Gu
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Mingfang Jiang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Hanying Hu
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Weijie Qiao
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Hong Jin
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Taiping Hou
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Ke Tao
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, P.R. China
| |
Collapse
|
11
|
Chu Z, Luo Y, Ni T, Zhu M, Feng X, Liu Y, Wang H. Betulonic Acid, as One of the Active Components of the Celastrus orbiculatus Extract, Inhibits the Invasion and Metastasis of Gastric Cancer Cells by Mediating Cytoskeleton Rearrangement In Vitro. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27031025. [PMID: 35164287 PMCID: PMC8840099 DOI: 10.3390/molecules27031025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/28/2022] [Accepted: 01/31/2022] [Indexed: 12/23/2022]
Abstract
Gastric cancer is a type of malignant tumor that seriously threatens human life and health. Invasion and metastasis present difficulties in the treatment of gastric cancer, and the remodeling of the tumor cytoskeleton plays an important role in mediating the ability of tumor cells to achieve invasion and metastasis. Previous experimental results suggest that Celastrus orbiculatus extract can regulate cytoskeletal remodeling in gastric cancer, but the active component has not been determined. Betulonic acid, as an effective component of COE, inhibits the invasion and metastasis of gastric cancer cells by regulating cytoskeletal remodeling in vitro; its specific mechanisms have been studied here. After betulonic acid was dissolved, it was diluted to various working concentrations in RPMI-1640 medium and added to AGS, HGC-27 and GES-1 cell lines. Cell viability was assessed by CCK-8 and colony formation assays. Cytoskeleton staining was used to detect changes in cytoskeleton morphology. Functional assays including wound healing assays and transwell assays were used to detect the invasion and migration of cells. The effect of betulonic acid on cell invasion and migration was clearly and precisely observed by high-content imaging technology. Western blotting was used to detect the regulation of matrix metalloproteinase-related proteins and epithelial–mesenchymal transformation-related proteins. We found that betulonic acid inhibited the migration and invasion of gastric cancer cells. Therefore, betulonic acid inhibits the invasion and metastasis of gastric cancer cells by mediating cytoskeletal remodeling and regulating epithelial mesenchymal transformation.
Collapse
Affiliation(s)
- Zewen Chu
- Department of Integrated Chinese and Western Medicine, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China; (Z.C.); (Y.L.); (T.N.); (M.Z.); (X.F.)
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Department of Integrated Chinese and Western Medicine, Yangzhou University, Yangzhou 225001, China
- Department of Integrated Chinese and Western Medicine, Yangzhou Cancer Research Institute, Yangzhou University, Yangzhou 225001, China
| | - Yuanyuan Luo
- Department of Integrated Chinese and Western Medicine, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China; (Z.C.); (Y.L.); (T.N.); (M.Z.); (X.F.)
| | - Tengyang Ni
- Department of Integrated Chinese and Western Medicine, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China; (Z.C.); (Y.L.); (T.N.); (M.Z.); (X.F.)
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Department of Integrated Chinese and Western Medicine, Yangzhou University, Yangzhou 225001, China
- Department of Integrated Chinese and Western Medicine, Yangzhou Cancer Research Institute, Yangzhou University, Yangzhou 225001, China
| | - Miao Zhu
- Department of Integrated Chinese and Western Medicine, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China; (Z.C.); (Y.L.); (T.N.); (M.Z.); (X.F.)
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Department of Integrated Chinese and Western Medicine, Yangzhou University, Yangzhou 225001, China
- Department of Integrated Chinese and Western Medicine, Yangzhou Cancer Research Institute, Yangzhou University, Yangzhou 225001, China
| | - Xinyi Feng
- Department of Integrated Chinese and Western Medicine, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China; (Z.C.); (Y.L.); (T.N.); (M.Z.); (X.F.)
| | - Yanqing Liu
- Department of Integrated Chinese and Western Medicine, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China; (Z.C.); (Y.L.); (T.N.); (M.Z.); (X.F.)
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Department of Integrated Chinese and Western Medicine, Yangzhou University, Yangzhou 225001, China
- Department of Integrated Chinese and Western Medicine, Yangzhou Cancer Research Institute, Yangzhou University, Yangzhou 225001, China
- Correspondence: (Y.L.); (H.W.)
| | - Haibo Wang
- Department of Integrated Chinese and Western Medicine, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China; (Z.C.); (Y.L.); (T.N.); (M.Z.); (X.F.)
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Department of Integrated Chinese and Western Medicine, Yangzhou University, Yangzhou 225001, China
- Department of Integrated Chinese and Western Medicine, Yangzhou Cancer Research Institute, Yangzhou University, Yangzhou 225001, China
- Correspondence: (Y.L.); (H.W.)
| |
Collapse
|
12
|
Sanaei MJ, Baghery Saghchy Khorasani A, Pourbagheri-Sigaroodi A, Shahrokh S, Zali MR, Bashash D. The PI3K/Akt/mTOR axis in colorectal cancer: Oncogenic alterations, non-coding RNAs, therapeutic opportunities, and the emerging role of nanoparticles. J Cell Physiol 2021; 237:1720-1752. [PMID: 34897682 DOI: 10.1002/jcp.30655] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/02/2021] [Accepted: 11/24/2021] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) is one of the deadliest human malignancies worldwide. Several molecular pathways have been demonstrated to be involved in the initiation and development of CRC which among them, the overactivation of the phosphatidyl-inositol 3-kinase (PI3K)/Akt/mTOR axis is of importance. The current review aims to unravel the mechanisms by which the PI3K/Akt/mTOR pathway affects CRC progression; and also, to summarize the original data obtained from international research laboratories on the oncogenic alterations and polymorphisms affecting this pathway in CRC. Besides, we provide a special focus on the regulatory role of noncoding RNAs targeting the PI3K/Akt/mTOR pathway in this malignancy. Questions on how this axis is involved in the inhibition of apoptosis, in the induction of drug resistance, and the angiogenesis, epithelial to mesenchymal transition, and metastasis are also responded. We also discussed the PI3K/Akt pathway-associated prognostic and predictive biomarkers in CRC. In addition, we provide a general overview of PI3K/Akt/mTOR pathway inhibition whether by chemical-based drugs or by natural-based medications in the context of CRC, either as monotherapy or in combination with other therapeutic agents; however, those treatments might have life-threatening side effects and toxicities. To the best of our knowledge, the current review is one of the first ones highlighting the emerging roles of nanotechnology to overcome challenges related to CRC therapy in the hope that providing a promising platform for the treatment of CRC.
Collapse
Affiliation(s)
- Mohammad-Javad Sanaei
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shabnam Shahrokh
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Li H, Liu Y, Tang S, Hu J, Wu Q, Wei Y, Niu M. Carbonic Anhydrase III Attenuates Hypoxia-Induced Apoptosis and Activates PI3K/Akt/mTOR Pathway in H9c2 Cardiomyocyte Cell Line. Cardiovasc Toxicol 2021; 21:914-926. [PMID: 34387844 DOI: 10.1007/s12012-021-09683-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022]
Abstract
Myocardial ischemia can cause insufficient oxygen and functional damage to myocardial cells. Carbonic anhydrase III (CAIII) has been found to be closely related to the abnormality of cardiomyocytes. To investigate the role of CAIII in the apoptosis of myocytes under hypoxic conditions and facilitate the strategy for treating hypoxia-induced damage, in vitro experiments in H9c2 were employed. The protein expression of CAIII in H9c2 cells after hypoxia or normoxia treatment was determined by western blotting and immunohistochemistry. MTT assay was employed for cells viability measurement and LDH release was monitored. The apoptotic cells were observed using immunofluorescence assay, flow cytometric analysis, and TUNEL assay. CAIII-overexpression or -knockdown cells were constructed to determine the role of CAIII in regulating apoptosis-related proteins caspase-3, Bax, Bcl-2, and anti-apoptosis pathway PI3K/Akt/mTOR. The mRNA levels of CAIII and genes related to CAIII synthesis including REN, IGHM, APOBEC 3F, and SKOR2 were significantly upregulated in hypoxia fetal sheep. The expression of CAIII protein and content of apoptotic H9c2 cells were increased at 1, 3, 6, and 12 h after hypoxia treatment. Overexpression of CAIII significantly upregulated Bcl2 level and downregulated Bax and caspase-3 cleavage levels, while its knockdown led to the contrary results. Overexpressed CAIII promoted the HIF-1α level and activated the PI3K/Akt/mTOR pathway, thereby exerting an inhibitory effect on hypoxia-induced apoptosis. In conclusion, our findings revealed that CAIII could protect cell from hypoxia-apoptosis of H9c2 cells, in which, activated PI3K/Akt/mTOR signaling pathway may be involved.
