1
|
Wang F, Liang L, Yu M, Wang W, Badar IH, Bao Y, Zhu K, Li Y, Shafi S, Li D, Diao Y, Efferth T, Xue Z, Hua X. Advances in antitumor activity and mechanism of natural steroidal saponins: A review of advances, challenges, and future prospects. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155432. [PMID: 38518645 DOI: 10.1016/j.phymed.2024.155432] [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: 08/12/2023] [Revised: 01/11/2024] [Accepted: 02/06/2024] [Indexed: 03/24/2024]
Abstract
BACKGROUND Cancer, the second leading cause of death worldwide following cardiovascular diseases, presents a formidable challenge in clinical settings due to the extensive toxic side effects associated with primary chemotherapy drugs employed for cancer treatment. Furthermore, the emergence of drug resistance against specific chemotherapeutic agents has further complicated the situation. Consequently, there exists an urgent imperative to investigate novel anticancer drugs. Steroidal saponins, a class of natural compounds, have demonstrated notable antitumor efficacy. Nonetheless, their translation into clinical applications has remained unrealized thus far. In light of this, we conducted a comprehensive systematic review elucidating the antitumor activity, underlying mechanisms, and inherent limitations of steroidal saponins. Additionally, we propose a series of strategic approaches and recommendations to augment the antitumor potential of steroidal saponin compounds, thereby offering prospective insights for their eventual clinical implementation. PURPOSE This review summarizes steroidal saponins' antitumor activity, mechanisms, and limitations. METHODS The data included in this review are sourced from authoritative databases such as PubMed, Web of Science, ScienceDirect, and others. RESULTS A comprehensive summary of over 40 steroidal saponin compounds with proven antitumor activity, including their applicable tumor types and structural characteristics, has been compiled. These steroidal saponins can be primarily classified into five categories: spirostanol, isospirostanol, furostanol, steroidal alkaloids, and cholestanol. The isospirostanol and cholestanol saponins are found to have more potent antitumor activity. The primary antitumor mechanisms of these saponins include tumor cell apoptosis, autophagy induction, inhibition of tumor migration, overcoming drug resistance, and cell cycle arrest. However, steroidal saponins have limitations, such as higher cytotoxicity and lower bioavailability. Furthermore, strategies to address these drawbacks have been proposed. CONCLUSION In summary, isospirostanol and cholestanol steroidal saponins demonstrate notable antitumor activity and different structural categories of steroidal saponins exhibit variations in their antitumor signaling pathways. However, the clinical application of steroidal saponins in cancer treatment still faces limitations, and further research and development are necessary to advance their potential in tumor therapy.
Collapse
Affiliation(s)
- Fengge Wang
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Lu Liang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR, PR China
| | - Ma Yu
- School of Life Science and Engineering, Southwest University of Science and Technology, 59 Qinglong Road, Mianyang, 621010, Sichuan, PR China
| | - Wenjie Wang
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Iftikhar Hussain Badar
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang, 150030, PR China; Department of Meat Science and Technology, University of Veterinary and Animal Sciences, Lahore, 54000, Pakistan
| | - Yongping Bao
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, United Kingdom
| | - Kai Zhu
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Yanlin Li
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Saba Shafi
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Dangdang Li
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Yongchao Diao
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz 55128, Germany.
| | - Zheyong Xue
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China.
| | - Xin Hua
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China.
| |
Collapse
|
2
|
Ou J, Wang Z, Huang H, Chen J, Liu X, Jia X, Song B, Cheong KL, Gao Y, Zhong S. Intervention effects of sulfate glycosaminoglycan from swim bladder against arsenic-induced damage in IEC-6 cells. Int J Biol Macromol 2023; 252:126460. [PMID: 37619679 DOI: 10.1016/j.ijbiomac.2023.126460] [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: 04/15/2023] [Revised: 07/26/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
In this study, a purified macromolecular sulfate glycosaminoglycan whose structural characterization is similar to chondroitin sulfate from the swim bladder of Aristichthys nobilis, named SBSG, was used to explore the intervention effects on arsenic-induced intestinal epithelial cells (IEC-6) damage. Arsenic exposure led to cell membrane rupture, mitochondrial dysfunction, oxidative damage, and down-regulation of tight junction proteins expression. Treatment with SBSG could alleviate arsenic exposure-induced cell damage by decreasing the extracellular lactate dehydrogenase activity and influencing mitochondrial membrane potential, reactive oxygen species level, malondialdehyde content, and anti-oxidative enzyme activity. On the other hand, SBSG could promote nitric oxide production to achieve potential immunoregulation. The Western blot showed that intervention of SBSG mainly could restrain the activation of the JNK signaling pathway and up-regulate the expression of ZO-1 against arsenic-induced cell damage. This study provides a new perspective for understanding the heavy metal detoxification of SBSG on the intestinal and indicates that SBSG could be used as natural antioxidant resistant to heavy metal toxicity.