Collapse
Affiliation(s)
- Hua Li
- Cardiac Ultrasonic Department, Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, No. 116 Huanghe Road, Shayibake District, Ürümqi, 830002, Xinjiang, China.
| | - Yibin Liu
- Ultrasonic Department, First Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830011, Xinjiang, China
| | - Sha Tang
- Cardiac Ultrasonic Department, Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, No. 116 Huanghe Road, Shayibake District, Ürümqi, 830002, Xinjiang, China
| | - Jie Hu
- Cardiac Ultrasonic Department, Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, No. 116 Huanghe Road, Shayibake District, Ürümqi, 830002, Xinjiang, China
| | - Qiuling Wu
- Cardiac Ultrasonic Department, Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, No. 116 Huanghe Road, Shayibake District, Ürümqi, 830002, Xinjiang, China
| | - Yang Wei
- Cardiac Ultrasonic Department, Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, No. 116 Huanghe Road, Shayibake District, Ürümqi, 830002, Xinjiang, China
| | - Ming Niu
- Cardiac Ultrasonic Department, Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, No. 116 Huanghe Road, Shayibake District, Ürümqi, 830002, Xinjiang, China
| |
Collapse
|
14
|
Wang Z, Liu C. Upregulated hsa_circRNA_100269 inhibits the growth and metastasis of gastric cancer through inactivating PI3K/Akt axis. PLoS One 2021; 16:e0250603. [PMID: 33901239 PMCID: PMC8075232 DOI: 10.1371/journal.pone.0250603] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/11/2021] [Indexed: 01/10/2023] Open
Abstract
The pathogenesis of GC involves the complex networking of multiple signaling pathways; however, the detailed mechanisms of tumorigenesis of GC remains largely unknown. Therefore, it is necessary to explore novel diagnostic/prognostic biomarkers for GC. In this study, the levels of hsa_circRNA_100269 in gastric cancer (GC) samples and cells were examined, and its effects on the biological functions of GC cells were elucidated. The levels of hsa_circRNA_100269 in specimens/cell lines were examined using RT-qPCR. Cell models with hsa_circRNA_100269 overexpression or knockdown were generated using lentiviral vectors. Cell viability was determined by MTT assay; cell migratory/invasive activity was evaluated using wound healing/Transwell assay. Cell cycle arrest and apoptosis were assessed by flow cytometry; expression of associated markers involved in cell apoptosis, EMT and the PI3K/Akt signaling were determined by RT-qPCR/immunoblotting. In vivo study was also performed using hsa_circRNA_100269 knockout mice. Our findings revealed downregulation of hsa_circRNA_100269 in GC tissues compared to non-cancerous control. Additionally, the levels of PI3K were remarkably elevated in GC tissues, where hsa_circRNA_100269 and PI3K was negatively correlated. Moreover, the expression of hsa_circRNA_100269 was associated with histology grade and occurrence of metastasis in GC patients. In addition, hsa_circRNA_100269 was downregulated in GC cells compared to normal gastric epithelial cells. Overexpressed hsa_circRNA_100269 notably inhibited the proliferation, migration, invasion and EMT of GC cells, whereas cell cycle arrest at G0/G1 phase was promoted and cell apoptosis was enhanced. Moreover, the PI3K/Akt signaling was involved in hsa_circRNA_100269-regulated GC cell proliferation, migration, invasion, EMT and apoptosis. Knockdown of hsa_circRNA_100269 also remarkably induced tumor growth in mouse model. In summary, our findings indicated that the levels of hsa_circRNA_100269 were reduced in GC. Furthermore, hsa_circRNA_100269 could suppress the development of GC by inactivating the PI3K/Akt pathway. More importantly, hsa_circRNA_100269/PI3K/Akt axis may be a novel therapeutic candidate for GC treatment.
Collapse
Affiliation(s)
- Zhongli Wang
- Department of General Surgery, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, P.R. China
| | - Chao Liu
- Department of Developmental Biology, Jinzhou Medical University, Jinzhou, Liaoning, P.R. China
- * E-mail:
| |
Collapse
|
15
|
Bone marrow-derived mesenchymal stem cells modulate autophagy in RAW264.7 macrophages via the phosphoinositide 3-kinase/protein kinase B/heme oxygenase-1 signaling pathway under oxygen-glucose deprivation/restoration conditions. Chin Med J (Engl) 2021; 134:699-707. [PMID: 33605598 PMCID: PMC7989993 DOI: 10.1097/cm9.0000000000001133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background Autophagy of alveolar macrophages is a crucial process in ischemia/reperfusion injury-induced acute lung injury (ALI). Bone marrow-derived mesenchymal stem cells (BM-MSCs) are multipotent cells with the potential for repairing injured sites and regulating autophagy. This study was to investigate the influence of BM-MSCs on autophagy of macrophages in the oxygen-glucose deprivation/restoration (OGD/R) microenvironment and to explore the potential mechanism. Methods We established a co-culture system of macrophages (RAW264.7) with BM-MSCs under OGD/R conditions in vitro. RAW264.7 cells were transfected with recombinant adenovirus (Ad-mCherry-GFP-LC3B) and autophagic status of RAW264.7 cells was observed under a fluorescence microscope. Autophagy-related proteins light chain 3 (LC3)-I, LC3-II, and p62 in RAW264.7 cells were detected by Western blotting. We used microarray expression analysis to identify the differently expressed genes between OGD/R treated macrophages and macrophages co-culture with BM-MSCs. We investigated the gene heme oxygenase-1 (HO-1), which is downstream of the phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) signaling pathway. Results The ratio of LC3-II/LC3-I of OGD/R treated RAW264.7 cells was increased (1.27 ± 0.20 vs. 0.44 ± 0.08, t = 6.67, P < 0.05), while the expression of p62 was decreased (0.77 ± 0.04 vs. 0.95 ± 0.10, t = 2.90, P < 0.05), and PI3K (0.40 ± 0.06 vs. 0.63 ± 0.10, t = 3.42, P < 0.05) and p-Akt/Akt ratio was also decreased (0.39 ± 0.02 vs. 0.58 ± 0.03, t = 9.13, P < 0.05). BM-MSCs reduced the LC3-II/LC3-I ratio of OGD/R treated RAW264.7 cells (0.68 ± 0.14 vs. 1.27 ± 0.20, t = 4.12, P < 0.05), up-regulated p62 expression (1.10 ± 0.20 vs. 0.77 ± 0.04, t = 2.80, P < 0.05), and up-regulated PI3K (0.54 ± 0.05 vs. 0.40 ± 0.06, t = 3.11, P < 0.05) and p-Akt/Akt ratios (0.52 ± 0.05 vs. 0.39 ± 0.02, t = 9.13, P < 0.05). A whole-genome microarray assay screened the differentially expressed gene HO-1, which is downstream of the PI3K/Akt signaling pathway, and the alteration of HO-1 mRNA and protein expression was consistent with the data on PI3K/Akt pathway. Conclusions Our results suggest the existence of the PI3K/Akt/HO-1 signaling pathway in RAW264.7 cells under OGD/R circumstances in vitro, revealing the mechanism underlying BM-MSC-mediated regulation of autophagy and enriching the understanding of potential therapeutic targets for the treatment of ALI.
Collapse
|
16
|
Li JQ, Li JL, Xie YH, Wang Y, Shen XN, Qian Y, Han JX, Chen YX, Fang JY. Saccharomyces cerevisiae may serve as a probiotic in colorectal cancer by promoting cancer cell apoptosis. J Dig Dis 2020; 21:571-582. [PMID: 33245627 DOI: 10.1111/1751-2980.12930] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 07/20/2020] [Accepted: 08/12/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Shotgun metagenomic sequencing of human fecal samples has shown that Saccharomyces cerevisiae (S. cerevisiae) is significantly suppressed in colorectal cancer (CRC) and probably plays an important role in CRC progression. However, these results need to be validated. Here we aimed to confirm the results of high-throughput sequencing and demonstrate the mechanisms mediating the effect of S. cerevisiae on progression from colorectal adenoma (CRA) to CRC. METHODS We used a quantitative polymerase chain reaction (qPCR) assay to examine the relative abundance of S. cerevisiae in 281 fecal samples collected from 106 healthy controls, 108 patients with CRA and 67 with CRC. C57BL/6 and APCMin/+ mouse models and in vitro cell assays were subsequntly used for additional analyses. The mouse models were treated or not treated with broad-spectrum antibiotics and given an S. cerevisiae gavage for 8 weeks. Western blot, 16S rRNA sequencing, qPCR, immunohistochemistry, RNA sequencing, cell counting kit-8 assay, colony formation assay and flow cytometry were performed. RESULTS S. cerevisiae was 2.68-fold and 3.94-fold less abundant in patients with CRA and CRC, respectively, than in the controls. In vivo experiments showed that S. cerevisiae reduced colorectal tumor progression by promoting epithelial cell apoptosis and modulated gut microbial structure and intestinal immunity. S. cerevisiae downregulated nuclear factor kappa light chain enhancer of activated B cells and the mechanistic target of rapamycin signaling pathways. Cell assays confirmed the pro-apoptotic effect of S. cerevisiae. CONCLUSIONS S. cerevisiae may play a probiotic role in CRC by promoting cancer cell apoptosis. It can reduce CRC progression by modulating the mucosal microbial structure.
Collapse
Affiliation(s)
- Jia Qi Li
- Division of Gastroenterology and Hepatology, Shanghai Cancer Institute, State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jia Lu Li
- Division of Gastroenterology and Hepatology, Shanghai Cancer Institute, State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuan Hong Xie
- Division of Gastroenterology and Hepatology, Shanghai Cancer Institute, State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yao Wang
- Division of Gastroenterology and Hepatology, Shanghai Cancer Institute, State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao Nan Shen
- Division of Gastroenterology and Hepatology, Shanghai Cancer Institute, State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yun Qian
- Division of Gastroenterology and Hepatology, Shanghai Cancer Institute, State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ji Xuan Han
- Division of Gastroenterology and Hepatology, Shanghai Cancer Institute, State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Xuan Chen
- Division of Gastroenterology and Hepatology, Shanghai Cancer Institute, State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, Shanghai Cancer Institute, State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
17
|
Tan S, Yang S, Chen G, Zhu L, Sun Z, Chen S. Trehalose alleviates apoptosis by protecting the autophagy-lysosomal system in alveolar macrophages during human silicosis. Life Sci 2020; 257:118043. [PMID: 32621922 DOI: 10.1016/j.lfs.2020.118043] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/12/2020] [Accepted: 06/29/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Alveolar macrophages (AMs) are the primary targets of silicosis. Blockade of autophagy may aggravate the apoptosis of AMs. Trehalose (Tre), a transcription factor EB (TFEB) activator, may impact the autophagy-lysosomal system in AMs during silicosis. However, the mechanism by which Tre acts upon AMs in silicosis is unknown. METHODS We collected AMs from twenty male workers exposed to silica and divided them into observer and silicosis patient groups. AMs from the two groups were then exposed to Tre. Western blot was used to measure the expression of autophagy-associated proteins. Lysosomal-associated membrane protein 1 (LAMP1) expression was observed using immunofluorescence and western blot. Apoptosis of the AMs was detected by TUNEL assay and western blot. RESULTS Tre induced localization of TFEB to the nucleus in the AMs of both groups. After Tre exposure, LAMP1 levels increased and LC3 levels decreased in the AMs of both groups, suggesting that Tre may increase the function of the autophagy-lysosomal system. The LC3-II/I ratio in the Tre-exposed AMs was lower than in the AMs not exposed to Tre. The LC3-II/I ratio in AMs subjected to Tre plus Bafilomycin (Baf) was higher than the ratio in cells exposed to Tre or Baf individually. Additionally, p62 levels decreased after Tre stimulation in the AMs of both groups. This indicates that Tre may accelerate the process of autophagic degradation. We also found decreased levels of cleaved caspase-3 after Tre treatment in the AMs of both groups. However, p-mTOR (Ser2448) and p-mTOR (Ser2481) levels did not change significantly after Tre treatment, suggesting that the mTOR signaling pathway was not affected by Tre treatment. CONCLUSION Our findings suggest that the restoration of autophagy-lysosomal function by Tre may be a potential protective strategy against silicosis.
Collapse
Affiliation(s)
- Shiyi Tan
- School of Medicine, Hunan Normal University, Changsha, Hunan Province 410013, PR China; Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University Changsha, Hunan Province 410013, PR China
| | - Shang Yang
- School of Medicine, Hunan Normal University, Changsha, Hunan Province 410013, PR China; Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University Changsha, Hunan Province 410013, PR China
| | - Gang Chen
- Department of Pneumoconiosis, Beidaihe Sanitarium for China Coal Miners, Qinhuangdao, Hebei Province 066104, PR China
| | - Li Zhu
- Department of Pneumoconiosis, Beidaihe Sanitarium for China Coal Miners, Qinhuangdao, Hebei Province 066104, PR China
| | - Zhiqian Sun
- Department of Pneumoconiosis, Beidaihe Sanitarium for China Coal Miners, Qinhuangdao, Hebei Province 066104, PR China
| | - Shi Chen
- School of Medicine, Hunan Normal University, Changsha, Hunan Province 410013, PR China.
| |
Collapse
|
18
|
Kim SY, Yi HK, Yun BS, Lee DY, Hwang PH, Park HR, Kim MS. The extract of the immature fruit of Poncirus trifoliata induces apoptosis in colorectal cancer cells via mitochondrial autophagy. FOOD SCIENCE AND HUMAN WELLNESS 2020. [DOI: 10.1016/j.fshw.2020.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
19
|
Li C, Gao H, Feng X, Bi C, Zhang J, Yin J. Ginsenoside Rh2 impedes proliferation and migration and induces apoptosis by regulating NF-κB, MAPK, and PI3K/Akt/mTOR signaling pathways in osteosarcoma cells. J Biochem Mol Toxicol 2020; 34:e22597. [PMID: 32762018 DOI: 10.1002/jbt.22597] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/07/2020] [Accepted: 07/23/2020] [Indexed: 12/14/2022]
Abstract
Ginsenoside Rh2 is a primary bioactive compound obtained from ginseng that indicated anticancer activities against several malignant tumors. However, previous studies have reported little about the inhibitory effect of Rh2 on osteosarcoma (OS). This study aims to explore whether Rh2 could exert anticancer effects in OS cells and further investigate the proliferation, migration, and apoptosis mechanisms induced by Rh2 in human OS U20S cell line. The viability of U20S cells was obtained by the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide assay. Cell migration property was analyzed by wound-healing assay. Apoptosis was visualized using terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL), 4',6-diamidino-2-phenylindole (DAPI), and annexin V/propidium iodide (PI) staining. Relative protein expressed was confirmed through Western blot analysis. Mitochondrial membrane potential was evaluated by JC-1 staining. In this study, we used broad-spectrum anticancer drug cisplatin (CP) as a positive control. The results indicated that Rh2 remarkably inhibited cell viability of U20S cells in a dose- and time-dependent manner, and suppressed migration. TUNEL, DAPI, annexin V/PI, and JC-1 assay suggested that Rh2 could induce cellular apoptosis. Rh2 could reduce the levels of Bcl-2, caspase 3, and caspase 9, and promote the expression level of Bax in U20S cells. Moreover, Rh2 could induce apoptosis by promoting mitogen-activated protein kinase (MAPK) signaling pathway and inhibit PI3K/Akt/mTOR and nuclear factor-κB (NF-κB) signaling pathway in U20S cells. These findings indicated that Rh2 has an anticancer effect on U20S cells by regulating MAPK, PI3K/Akt/mTOR, and NF-κB signaling pathway.
Collapse
Affiliation(s)
- Chenchen Li
- Department of Natural Products Chemistry, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Huan Gao
- Department of Pharmacy, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xuemei Feng
- Department of Natural Products Chemistry, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Chuyao Bi
- Department of Natural Products Chemistry, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Jing Zhang
- Department of Natural Products Chemistry, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Jianyuan Yin
- Department of Natural Products Chemistry, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| |
Collapse
|
20
|
Anti-Inflammatory Activity of Diterpenoids from Celastrus orbiculatus in Lipopolysaccharide-Stimulated RAW264.7 Cells. J Immunol Res 2020; 2020:7207354. [PMID: 32802895 PMCID: PMC7414338 DOI: 10.1155/2020/7207354] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 04/29/2020] [Accepted: 05/07/2020] [Indexed: 12/14/2022] Open
Abstract
Celastrus orbiculatus Thunb has been known as an ethnopharmacological medicinal plant for antitumor, anti-inflammatory, and analgesic effects. Although various pharmacological studies of C. orbiculatus extract has been reported, an anti-inflammatory mechanism study of their phytochemical constituents has not been fully elucidated. In this study, compounds 1-17, including undescribed podocarpane-type trinorditerpenoid (3), were purified from C. orbiculatus and their chemical structure were determined by high-resolution electrospray ionization mass (HRESIMS) and nuclear magnetic resonance (NMR) spectroscopic data. To investigate the anti-inflammatory activity of compounds 1-17, nitric oxide (NO) secretion was evaluated in LPS-treated murine macrophages, RAW264.7 cells. Among compounds 1-17, deoxynimbidiol (1) and new trinorditerpenoid (3) showed the most potent inhibitory effects (IC50: 4.9 and 12.6 μM, respectively) on lipopolysaccharide- (LPS-) stimulated NO releases as well as proinflammatory mediators, such as inducible nitric oxide (iNOS), cyclooxygenase- (COX-) 2, interleukin- (IL-) 1β, IL-6, and tumor necrosis factor- (TNF-) α. Its inhibitory activity of proinflammatory mediators is contributed by suppressing the activation of nuclear transcription factor- (NF-) κB and mitogen-activated protein kinase (MAPK) signaling cascades including p65, inhibition of NF-κB (IκB), extracellular signal-regulated kinase (ERK), c-Jun NH2-terminal kinase (JNK), and p38. Therefore, these results demonstrated that diterpenoids 1 and 3 obtained from C. orbiculatus may be considered a potential candidate for the treatment of inflammatory diseases.