Collapse
Affiliation(s)
- Jieying Ou
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Zhanjiang 524088, China
| | - Zhuo Wang
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Zhanjiang 524088, China.
| | - Houpei Huang
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Zhanjiang 524088, China
| | - Jing Chen
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Zhanjiang 524088, China
| | - Xiaofei Liu
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Zhanjiang 524088, China
| | - Xuejing Jia
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Zhanjiang 524088, China
| | - Bingbing Song
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Zhanjiang 524088, China
| | - Kit-Leong Cheong
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Zhanjiang 524088, China
| | - Yuan Gao
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Zhanjiang 524088, China
| | - Saiyi Zhong
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Zhanjiang 524088, China; Shenzhen Research Institute, Guangdong Ocean University, Shenzhen 518108, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China.
| |
Collapse
|
3
|
Yang L, Liu YN, Gu Y, Guo Q. Deltonin enhances gastric carcinoma cell apoptosis and chemosensitivity to cisplatin via inhibiting PI3K/AKT/mTOR and MAPK signaling. World J Gastrointest Oncol 2023; 15:1739-1755. [PMID: 37969408 PMCID: PMC10631430 DOI: 10.4251/wjgo.v15.i10.1739] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/23/2023] [Accepted: 07/19/2023] [Indexed: 10/10/2023] Open
Abstract
BACKGROUND As an active ingredient derived from Dioscorea zingiberensis C.H. Wright, deltonin has been reported to show anti-cancer effects in a variety of malignancies. AIM To investigate the role and mechanism of action of deltonin in promoting gastric carcinoma (GC) cell apoptosis and chemosensitivity to cisplatin. METHODS The GC cell lines AGS, HGC-27, and MKN-45 were treated with deltonin and then subjected to flow cytometry and 3-(4,5-dimethylthiazol-2-yl)-3,5-diphenyltetrazolium bromide assays for cell apoptosis and viability determination. Western blot analysis was conducted to examine alterations in the expression of apoptosis-related proteins (Bax, Bid, Bad, and Fas), DNA repair-associated proteins (Rad51 and MDM2), and phosphatidylinositol 3-kinase/protein kinase B/mammalian target of the rapamycin (PI3K/AKT/mTOR) and p38-mitogen-activated protein kinase (MAPK) axis proteins. Additionally, the influence of deltonin on GC cell chemosensitivity to cisplatin was evaluated both in vitro and in vivo. RESULTS Deltonin treatment weakened viability, enhanced apoptosis, and dampened DNA repair in GC cell lines in a dose-dependent pattern. Furthermore, deltonin mitigated PI3K, AKT, mTOR, and p38-MAPK phosphorylation. HS-173, an inhibitor of PI3K, attenuated GC cell viability and abolished deltonin inhibition of GC cell viability and PI3K/AKT/mTOR and p38-MAPK pathway activation. Deltonin also promoted the chemosensitivity of GC cells to cisplatin via repressing GC cell proliferation and growth and accelerating apoptosis. CONCLUSION Deltonin can boost the chemosensitivity of GC cells to cisplatin via inactivating p38-MAPK and PI3K/AKT/mTOR signaling.