Collapse
|
21
|
Kim OH, Lee JH, Mah S, Park SY, Hong S, Hong SS. HS‑146, a novel phosphoinositide 3‑kinase α inhibitor, induces the apoptosis and inhibits the metastatic ability of human breast cancer cells. Int J Oncol 2020; 56:1509-1520. [PMID: 32236634 DOI: 10.3892/ijo.2020.5018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 02/21/2020] [Indexed: 11/06/2022] Open
Abstract
The phosphoinositide 3‑kinase (PI3K) signaling pathway plays an important role in human cancer as it regulates critical cellular functions, such as survival, proliferation and metabolism. In the present study, a novel PI3Kα inhibitor (HS‑146) was synthesized and its anticancer effects on MCF‑7, MDA‑MB‑231, SKBR3 and BT‑474 human breast cancer cell lines were confirmed. HS‑146 was found to be most effective in inhibiting the proliferation of MCF‑7 cells and in inducing cell cycle arrest in the G0/G1 phase by downregulating cyclin D1, cyclin E, cyclin‑dependent kinase (Cdk)2 and Cdk4, and upregulating p21Waf1/Cip1 protein levels in this cell line. The induction of apoptosis by HS‑146 was confirmed by DAPI staining and western blot analysis. Cell shrinkage and nuclear condensation, which are typical morphological markers of apoptosis, were increased by HS‑146 in the MCF‑7 cells in a concentration‑dependent manner, and HS‑146 also increased the protein expression levels of cleaved poly(ADP‑ribose) polymerase (PARP) and decreased the protein expression levels of Mcl‑1 and caspase‑7. In addition, HS‑146 effectively decreased the phosphorylation levels of downstream PI3K effectors, such as Akt, mammalian target of rapamycin (mTOR), glycogen synthase kinase 3β (GSK3β), p70S6K1 and eukaryotic translation initiation factor 4E‑binding protein 1 (4E‑BP1). Hypoxia‑inducible factor (HIF)‑1α and vascular endothelial growth factor (VEGF) expression were also suppressed by HS‑146 under hypoxic conditions, and HS‑146 inhibited the migration and invasion of MCF‑7 cells in a concentration‑dependent manner. On the whole, the findings of the present study suggest that HS‑146, a novel PI3Kα inhibitor, may be an effective novel therapeutic candidate that suppresses breast cancer proliferation and metastasis by inhibiting the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Ok Hyeon Kim
- College of Korean Medicine, Dongguk University, Goyang, Gyeonggi 10326, Republic of Korea
| | - Ju-Hee Lee
- College of Korean Medicine, Dongguk University, Goyang, Gyeonggi 10326, Republic of Korea
| | - Shinmee Mah
- Center for Catalytic Hydrocarbon Functionalizations, Institute of Basic Science (IBS), Daejeon 34141, Republic of Korea
| | - Sung Yun Park
- College of Korean Medicine, Dongguk University, Goyang, Gyeonggi 10326, Republic of Korea
| | - Sungwoo Hong
- Center for Catalytic Hydrocarbon Functionalizations, Institute of Basic Science (IBS), Daejeon 34141, Republic of Korea
| | - Soon-Sun Hong
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon 22332, Republic of Korea
| |
Collapse
|
22
|
Jiang W, Kai J, Li D, Wei Z, Wang Y, Wang W. lncRNA HOXB-AS3 exacerbates proliferation, migration, and invasion of lung cancer via activating the PI3K-AKT pathway. J Cell Physiol 2020; 235:7194-7203. [PMID: 32039488 DOI: 10.1002/jcp.29618] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 01/24/2020] [Indexed: 12/17/2022]
Abstract
Lung cancer remains the leading cause of cancer-related death all over the world. In spite of the great advances made in surgery and chemotherapy, the prognosis of lung cancer patients is poor. A substantial fraction of long noncoding RNAs (lncRNAs) can regulate various cancers. A recent study has reported that lncRNA HOXB-AS3 plays a critical role in cancers. However, its biological function remains unclear in lung cancer progression. In the current research, we found HOXB-AS3 was obviously elevated in NSCLC tissues and cells. Functional assays showed that inhibition of HOXB-AS3 was able to repress A549 and H1975 cell proliferation, cell colony formation ability and meanwhile, triggered cell apoptosis. Furthermore, the lung cancer cell cycle was mostly blocked in the G1 phase whereas the cell ratio in the S phase was reduced. Also, A549 and H1975 cell migration and invasion capacity were significantly repressed by the loss of HOXB-AS3. The PI3K/AKT pathway has been implicated in the carcinogenesis of multiple cancers. Here, we displayed that inhibition of HOXB-AS3 suppressed lung cancer cell progression via inactivating the PI3K/AKT pathway. Subsequently, in vivo experiments were utilized in our study and it was demonstrated that HOXB-AS3 contributed to lung cancer tumor growth via modulating the PI3K/AKT pathway. Overall, we implied that HOXB-AS3 might provide a new perspective for lung cancer treatment via targeting PI3K/AKT.
Collapse
Affiliation(s)
- Wenyang Jiang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jindan Kai
- Department of Thoracic Surgery, Hubei Cancer Hospital, Wuhan, China
| | - Donghang Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhongheng Wei
- Department of Oncology, Affiliated Hospital of Youjiang Medical College for Nationalities, Baise, Guangxi, China
| | - Ying Wang
- Department of Respiratory Medicine, The First People's Hospital of Tianmen, Tianmen, Hubei, China
| | - Wei Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
23
|
Tewari D, Patni P, Bishayee A, Sah AN, Bishayee A. Natural products targeting the PI3K-Akt-mTOR signaling pathway in cancer: A novel therapeutic strategy. Semin Cancer Biol 2019; 80:1-17. [PMID: 31866476 DOI: 10.1016/j.semcancer.2019.12.008] [Citation(s) in RCA: 302] [Impact Index Per Article: 60.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/01/2019] [Accepted: 12/03/2019] [Indexed: 02/07/2023]
Abstract
The phosphatidylinositol 3-kinase (PI3K)-Akt and the mammalian target of rapamycin (mTOR) represent two vital intracellular signaling pathways, which are associated with various aspects of cellular functions. These functions play vital roles in quiescence, survival, and growth in normal physiological circumstances as well as in various pathological disorders, including cancer. These two pathways are so intimately connected to each other that in some instances these are considered as one unique pathway crucial for cell cycle regulation. The purpose of this review is to emphasize the role of PI3K-Akt-mTOR signaling pathway in different cancer conditions and the importance of natural products targeting the PI3K-Akt-mTOR signaling pathway. This review also aims to draw the attention of scientists and researchers to the assorted beneficial effects of the numerous classes of natural products for the development of new and safe drugs for possible cancer therapy. We also summarize and critically analyze various preclinical and clinical studies on bioactive compounds and constituents, which are derived from natural products, to target the PI3K-Akt-mTOR signaling pathway for cancer prevention and intervention.
Collapse
Affiliation(s)
- Devesh Tewari
- Department of Pharmacognosy, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144 411, Punjab, India.
| | - Pooja Patni
- Sharda School of Pharmacy, Gujarat Technical University, Gandhinagar 382 610, Gujarat, India
| | | | - Archana N Sah
- Department of Pharmaceutical Sciences, Faculty of Technology, Bhimtal Campus, Kumaun University, Nainital 263 136, Uttarakhand, India
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA.
| |
Collapse
|
24
|
Xu Z, Han X, Ou D, Liu T, Li Z, Jiang G, Liu J, Zhang J. Targeting PI3K/AKT/mTOR-mediated autophagy for tumor therapy. Appl Microbiol Biotechnol 2019; 104:575-587. [PMID: 31832711 DOI: 10.1007/s00253-019-10257-8] [Citation(s) in RCA: 348] [Impact Index Per Article: 69.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/05/2019] [Accepted: 11/12/2019] [Indexed: 12/11/2022]
Abstract
Autophagy is a highly conserved catabolic process and participates in a variety of cellular biological activities. The phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway, as a critical regulator of autophagy, is involved in the initiation and promotion of a series of pathological disorders including various tumors. Autophagy also participates in regulating the balance between the tumor and the tumor microenvironment. Natural products have been considered a treasure of new drug discoveries and are of great value to medicine. Mounting evidence has suggested that numerous natural products are targeting PI3K/AKT/mTOR-mediated autophagy, thereby suppressing tumor growth. Furthermore, autophagy plays a "double-edged sword" role in different tumors. Targeting PI3K/AKT/mTOR-mediated autophagy is an important therapeutic strategy for a variety of tumors, and plays important roles in enhancing the chemosensitivity of tumor cells and avoiding drug resistance. Therefore, we summarized the roles of PI3K/AKT/mTOR-mediated autophagy in tumorigenesis, progression, and drug resistance of tumors, which may be utilized to design preferably therapeutic strategies for various tumors.
Collapse
Affiliation(s)
- Zhenru Xu
- Department of Rheumatology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Xu Han
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Daming Ou
- Department of Rheumatology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Ting Liu
- Department of Rheumatology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Zunxiong Li
- University of South China, Hengyang, Hunan, China
| | - Guanmin Jiang
- Department of Clinical Laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Jing Liu
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.
| | - Ji Zhang
- Department of Rheumatology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China.
| |
Collapse
|
25
|
28-Hydroxy-3-oxoolean-12-en-29-oic Acid, a Triterpene Acid from Celastrus Orbiculatus Extract, Inhibits the Migration and Invasion of Human Gastric Cancer Cells In Vitro. Molecules 2019; 24:molecules24193513. [PMID: 31569766 PMCID: PMC6803947 DOI: 10.3390/molecules24193513] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/25/2019] [Accepted: 09/25/2019] [Indexed: 12/29/2022] Open
Abstract
Gastric cancer is the fifth most common tumor and has the third-highest mortality rate among various malignant tumors, and the survival rate of patients is low. Celastrus orbiculatus extract has been shown to inhibit the activity of a variety of tumors. This study explored the inhibitory effect of the oleanane-type triterpenoid acid 28-hydroxy-3-oxoolean-12-en-29-oic acid molecule from Celastrus orbiculatus extract on gastric cancer cell invasion and metastasis and determined its mechanism. 28-Hydroxy-3-oxoolean-12-en-29-oic acid was first diluted to various concentrations and then used to treat SGC-7901 and BGC-823 cells. Cell proliferation was assessed by an MTT (thiazole blue) assay. Transwell and wound healing assays were used to assess cell invasion and migration. High-content imaging technology was used to further observe the effects of the drug on cell invasion and migration. Western blotting was used to assess the effects on the expression of matrix metalloproteinases (MMPs) and the effects on epithelial-mesenchymal transition (EMT)-related proteins and phosphorylation-related proteins. We found that 28-Hydroxy-3-oxoolean-12-en-29-oic acid inhibited the migration and invasion of SGC-7901 and BGC-823 gastric cancer cells in a dose-dependent manner. Consequently, 28-hydroxy-3-oxoolean-12-en-29-oic acid decreased the expression of EMT-related proteins and MMPs in gastric cancer cells and reduced protein phosphorylation, inhibiting the migration and invasion of gastric cancer cells.
Collapse
|
26
|
Shen Y, Chen BL, Zhang QX, Zheng YZ, Fu Q. Traditional uses, secondary metabolites, and pharmacology of Celastrus species - a review. JOURNAL OF ETHNOPHARMACOLOGY 2019; 241:111934. [PMID: 31129308 DOI: 10.1016/j.jep.2019.111934] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 05/02/2019] [Accepted: 05/02/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Plants of genus Celastrus (Celastraceae) have been widely used in traditional Chinese medicine (TCM) and Indian medicine to treat cognitive dysfunction, epilepsy, insomnia, rheumatism, gout, and dyspepsia for thousands of years. AIM OF STUDY We critically summarized the current evidence on the botanic characterization and distribution, ethnopharmacology, secondary metabolites, pharmacological activities, qualitative and quantitative analysis, and toxicology of Celastrus species to provide perspectives for developing more attractive pharmaceuticals of plant origin. MATERIALS AND METHODS The relevant information on Celastrus species was gathered from worldwide accepted scientific databases via electronic search (Web of Science, SciFinder, PubMed, Elsevier, SpringerLink, Wiley Online, China Knowledge Resource Integrated, and Google Scholar). Information was also obtained from the literature and books as well as PhD and MSc dissertations. Plant names were validated by "The Plant List" (www.theplantlist.org). RESULTS Comprehensive analysis of the above mentioned databases and other sources confirmed that ethnomedical uses of plants of Celastrus genus had been recorded in China, India, and other countries in Southern Asia. The phytochemical investigation revealed the presence of β-dihydroagarofuranoids, diterpenoids, triterpenoids, tetraterpenes, phenylpropanoids, alkaloids, flavonoids, lignans, and others. The crude extracts and isolated constituents have exhibited a wide range of in vitro and in vivo pharmacological effects, including antitumor, cytotoxic, insecticidal, antimicrobial, anti-rheumatoid arthritis (RA), anti-inflammatory, anti-ageing and antioxidative, and neuroprotective activities. CONCLUSION Plants of genus Celastrus have been confirmed to show a strong potential for therapeutic and health-maintaining effects, in light of their long traditional use and the phytochemical and pharmacological studies summarized here. Currently, pharmacological studies of this genus mainly focus on Celastrus paniculatus Willd. and Celastrus orbiculatus Thunb. Therefore, more pharmacological investigations should be implemented to support traditional uses of other medicinal plants of the genus Celastrus. Moreover, studies on the toxicity, bioavailability, and pharmacokinetics, in addition to clinical trials, are indispensable for assessing the safety and efficacy of the secondary metabolites or extracts obtained from plants belonging to this genus.
Collapse
Affiliation(s)
- Yue Shen
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture Rural Affairs, College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Bi-Lian Chen
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture Rural Affairs, College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Qin-Xiu Zhang
- School of Medical and Life Sciences, Chengdu University of TCM, Chengdu, 610072, China
| | - Yu-Zhong Zheng
- School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou, 521041, China
| | - Qiang Fu
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture Rural Affairs, College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, 610106, China.
| |
Collapse
|
27
|
Narayanankutty A. PI3K/ Akt/ mTOR Pathway as a Therapeutic Target for Colorectal Cancer: A Review of Preclinical and Clinical Evidence. Curr Drug Targets 2019; 20:1217-1226. [DOI: 10.2174/1389450120666190618123846] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 05/21/2019] [Accepted: 05/29/2019] [Indexed: 02/06/2023]
Abstract
Background:
Phosphoinositide 3-kinase (PI3Ks) is a member of intracellular lipid kinases
and involved in the regulation of cellular proliferation, differentiation and survival. Overexpression of
the PI3K/Akt/mTOR signalling has been reported in various forms of cancers, especially in colorectal
cancers (CRC). Due to their significant roles in the initiation and progression events of colorectal cancer,
they are recognized as a striking therapeutic target.
Objective:
The present review is aimed to provide a detailed outline on the role of PI3K/Akt/mTOR
pathway in the initiation and progression events of colorectal cancers as well as its function in drug
resistance. Further, the role of PI3K/Akt/mTOR inhibitors alone and in combination with other chemotherapeutic
drugs, in alleviating colorectal cancer is also discussed. The review contains preclinical
and clinical evidence as well as patent literature of the pathway inhibitors which are natural
and synthetic in origin.
Methods:
The data were obtained from PubMed/Medline databases, Scopus and Google patent literature.
Results:
PI3K/Akt/mTOR signalling is an important event in colorectal carcinogenesis. In addition, it
plays significant roles in acquiring drug resistance as well as metastatic initiation events of CRCs.
Several small molecules of natural and synthetic origin have been found to be potent inhibitors of
CRCs by effectively downregulating the pathway. Data from various clinical studies also support
these pathway inhibitors and several among them are patented.
Conclusion:
Inhibitors of the PI3K/mTOR pathway have been successful for the treatment of primary
and metastatic colorectal cancers, rendering the pathway as a promising clinical cancer therapeutic target.