Collapse
Affiliation(s)
- Lin Yang
- Intensive Care Unit, Second Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Ya-Nan Liu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Yi Gu
- Nursing Department of Obstetrics and Gynecology, Second Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Qi Guo
- Department of Radiotherapy, Second Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| |
Collapse
|
4
|
Chen Y, Luo X, Zou Z, Liang Y. The Role of Reactive Oxygen Species in Tumor Treatment and its Impact on Bone Marrow Hematopoiesis. Curr Drug Targets 2021; 21:477-498. [PMID: 31736443 DOI: 10.2174/1389450120666191021110208] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/21/2019] [Accepted: 10/08/2019] [Indexed: 02/08/2023]
Abstract
Reactive oxygen species (ROS), an important molecule inducing oxidative stress in organisms, play a key role in tumorigenesis, tumor progression and recurrence. Recent findings on ROS have shown that ROS can be used to treat cancer as they accelerate the death of tumor cells. At present, pro-oxidant drugs that are intended to increase ROS levels of the tumor cells have been widely used in the clinic. However, ROS are a double-edged sword in the treatment of tumors. High levels of ROS induce not only the death of tumor cells but also oxidative damage to normal cells, especially bone marrow hemopoietic cells, which leads to bone marrow suppression and (or) other side effects, weak efficacy of tumor treatment and even threatening patients' life. How to enhance the killing effect of ROS on tumor cells while avoiding oxidative damage to the normal cells has become an urgent issue. This study is a review of the latest progress in the role of ROS-mediated programmed death in tumor treatment and prevention and treatment of oxidative damage in bone marrow induced by ROS.
Collapse
Affiliation(s)
- Yongfeng Chen
- Taizhou University Hosipital, Taizhou University, Taizhou, 318000, Zhejiang, China.,Department of Basic Medical Sciences, Medical College of Taizhou University, Taizhou, 318000, Zhejiang, China
| | - Xingjing Luo
- Taizhou University Hosipital, Taizhou University, Taizhou, 318000, Zhejiang, China.,Department of Basic Medical Sciences, Medical College of Taizhou University, Taizhou, 318000, Zhejiang, China
| | - Zhenyou Zou
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, 541199, Guangxi, China
| | - Yong Liang
- Taizhou University Hosipital, Taizhou University, Taizhou, 318000, Zhejiang, China.,Department of Basic Medical Sciences, Medical College of Taizhou University, Taizhou, 318000, Zhejiang, China
| |
Collapse
|
5
|
Plasma-Treated Flammulina velutipes-Derived Extract Showed Anticancer Potential in Human Breast Cancer Cells. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10238395] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Natural products with medicinal properties are among alternative therapies of interest due to their high body tolerance. We aimed to determine whether nonthermal gas plasma could enhance the medicinal value of Flammulina velutipes mushrooms. Generated gas plasma was characterized by its emission spectrum in ambient air, pH, temperature, and H2O2 and NOx concentrations after exposure for various periods. Phenolic and flavonoid contents in the extracts were measured using antioxidant assays and Fourier transform infrared and ultraviolet-visible spectroscopy. We analyzed the effects of the plasma-treated mushroom-derived extracts against breast carcinoma using the MCF7 and MDA-MB231 cell lines. The extracts significantly and concentration dependently inhibited the growth of breast cancer cells without inducing toxicity in normal MCF10A cells, and induced apoptosis via oxidative stress, evidenced by DNA damage (γ-H2AX foci formation), and increased the population of MCF7 breast cancer cells arrested in the G2/M phase of the cell cycle. The extracts also induced mitochondrion-mediated apoptosis of MCF7 cells through cytochrome c release and caspase cleavage activity. The plasma improved the biological activity of mushrooms by increasing their phenolic compounds that prevented the growth of breast cancer cells in vitro.
Collapse
|
6
|
Obidiegwu JE, Lyons JB, Chilaka CA. The Dioscorea Genus (Yam)-An Appraisal of Nutritional and Therapeutic Potentials. Foods 2020; 9:E1304. [PMID: 32947880 PMCID: PMC7555206 DOI: 10.3390/foods9091304] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/11/2020] [Accepted: 09/12/2020] [Indexed: 12/19/2022] Open
Abstract
The quest for a food secure and safe world has led to continuous effort toward improvements of global food and health systems. While the developed countries seem to have these systems stabilized, some parts of the world still face enormous challenges. Yam (Dioscorea species) is an orphan crop, widely distributed globally; and has contributed enormously to food security especially in sub-Saharan Africa because of its role in providing nutritional benefits and income. Additionally, yam has non-nutritional components called bioactive compounds, which offer numerous health benefits ranging from prevention to treatment of degenerative diseases. Pharmaceutical application of diosgenin and dioscorin, among other compounds isolated from yam, has shown more prospects recently. Despite the benefits embedded in yam, reports on the nutritional and therapeutic potentials of yam have been fragmented and the diversity within the genus has led to much confusion. An overview of the nutritional and health importance of yam will harness the crop to meet its potential towards combating hunger and malnutrition, while improving global health. This review makes a conscious attempt to provide an overview regarding the nutritional, bioactive compositions and therapeutic potentials of yam diversity. Insights on how to increase its utilization for a greater impact are elucidated.