Collapse
Affiliation(s)
- Arunaksharan Narayanankutty
- Post Graduate & Research Department of Zoologyid1, St. Joseph's College (Autonomous), Devagiri, Calicut, Kerala, 673008, India
| |
Collapse
|
28
|
Zhang R, Yu Q, Lu W, Shen J, Zhou D, Wang Y, Gao S, Wang Z. Grape seed procyanidin B2 promotes the autophagy and apoptosis in colorectal cancer cells via regulating PI3K/Akt signaling pathway. Onco Targets Ther 2019; 12:4109-4118. [PMID: 31213831 PMCID: PMC6538883 DOI: 10.2147/ott.s195615] [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] [Received: 11/22/2018] [Accepted: 03/07/2019] [Indexed: 12/17/2022] Open
Abstract
Aim: Colorectal cancer (CRC) is a major malignancy in China, which is the critical risk of people health. Many natural herbs extracts have been found to exhibit good therapeutic effect on CRC. Our previous study found that grape seed procyanidins B2 (PB2) would induce CRC cell death. However, the molecular mechanism underlying its anti-tumor effect on CRC remains unclear. Thereby, this study aimed to investigate the anti-tumor mechanism of PB2 on CRC. Methods: CCK-8, western blotting, flow cytometry, qRT-PCR and animal study were used in the current study. Results: The in vitro and in vivo data demonstrated that PB2 could promote the apoptosis of CRC cells in a dose-dependent manner, which was significantly reversed by caspase 3 inhibitor. Meanwhile, PB2 dose-dependently induced autophagy in CRC cells, which was markedly attenuated by autophagy inhibitor 3-MA. In addition, PB2 dose-dependently inhibited the expressions of p-PI3K, p-Akt and p-mTOR in the cells. Conclusion: PB2 dose-dependently induced apoptosis and autophagy in CRC cells via downregulation of PI3K/Akt pathway. This study provided the experimental basis for further development of PB2 as a new effective anticancer drug for the patients with CRC.
Collapse
Affiliation(s)
- Ruijuan Zhang
- Department of TCM, Shanghai Putuo District People's Hospital, Shanghai 200060, People's Republic of China
| | - Qianyun Yu
- Department of TCM, Shanghai Huangpu District Wuliqiao Community Health Center, Shanghai, 200023, People's Republic of China
| | - Wenqiang Lu
- Department of TCM, Shanghai Putuo District People's Hospital, Shanghai 200060, People's Republic of China
| | - Jun Shen
- Department of TCM, Shanghai Putuo District People's Hospital, Shanghai 200060, People's Republic of China
| | - Dongqing Zhou
- Department of TCM, Shanghai Putuo District People's Hospital, Shanghai 200060, People's Republic of China
| | - Yingjue Wang
- Department of TCM, Shanghai Putuo District People's Hospital, Shanghai 200060, People's Republic of China
| | - Shurong Gao
- Department of TCM, Shanghai Putuo District People's Hospital, Shanghai 200060, People's Republic of China
| | - Zhijun Wang
- Department of TCM, Shanghai Putuo District People's Hospital, Shanghai 200060, People's Republic of China
| |
Collapse
|
29
|
Wan Z, Chai R, Yuan H, Chen B, Dong Q, Zheng B, Mou X, Pan W, Tu Y, Yang Q, Tu S, Hu X. MEIS2 promotes cell migration and invasion in colorectal cancer. Oncol Rep 2019; 42:213-223. [PMID: 31115559 PMCID: PMC6549210 DOI: 10.3892/or.2019.7161] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 03/18/2019] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common types of malignancy worldwide. Distant metastasis is a key cause of CRC-associated mortality. MEIS2 has been identified to be dysregulated in several types of human cancer. However, the mechanisms underlying the regulatory role of MEIS2 in CRC metastasis remain largely unknown. For the first time, the present study demonstrated that MEIS2 serves a role as a promoter of metastasis in CRC. In vivo and in vitro experiments revealed that knockdown of MEIS2 significantly suppressed CRC migration, invasion and the epithelial-mesenchymal transition. Furthermore, microarray and bioinformatics analyses were performed to investigate the underlying mechanisms of MEIS2 in the regulation of CRC metastasis. Additionally, it was identified that a high expression of MEIS2 was significantly associated with a shorter overall survival time for patients with CRC. The present study demonstrated that MEIS2 may serve as a novel biomarker for CRC.
Collapse
Affiliation(s)
- Ziang Wan
- Department of Colorectal Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Rui Chai
- Department of Colorectal Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Hang Yuan
- Department of Colorectal Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Bingchen Chen
- Department of Colorectal Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Quanjin Dong
- Department of Colorectal Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Boan Zheng
- Department of Colorectal Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Xiaozhou Mou
- Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Wensheng Pan
- Department of Gastroenterology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Yifeng Tu
- Department of Pathology, College of Basic Medical Sciences, Shenyang Medical College, Shenyang, Liaoning 110034, P.R. China
| | - Qing Yang
- Department of Academy of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Shiliang Tu
- Department of Colorectal Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Xinye Hu
- Department of Colorectal Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
30
|
Tang Y, Ren F, Cong X, Kong Y, Tian Y, Xu Y, Fan J. Overexpression of ribonuclease inhibitor induces autophagy in human colorectal cancer cells via the Akt/mTOR/ULK1 pathway. Mol Med Rep 2019; 19:3519-3526. [PMID: 30896869 PMCID: PMC6472130 DOI: 10.3892/mmr.2019.10030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 03/06/2019] [Indexed: 12/18/2022] Open
Abstract
Ribonuclease inhibitor (RI), also termed angiogenin inhibitor, acts as the inhibitor of ribonucleolytic activity of RNase A and angiogenin. The expression of RI has been investigated in melanoma and bladder cancer cells. However, the precise role of RI in tumorigenesis, in addition to RI‑induced autophagy, remains poorly understood. In the present study, it was demonstrated that RI positively regulated autophagy in human colorectal cancer (CRC) cells as indicated by an increase in light chain 3 (LC3)‑II levels. Furthermore, RI regulated cell survival in HT29 cells. In addition, autophagy‑associated proteins, including beclin‑1 and autophagy‑related protein 13, were increased in response to RI‑induced autophagy, and the protein kinase B (Akt)/mechanistic target of rapamycin (mTOR)/Unc‑51 like autophagy activating kinase (ULK1) pathway may be involved in the activation of autophagy induced by RI overexpression. Taken together, the evidence of the present study indicated that the overexpression of RI induced ATG‑dependent autophagy in CRC cells via the Akt/mTOR/ULK1 pathway, suggesting that the upregulation of RI activity may constitute a novel approach for the treatment of human colorectal carcinoma.
Collapse
Affiliation(s)
- Ying Tang
- Department of Pathology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Feng Ren
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Xi Cong
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Ying Kong
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Yuxiang Tian
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Yuefei Xu
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Jianhui Fan
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| |
Collapse
|
31
|
Zhu G, Wang HB, Jin F, Tao LD, Li D, Ni TY, Li WY, Pan B, Xiao WM, Ding YB, Sunagawa M, Liu YQ. Celastrus Orbiculatus Extract Suppresses Migration and Invasion
of Gastric Cancer by Inhibiting Prohibitin and c-Raf/ERK Signaling
Pathway. INT J PHARMACOL 2018. [DOI: 10.3923/ijp.2019.40.49] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
32
|
Zheng Y, Tan K, Huang H. Long noncoding RNA HAGLROS regulates apoptosis and autophagy in colorectal cancer cells via sponging miR-100 to target ATG5 expression. J Cell Biochem 2018; 120:3922-3933. [PMID: 30430634 DOI: 10.1002/jcb.27676] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/21/2018] [Indexed: 12/11/2022]
Abstract
The aim of this study was to explore the relationship between the expression of HOXD antisense growth-associated long noncoding RNA (HAGLROS) and prognosis of patients with colorectal cancer (CRC), as well as the roles and regulatory mechanism of HAGLROS in CRC development. The HAGLROS expression in CRC tissues and cells was detected. The correlation between HAGLROS expression and survival time of CRC patients was investigated. Moreover, HAGLROS was overexpressed and suppressed in HCT-116 cells, followed by detection of cell viability, apoptosis, and the expression of apoptosis-related proteins and autophagy markers. Furthermore, the association between HAGLROS and miR-100 and the potential targets of miR-100 were investigated. Besides, the regulatory relationship between HAGLROS and PI3K/AKT/mTOR pathway was elucidated. The results showed that HAGLROS was highly expressed in CRC tissues and cells. Highly expression of HAGLROS correlated with a shorter survival time of CRC patients. Moreover, knockdown of HAGLROS in HCT-116 cells induced apoptosis by increasing the expression of Bax/Bcl-2 ratio, cleaved-caspase-3, and cleaved-caspase-9, and inhibited autophagy by decreasing the expression of LC3II/LC3I and Beclin-1 and increasing P62 expression. Furthermore, HAGLROS negatively regulated the expression of miR-100, and HAGLROS controlled HCT-116 cell apoptosis and autophagy through negatively regulation of miR-100. Autophagy related 5 (ATG5) was verified as a functional target of miR-100 and miR-100 regulated HCT-116 cell apoptosis and autophagy through targeting ATG5. Besides, HAGLROS overexpression activated phosphatidylinositol-3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) pathway. In conclusion, a highly expression of HAGLROS correlated with shorter survival time of CRC patients. Downregulation of HAGLROS may induce apoptosis and inhibit autophagy in CRC cells by regulation of miR-100/ATG5 axis and PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Yansheng Zheng
- Department of Gastrointestinal Surgery, Traditional Chinese Medicine Hospital of Guangdong Province, Guangzhou, China
| | - Kanglian Tan
- Department of Gastrointestinal Surgery, Traditional Chinese Medicine Hospital of Guangdong Province, Guangzhou, China
| | - Haipeng Huang
- Department of Gastrointestinal Surgery, Traditional Chinese Medicine Hospital of Guangdong Province, Guangzhou, China
| |
Collapse
|
33
|
Nitulescu GM, Van De Venter M, Nitulescu G, Ungurianu A, Juzenas P, Peng Q, Olaru OT, Grădinaru D, Tsatsakis A, Tsoukalas D, Spandidos DA, Margina D. The Akt pathway in oncology therapy and beyond (Review). Int J Oncol 2018; 53:2319-2331. [PMID: 30334567 PMCID: PMC6203150 DOI: 10.3892/ijo.2018.4597] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/10/2018] [Indexed: 02/07/2023] Open
Abstract
Protein kinase B (Akt), similar to many other protein kinases, is at the crossroads of cell death and survival, playing a pivotal role in multiple interconnected cell signaling mechanisms implicated in cell metabolism, growth and division, apoptosis suppression and angiogenesis. Akt protein kinase displays important metabolic effects, among which are glucose uptake in muscle and fat cells or the suppression of neuronal cell death. Disruptions in the Akt-regulated pathways are associated with cancer, diabetes, cardiovascular and neurological diseases. The regulation of the Akt signaling pathway renders Akt a valuable therapeutic target. The discovery process of Akt inhibitors using various strategies has led to the identification of inhibitors with great selectivity, low side-effects and toxicity. The usefulness of Akt emerges beyond cancer therapy and extends to other major diseases, such as diabetes, heart diseases, or neurodegeneration. This review presents key features of Akt structure and functions, and presents the progress of Akt inhibitors in regards to drug development, and their preclinical and clinical activity in regards to therapeutic efficacy and safety for patients.
Collapse
Affiliation(s)
- George Mihai Nitulescu
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Maryna Van De Venter
- Department of Biochemistry and Microbiology, Nelson Mandela University, Port Elizabeth 6031, South Africa
| | - Georgiana Nitulescu
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Anca Ungurianu
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Petras Juzenas
- Department of Pathology, Radiumhospitalet, Oslo University Hospital, 0379 Oslo, Norway
| | - Qian Peng
- Department of Pathology, Radiumhospitalet, Oslo University Hospital, 0379 Oslo, Norway
| | - Octavian Tudorel Olaru
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Daniela Grădinaru
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Aristides Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Dimitris Tsoukalas
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Denisa Margina
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| |
Collapse
|
34
|
The Ethyl Acetate Extract of Gynura formosana Kitam. Leaves Inhibited Cervical Cancer Cell Proliferation via Induction of Autophagy. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4780612. [PMID: 29992145 PMCID: PMC5994325 DOI: 10.1155/2018/4780612] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/16/2018] [Accepted: 04/02/2018] [Indexed: 11/22/2022]
Abstract
Gynura formosana Kitam. belongs to the Compositae family and has been traditionally used for the prevention of cancer, diabetes, and inflammation in China. Previous studies had indicated that the ethyl acetate extract of Gynura formosana Kitam. leaves (EAEG) exhibited antioxidant and anti-inflammatory activity. In this report, we demonstrated that EAEG possessed potent anticancer activity through autophagy-mediated inhibition of cell proliferation. EAEG induced a strong cytostatic effect towards HeLa cells and, to a lesser extent, HepG2 and MCF-7 cells. This cytostatic effect of EAEG was not a consequence of increased apoptosis, as neither DNA fragmentation nor change in protein expression level for a number of apoptosis-related genes including Bid, Bax, Bcl-2, and caspase-3 was observed after EAEG treatment, and the apoptosis inhibitor Z-VAD-FMK did not inhibit the EAEG-elicited cytostatic effect. On the other hand, EAEG induced autophagy in a dose-dependent fashion, as shown by increased GFP puncta formation, enhanced conversion of the microtubule-associated protein light chain LC3-I to LC3-II, and downregulation of the p62 protein. Treating the HeLa cells with EAEG together with Chloroquine (CQ) further accelerated LC3 conversion and p62 clearance, indicating that EAEG induced complete autophagy flux. Importantly, the autophagy inhibitor 3-methyladenine (3MA) significantly abrogated the cytostatic effect of EAEG, strongly suggesting that EAEG inhibited HeLa cell proliferation through the induction of autophagy rather than apoptosis. Our results provided a novel and interesting mechanistic insight into the anticancer action of EAEG, supporting the traditional use of this plant for the treatment of the cancer.
Collapse
|
35
|
Vengoji R, Macha MA, Batra SK, Shonka NA. Natural products: a hope for glioblastoma patients. Oncotarget 2018; 9:22194-22219. [PMID: 29774132 PMCID: PMC5955138 DOI: 10.18632/oncotarget.25175] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 03/28/2018] [Indexed: 12/27/2022] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive malignant tumors with an overall dismal survival averaging one year despite multimodality therapeutic interventions including surgery, radiotherapy and concomitant and adjuvant chemotherapy. Few drugs are FDA approved for GBM, and the addition of temozolomide (TMZ) to standard therapy increases the median survival by only 2.5 months. Targeted therapy appeared promising in in vitro monolayer cultures, but disappointed in preclinical and clinical trials, partly due to the poor penetration of drugs through the blood brain barrier (BBB). Cancer stem cells (CSCs) have intrinsic resistance to initial chemoradiation therapy (CRT) and acquire further resistance via deregulation of many signaling pathways. Due to the failure of classical chemotherapies and targeted drugs, research efforts focusing on the use of less toxic agents have increased. Interestingly, multiple natural compounds have shown antitumor and apoptotic effects in TMZ resistant and p53 mutant GBM cell lines and also displayed synergistic effects with TMZ. In this review, we have summarized the current literature on natural products or product analogs used to modulate the BBB permeability, induce cell death, eradicate CSCs and sensitize GBM to CRT.
Collapse
Affiliation(s)
- Raghupathy Vengoji
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Muzafar A. Macha
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Otolaryngology/Head and Neck Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Eppley Institute for Research in Cancer and Allied Diseases and Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Nicole A. Shonka
- Eppley Institute for Research in Cancer and Allied Diseases and Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Internal Medicine, Division of Oncology and Hematology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| |
Collapse
|
36
|
Wang X, Liu Y, Liu W, Zhang Y, Guo F, Zhang L, Cui M, Liu S, Wu R. Ubenimex, an APN inhibitor, could serve as an anti‑tumor drug in RT112 and 5637 cells by operating in an Akt‑associated manner. Mol Med Rep 2018; 17:4531-4539. [PMID: 29328441 PMCID: PMC5802231 DOI: 10.3892/mmr.2018.8402] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 12/29/2017] [Indexed: 02/07/2023] Open
Abstract
Bladder cancer, a common urinary tract tumor, has high mortality and recurrence rates associated with metastasis. Aminopeptidase N (APN) expression and metastasis have been indicated to be associated with one another. Ubenimex may function as an APN inhibitor to inhibit the degradation of the extracellular matrix during tumorigenesis. Furthermore, APN has been widely used as an adjuvant therapy for the treatment of tumors; however, little information is available regarding the impact of ubenimex on patients. Autophagy is suggested to be important in the transformation and progression of cancer. Additionally, apoptosis, which leads to the rapid demolition of cellular organelles and structures, has also been suggested as an important factor. Thus, the present study investigated the role of ubenimex in inhibiting migration and invasion by downregulating APN expression levels to induce autophagic cell death and apoptosis in bladder cancer cells. RT112 and 5637 cell lines were treated with varying doses of ubenimex. Cell viability was measured by CCK8 colorimetry and flow cytometry. Using fluorescence microscopy, autophagic cell death was assessed using acridine orange/ethidium bromide staining. Furthermore, apoptotic cell death was assessed using flow cytometry and Trypan blue staining was used to evaluate the cell death rate. Protein expression was determined by western blot analysis. Matrigel invasion assays were exploited to assess the invasion capabilities of 5637 cells. Wound-healing migration assays and Matrigel migration assays were exploited to assess the migratory abilities of 5637 cells. Treatment with ubenimex was accompanied by decreased Akt expression, indicating that ubenimex may have similar functions to Akt inhibitors. Results also indicated that ubenimex inhibited cell migration and invasion in bladder cancer cells. Furthermore, ubenimex also induced autophagic cell death and apoptosis, which suggested that mixed programmed cell death occurred in ubenimex-treated bladder cancer cells. The results from the present study suggest that ubenimex may be a potential adjuvant therapy for the treatment of bladder cancer.