Collapse
Affiliation(s)
- Jude E. Obidiegwu
- National Root Crops Research Institute, Umudike, Km 8 Umuahia-Ikot Ekpene Road, P.M.B 7006 Umuahia, Abia State, Nigeria
| | - Jessica B. Lyons
- Department of Molecular and Cell Biology and Innovative Genomics Institute, University of California, Berkeley, 142 Weill Hall #3200, Berkeley, CA 94720-3200, USA;
| | - Cynthia A. Chilaka
- Institute of Pharmacology and Toxicology, Julius Maximilian University of Würzburg, Versbacher Straβe 9, 97078 Würzburg, Germany; or
| |
Collapse
|
7
|
Zhang Y, Tian Z, Wan H, Liu W, Kong F, Ma G. Deltonin Ameliorates Cerebral Ischemia/Reperfusion Injury in Correlation with Modulation of Autophagy and Inflammation. Neuropsychiatr Dis Treat 2020; 16:871-879. [PMID: 32280228 PMCID: PMC7127787 DOI: 10.2147/ndt.s227988] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 01/06/2020] [Indexed: 01/31/2023] Open
Abstract
INTRODUCTION Deltonin, an active component extracted from Dioscorea zingiberensis C.H. WRIGHT, was widely utilized in traditional Chinese medicines. It has been shown to have anti-cancer functions such as colon cancer, breast cancer, and head and neck squamous carcinoma. Herein, we will investigate the role of deltonin in cerebral ischemia/reperfusion injuries. METHODS Ly294002 and anisomycin were used as inhibitors to monitor the effects of deltonin. Middle cerebral artery occlusion I/R model was constructed. Infarct volumes, neurological deficits and brain water contents were evaluated under different conditions. Rotarod test, ELISA, and Western blotting were carried to investigate the effects in vitro. RESULTS We found that deltonin in ischemia/reperfusion (I/R) rats greatly enhanced brain damages as well as neurological functions through up-regulating p-Akt and p-mTOR as well as inhibiting the expressions of LC3-II/LC3-I, Beclin-1, IL-1, TLR4, and p-p38. Deltonin exerted neuroprotection effect through relieving autophagy activity by regulating PI3K/Akt/mTOR signaling. Deltonin suppressed inflammation reactions through modulation TLR4/p38/MAPK signaling as well. CONCLUSION Overall, our data suggested that deltonin could suppress ischemic brain injury by regulating autophagy and inflammation during I/R. Deltonin can be a potential therapeutic method for patient with I/R.
Collapse
Affiliation(s)
- Yi Zhang
- Cerebral Vascular Center, Zhongda Hospital, Southeast University, Nanjing City, Jiangsu Province 210044, People's Republic of China
| | - Zhiming Tian
- Cerebral Vascular Center, Zhongda Hospital, Southeast University, Nanjing City, Jiangsu Province 210044, People's Republic of China
| | - Hongyan Wan
- Cerebral Vascular Center, Zhongda Hospital, Southeast University, Nanjing City, Jiangsu Province 210044, People's Republic of China
| | - Wen Liu
- Cerebral Vascular Center, Zhongda Hospital, Southeast University, Nanjing City, Jiangsu Province 210044, People's Republic of China
| | - Fanping Kong
- Department of Neurology, Fu-Ning People's Hospital, Yancheng City, Jiangsu Province 224400, People's Republic of China
| | - Guoping Ma
- Department of Neurology, Tian-Shui First People's Hospital, Tianshui City, Gansu Province 741000, People's Republic of China
| |
Collapse
|
8
|
Yang HL, Thiyagarajan V, Shen PC, Mathew DC, Lin KY, Liao JW, Hseu YC. Anti-EMT properties of CoQ0 attributed to PI3K/AKT/NFKB/MMP-9 signaling pathway through ROS-mediated apoptosis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:186. [PMID: 31068208 PMCID: PMC6505074 DOI: 10.1186/s13046-019-1196-x] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 04/25/2019] [Indexed: 01/08/2023]
Abstract
Background Breast cancer is the most prevalent cancer among women. In triple-negative breast cancer (TNBC) cells, a novel quinone derivative, coenzyme Q0 (CoQ0), promotes apoptosis and cell-cycle arrest. This study explored the anti-epithelial–mesenchymal transition (EMT) and antimetastatic attributes of CoQ0 in TNBC (MDA-MB-231). Methods Invasion, as well as MTT assays were conducted. Lipofectamine RNAiMAX was used to transfect cells with β-catenin siRNA. Through Western blotting and RT-PCR, the major signaling pathways’ protein expressions were examined, and the biopsied tumor tissues underwent immunohistochemical and hematoxylin and eosin staining as well as Western blotting. Results CoQ0 (0.5–2 μM) hindered tumor migration, invasion, and progression. Additionally, it caused MMP-2/− 9, uPA, uPAR, and VEGF downregulation. Furthermore, in highly metastatic MDA-MB-231 cells, TIMP-1/2 expression was subsequently upregulated and MMP-9 expression was downregulated. In addition, CoQ0 inhibited metastasis and EMT in TGF-β/TNF-α-stimulated non-tumorigenic MCF-10A cells. Bioluminescence imaging of MDA-MB-231 luciferase–injected live mice demonstrated that CoQ0 significantly inhibited metastasis of the breast cancer to the lungs and inhibited the development of tumors in MDA-MB-231 xenografted nude mice. Silencing of β-catenin with siRNA stimulated CoQ0-inhibited EMT. Western blotting as well as histological analysis established that CoQ0 reduced xenografted tumor development because apoptosis induction, cell-cycle inhibition, E-cadherin upregulation, β-catenin downregulation, and metastasis and EMT regulatory protein modulation were observed. Conclusions CoQ0 inhibited the progression of metastasis as well as EMT (in vitro and in vivo). The described approach has potential in treating human breast cancer metastasis. Electronic supplementary material The online version of this article (10.1186/s13046-019-1196-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hsin-Ling Yang
- Institute of Nutrition, College of Biopharmaceuticals and Food Sciences, China Medical University, Taichung, 40402, Taiwan
| | - Varadharajan Thiyagarajan
- Department of Cosmeceutics, College of Biopharmaceutical and Food Sciences, China Medical University, No. 91, Hsueh-Shih Road, Taichung, 40402, Taiwan
| | - Pei-Chun Shen
- Institute of Nutrition, College of Biopharmaceuticals and Food Sciences, China Medical University, Taichung, 40402, Taiwan
| | - Dony Chacko Mathew
- Institute of Nutrition, College of Biopharmaceuticals and Food Sciences, China Medical University, Taichung, 40402, Taiwan
| | - Kai-Yuan Lin
- Department of Medical Research, Chi-Mei Medical Center, Tainan, 710, Taiwan
| | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathology, National Chung Hsing University, Taichung, 40227, Taiwan
| | - You-Cheng Hseu
- Department of Cosmeceutics, College of Biopharmaceutical and Food Sciences, China Medical University, No. 91, Hsueh-Shih Road, Taichung, 40402, Taiwan. .,Department of Health and Nutrition Biotechnology, Asia University, Taichung, 41354, Taiwan. .,Chinese Medicine Research Center, China Medical University, Taichung, 40402, Taiwan. .,Research Center of Chinese Herbal Medicine, China Medical University, Taichung, 40402, Taiwan.
| |
Collapse
|
9
|
A new pyrrole based small molecule from Tinospora cordifolia induces apoptosis in MDA-MB-231 breast cancer cells via ROS mediated mitochondrial damage and restoration of p53 activity. Chem Biol Interact 2019; 299:120-130. [DOI: 10.1016/j.cbi.2018.12.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 11/26/2018] [Accepted: 12/07/2018] [Indexed: 12/17/2022]
|
10
|
Wong JH, Sze SCW, Ng TB, Cheung RCF, Tam C, Zhang KY, Dan X, Chan YS, Shing Cho WC, Ng CCW, Waye MMY, Liang W, Zhang J, Yang J, Ye X, Lin J, Ye X, Wang H, Liu F, Chan DW, Ngan HYS, Sha O, Li G, Tse R, Tse TF, Chan H. Apoptosis and Anti-cancer Drug Discovery: The Power of Medicinal Fungi and Plants. Curr Med Chem 2019; 25:5613-5630. [DOI: 10.2174/0929867324666170720165005] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 05/16/2017] [Accepted: 05/16/2017] [Indexed: 01/21/2023]
Abstract
The purpose of this account is to review the compounds capable of eliciting
mitochondria-mediated apoptosis in cancer cells produced by medicinal fungi and plants.