Collapse
Affiliation(s)
- Xiaoqing Wang
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Yang Liu
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Wei Liu
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Yongfei Zhang
- Department of Dermatology, Shandong Provincial Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China
| | - Feng Guo
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Lijuan Zhang
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Mingyu Cui
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Shuai Liu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Rongde Wu
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
37
|
Han L, Zhang Y, Liu S, Zhao Q, Liang X, Ma Z, Gupta PK, Zhao M, Wang A. Autophagy flux inhibition, G2/M cell cycle arrest and apoptosis induction by ubenimex in glioma cell lines. Oncotarget 2017; 8:107730-107743. [PMID: 29296201 PMCID: PMC5746103 DOI: 10.18632/oncotarget.22594] [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: 08/24/2017] [Accepted: 11/03/2017] [Indexed: 12/21/2022] Open
Abstract
This study aimed to investigate whether ubenimex could work as an anti-tumor drug alone in glioma cells and figure out the underlying potential mechanisms. Ubenimex is widely used as an adjunct therapy in multiple solid cancers. However, it is rarely used to treat glioblastoma. The function of ubenimex in enhancing JQ1 treatment sensitivity of glioma cells by blocking autophagic degradation of HEXIM1 was previously studied. However, the detailed mechanism of autophagy regulation by ubenimex remains unclear. The U87 and U251 cell lines were treated with different doses of ubenimex. Cell viability was measured by using the WST-8 assay. Cell death was assessed using trypan blue staining and flow cytometry. The migration and invasive ability of glioma cells were examined by transwell migration/invasion assay. LC3-GFP-RFP was used to measure autophagic flux. Protein expression was assessed by Western blot analysis. Autophagosomes were evaluated using the transmission electron microscopy. Moreover, cell cycle arrest (PI Staining) was measured by flow cytometry. Results revealed that ubenimex inhibited cell proliferation as well as migration/invasion in glioma cells. Besides, ubenimex increased glioma cell death via autophagic flux inhibition. Meanwhile, ubenimex induced G2/M phase arrest and apoptosis, and this effect was accompanied by the decreased levels of p-Akt, indicating the role of ubenimex in the regulation of glioma cell proliferation and metastasis. To sum up, this study concluded that ubenimex could work as an anti-tumor drug alone in the glioma cells via inhibiting autophagic flux and inducing G2/M arrest as well as apoptosis.
Collapse
Affiliation(s)
- Liping Han
- Department of Neurology, Qianfoshan Hospital Affiliated to Shandong University, Jinan, P.R. China.,Department of Neurology, Shandong Police Hospital, Jinan, P.R. China
| | - Yongfei Zhang
- Department of Dermatology, Qianfoshan Hospital Affiliated to Shandong University, Jinan, P.R. China
| | - Shuai Liu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, P.R. China
| | - Qingwei Zhao
- Department of Neurology, Shandong Police Hospital, Jinan, P.R. China
| | - Xianhong Liang
- Department of Neurology, Shandong Police Hospital, Jinan, P.R. China
| | - Zhiguo Ma
- Department of Neurology, Shandong Police Hospital, Jinan, P.R. China
| | | | - Miaoqing Zhao
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, P.R. China
| | - Aihua Wang
- Department of Neurology, Qianfoshan Hospital Affiliated to Shandong University, Jinan, P.R. China
| |
Collapse
|
38
|
Guo F, Lin SC, Zhao MS, Yu B, Li XY, Gao Q, Lin DJ. microRNA-142-3p inhibits apoptosis and inflammation induced by bleomycin through down-regulation of Cox-2 in MLE-12 cells. ACTA ACUST UNITED AC 2017; 50:e5974. [PMID: 28678919 PMCID: PMC5496156 DOI: 10.1590/1414-431x20175974] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 04/27/2017] [Indexed: 11/21/2022]
Abstract
microRNA (miR)-142-3p is implicated in malignancy and has been identified as a biomarker for aggressive and recurrent lung adenocarcinomas. This study aimed to evaluate the inhibitory effect of miR-142-3p on apoptosis and inflammation induced by bleomycin in MLE-12 cells. MLE-12 cells were first transfected either with miR-142-3p mimic or miR-142-3p inhibitor and then the cells were exposed to 50 μg/mL of bleomycin. Thereafter, cell viability, apoptosis and the expression of pro-inflammatory cytokines were assessed using CCK-8, flow cytometry, RT-PCR and western blot analyses. Cox-2, PI3K, AKT and mTOR expressions were detected by western blotting after bleomycin was administered together with NS-398 (an inhibitor of Cox-2). As a result, cell viability was significantly decreased, as well as apoptosis and the expression of IL-1 and TNF-α were remarkably increased after 50 and 100 μg/mL of bleomycin administration. miR-142-3p overexpression alleviated bleomycin-induced apoptosis and overproduction of these two pro-inflammatory cytokines, while miR-142-3p suppression exhibited completely opposite results. Up-regulation of Cox-2 and inactivation of PI3K/AKT/mTOR were found in bleomycin-pretreated cells, while these abnormal regulations were partially abolished by miR-142-3p overexpression and NS-398. In conclusion, this study demonstrated that miR-142-3p overexpression protected bleomycin-induced injury in lung epithelial MLE-12 cells, possibly via regulating Cox-2 expression and PI3K/AKT/mTOR signaling pathway. These findings provide evidence that miR-142-3p may be a therapeutic strategy for idiopathic pulmonary fibrosis (IPF) treatment.
Collapse
Affiliation(s)
- F Guo
- Department of Respiratory Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China.,Department of Respiratory Medicine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - S C Lin
- Department of Respiratory Medicine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - M S Zhao
- Department of Respiratory Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - B Yu
- Department of Respiratory Medicine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - X Y Li
- Department of Respiratory Medicine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Q Gao
- Department of Respiratory Medicine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - D J Lin
- Department of Respiratory Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| |
Collapse
|
39
|
Qian HR, Shi ZQ, Zhu HP, Gu LH, Wang XF, Yang Y. Interplay between apoptosis and autophagy in colorectal cancer. Oncotarget 2017; 8:62759-62768. [PMID: 28977986 PMCID: PMC5617546 DOI: 10.18632/oncotarget.18663] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 05/15/2017] [Indexed: 12/15/2022] Open
Abstract
Autophagy and apoptosis are two pivotal mechanisms in mediating cell survival and death. Cross-talk of autophagy and apoptosis has been documented in the tumorigenesis and progression of cancer, while the interplay between the two pathways in colorectal cancer (CRC) has not yet been comprehensively summarized. In this study, we outlined the basis of apoptosis and autophagy machinery firstly, and then reviewed the recent evidence in cellular settings or animal studies regarding the interplay between them in CRC. In addition, several key factors that modulate the cross-talk between autophagy and apoptosis as well as its significance in clinical practice were discussed. Understanding of the interplay between the cell death mechanisms may benefit the translation of CRC treatment from basic research to clinical use.
Collapse
Affiliation(s)
- Hao-Ran Qian
- Department of General Surgery, Institute of Minimally Invasive, Surgery of Zhejiang University, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang, PR China
| | - Zhao-Qi Shi
- Department of General Surgery, Institute of Minimally Invasive, Surgery of Zhejiang University, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang, PR China
| | - He-Pan Zhu
- Department of General Surgery, Institute of Minimally Invasive, Surgery of Zhejiang University, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang, PR China
| | - Li-Hu Gu
- Department of General Surgery, Institute of Minimally Invasive, Surgery of Zhejiang University, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang, PR China
| | - Xian-Fa Wang
- Department of General Surgery, Institute of Minimally Invasive, Surgery of Zhejiang University, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang, PR China
| | - Yi Yang
- Department of Pharmacology, Hangzhou Key Laboratory of Medical Neurobiology, School of Medicine, Hangzhou Normal University, Hangzhou 310036, Zhejiang, PR China
| |
Collapse
|