The medicinal fungi discussed encompass Cordyceps, Ganoderma species, Coriolus versicolor
and Hypsizygus marmoreus. The medicinal plants discussed comprise Astragalus
complanatus, Dendrobium spp, Dioscorea spp, Glycyrrhiza spp, Panax notoginseng,
Panax ginseng, and Momordica charantia. These compounds have the potential of development
into anticancer drugs.
Collapse
Affiliation(s)
- Jack Ho Wong
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Stephen Cho Wing Sze
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Sassoon Road, Hong Kong, China
| | - Tzi Bun Ng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Randy Chi Fai Cheung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Chit Tam
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Kalin Yanbo Zhang
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Sassoon Road, Hong Kong, China
| | - Xiuli Dan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Yau Sang Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - William Chi Shing Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong, China
| | | | - Mary Miu Yee Waye
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Weicheng Liang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Jinfang Zhang
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - Jie Yang
- Fujian Key Laboratory of Marine Enzyme Engineering, Fuzhou University, Fuzhou 350116, Fujian, China
| | - Xiuyun Ye
- Fujian Key Laboratory of Marine Enzyme Engineering, Fuzhou University, Fuzhou 350116, Fujian, China
| | - Juan Lin
- Fujian Key Laboratory of Marine Enzyme Engineering, Fuzhou University, Fuzhou 350116, Fujian, China
| | - Xiujuan Ye
- Key Laboratory of Biopesticide and Chemical Biology, Ministry of Education, and Key Laboratory of Plant Virology of Fujian Province, Institute of Plant Virology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Hexiang Wang
- State Key Laboratory for Agrobiotechnology and Department of Microbiology, China Agricultural University, Beijing 100193, China
| | - Fang Liu
- Department of Microbiology, College of Life Science, Nankai University, Tianjin 300071, China
| | - David Wai Chan
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Hextan Yuen Sheung Ngan
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Ou Sha
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, China
| | - Guohui Li
- Vita Green Pharmaceuticals (HK) Ltd, Vita Green Health Products (HK) Ltd Genning Partners Company Limited, and Hong Kong Institute of Medical Research, Hong Kong, China
| | - Ryan Tse
- Vita Green Pharmaceuticals (HK) Ltd, Vita Green Health Products (HK) Ltd Genning Partners Company Limited, and Hong Kong Institute of Medical Research, Hong Kong, China
| | - Tak Fu Tse
- Vita Green Pharmaceuticals (HK) Ltd, Vita Green Health Products (HK) Ltd Genning Partners Company Limited, and Hong Kong Institute of Medical Research, Hong Kong, China
| | - Helen Chan
- Vita Green Pharmaceuticals (HK) Ltd, Vita Green Health Products (HK) Ltd Genning Partners Company Limited, and Hong Kong Institute of Medical Research, Hong Kong, China
| |
Collapse
|
11
|
Wang J, Li Y, Gao L, Yan F, Gao G, Li L. GSK-3β Inhibitor Alsterpaullone Attenuates MPP +-Induced Cell Damage in a c-Myc-Dependent Manner in SH-SY5Y Cells. Front Cell Neurosci 2018; 12:283. [PMID: 30233322 PMCID: PMC6127625 DOI: 10.3389/fncel.2018.00283] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/09/2018] [Indexed: 12/13/2022] Open
Abstract
Mitochondrial dysfunction plays significant roles in the pathogenesis of Parkinson’s Disease (PD). The inactivation of c-Myc, a down-stream gene of Wnt/β-catenin signaling, may contribute to the mitochondria dysfunction. Inhibition of glycogen synthase kinase 3β (GSK-3β) with Alsterpaullone (Als) can activate the down-stream events of Wnt signaling. Here, we investigated the protective roles of Als against MPP+-induced cell apoptosis in SH-SY5Y cells. The data showed that Als effectively rescued c-Myc from the MPP+-induced decline via Wnt signaling. Furthermore, Als protected SH-SY5Y cells from the MPP+-induced mitochondrial fission and cell apoptosis. However, the protective roles of Als were lost under β-catenin-deficient conditions. These findings indicate that Als, a GSK-3β inhibitor, attenuated the MPP+-induced mitochondria-dependent apoptotic via up-regulation of the Wnt signaling.
Collapse
Affiliation(s)
- Jiancai Wang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yuqian Li
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Li Gao
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Fengqi Yan
- Department of Urology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Guodong Gao
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Lihong Li
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
12
|
Castellanos-Esparza YC, Wu S, Huang L, Buquet C, Shen R, Sanchez-Gonzalez B, García Latorre EA, Boyer O, Varin R, Jiménez-Zamudio LA, Janin A, Vannier JP, Li H, Lu H. Synergistic promoting effects of pentoxifylline and simvastatin on the apoptosis of triple-negative MDA-MB-231 breast cancer cells. Int J Oncol 2018; 52:1246-1254. [PMID: 29436616 DOI: 10.3892/ijo.2018.4272] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 11/14/2017] [Indexed: 11/06/2022] Open
Abstract
Pentoxifylline (PTX), a xanthine family molecule and simvastatin (SIM), an anti-hypercholesterolemic agent, have recently been considered as sensitizers to chemotherapy and radiotherapy. The present in vitro study evaluated their antitumor synergistic effects on MDA‑MB‑231 breast cancer cells characterized by the triple‑negative phenotype (TNP). The anti-proliferative effects of these two agents were evaluated by MTT and clonogenic assays. Cell cycle progression was examined using propidium iodide staining. Apoptosis was investigated by Annexin V labeling, and by examining caspase 3 activity and DNA fragmentation. Autophagic vesicles and reactive oxygen species (ROS) levels were monitored by flow cytometry. Western blot analysis was performed to evaluate molecular targets. Our results revealed that when used alone, PTX and SIM exerted antitumor effects. Nevertheless, used in combination, the inhibition of cell proliferation was synergistically superior (80% vs 42%) than that observed following treatment with each agent alone after 48 h. PTX alone (0.5 mM) induced both apoptosis (25%) and autophagy (25%); however, when used in combination with SIM (0.5 µM), the balance between these processes was disrupted and the cells underwent apoptosis (>65%) as opposed to autophagy (<13%). This imbalance was associated with an increase in ERK1/2 and AKT activation, but not with an increase in mTOR phosphorylation, and with the suppression of the NF-κB pathway. In addition, in the cells treated with both agents, almost 78% of the cells were arrested at the G0/G1 phase and lost their colony-forming ability (38±5%) compared to the cells treated with PTX alone (115±5%). On the whole, these results suggest that the induction of autophagy may be a protective mechanism preventing MDA‑MB‑231 cancer cell death. The combined use of PTX and SIM may drive dormant autophagic cancer cells to undergo apoptosis and thus this may be a novel treatment strategy for breast cancer characterized by the TNP.
Collapse
Affiliation(s)
- Yessica Cristina Castellanos-Esparza
- National Institute of Health and Medical Research, Medical Research Unit S-1165/Paris Diderot University, University Institute of Hematology, Saint-Louis Hospital, 75010 Paris, France
| | - Shuang Wu
- National Institute of Health and Medical Research, Medical Research Unit S-1165/Paris Diderot University, University Institute of Hematology, Saint-Louis Hospital, 75010 Paris, France
| | - Limin Huang
- National Institute of Health and Medical Research, Medical Research Unit S-1165/Paris Diderot University, University Institute of Hematology, Saint-Louis Hospital, 75010 Paris, France
| | - Catherine Buquet
- National Institute of Health and Medical Research, Unit 1234/Rouen University, Faculty of Medicine and Pharmacy, 76183 Rouen, France
| | - Rong Shen
- National Institute of Health and Medical Research, Medical Research Unit S-1165/Paris Diderot University, University Institute of Hematology, Saint-Louis Hospital, 75010 Paris, France
| | - Berenice Sanchez-Gonzalez
- Immunochemistry Laboratory I, Immunology Department, National School of Biological Sciences, National Polytechnic Institute, Mexico City 11340, Mexico
| | - Ethel Awilda García Latorre
- Immunochemistry Laboratory I, Immunology Department, National School of Biological Sciences, National Polytechnic Institute, Mexico City 11340, Mexico
| | - Olivier Boyer
- National Institute of Health and Medical Research, Unit 1234/Rouen University, Faculty of Medicine and Pharmacy, 76183 Rouen, France
| | - Remi Varin
- National Institute of Health and Medical Research, Unit 1234/Rouen University, Faculty of Medicine and Pharmacy, 76183 Rouen, France
| | - Luis Antonio Jiménez-Zamudio
- Immunochemistry Laboratory I, Immunology Department, National School of Biological Sciences, National Polytechnic Institute, Mexico City 11340, Mexico
| | - Anne Janin
- National Institute of Health and Medical Research, Medical Research Unit S-1165/Paris Diderot University, University Institute of Hematology, Saint-Louis Hospital, 75010 Paris, France
| | - Jean-Pierre Vannier
- National Institute of Health and Medical Research, Unit 1234/Rouen University, Faculty of Medicine and Pharmacy, 76183 Rouen, France
| | - Hong Li
- National Institute of Health and Medical Research, Unit 1234/Rouen University, Faculty of Medicine and Pharmacy, 76183 Rouen, France
| | - He Lu
- National Institute of Health and Medical Research, Medical Research Unit S-1165/Paris Diderot University, University Institute of Hematology, Saint-Louis Hospital, 75010 Paris, France
| |
Collapse
|
13
|
Wang Y, Mo H, Gu J, Chen K, Han Z, Liu Y. Cordycepin induces apoptosis of human acute monocytic leukemia cells via downregulation of the ERK/Akt signaling pathway. Exp Ther Med 2017; 14:3067-3073. [PMID: 28912858 PMCID: PMC5585717 DOI: 10.3892/etm.2017.4855] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 05/19/2017] [Indexed: 12/25/2022] Open
Abstract
The aim of the present study was to examine the apoptotic effect of cordycepin (COR) on human THP-1 acute monocytic leukemia cells. THP-1 cells were exposed to different concentrations of COR for 24, 48, 72 or 96 h. The cell viability and apoptotic rate were analyzed. The gene expression of Akt1, Akt2, Akt3, B-cell lymphoma 2 (Bcl-2) and Bcl-2-associated X protein (Bax) were assessed by reverse-transcription quantitative PCR. Western blot analysis was used to detect the protein levels of phosphorylated (p)-Akt, p-extracellular signal-regulated kinase (ERK) and cleaved caspase-3. It was found that the viability of THP-1 cells was inhibited by COR in a dose- and time-dependent manner. After treatment with 200 µM COR for 24 h, the percentage of apoptotic cells was significantly increased. COR also downregulated the levels of Bcl-2, Akt1, Akt2 and Akt3, and elevated the expression of Bax. The protein levels of p-Akt and p-ERK were suppressed and cleaved caspase-3 was increased after treatment of COR. In conclusion, COR was found to induce apoptosis of THP-1 acute monocytic leukemia cells through downregulation of ERK/Akt signaling.
Collapse
Affiliation(s)
- Yue Wang
- Department of Cardiology, Ninth People's Hospital, Shanghai Jiaotong University Medical School, Shanghai 200011, P.R. China
| | - Huimin Mo
- Institute of Hematology, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Jun Gu
- Department of Cardiology, Ninth People's Hospital, Shanghai Jiaotong University Medical School, Shanghai 200011, P.R. China
| | - Kan Chen
- Department of Cardiology, Ninth People's Hospital, Shanghai Jiaotong University Medical School, Shanghai 200011, P.R. China
| | - Zhihua Han
- Department of Cardiology, Ninth People's Hospital, Shanghai Jiaotong University Medical School, Shanghai 200011, P.R. China
| | - Yi Liu
- Department of Ultrasound, Renji Hospital, Shanghai Jiaotong University Medical School, Shanghai 200127, P.R. China
| |
Collapse
|
14
|
Abdelrahman M, Mahmoud HYAH, El-Sayed M, Tanaka S, Tran LS. Isolation and characterization of Cepa2, a natural alliospiroside A, from shallot (Allium cepa L. Aggregatum group) with anticancer activity. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2017; 116:167-173. [PMID: 28577504 DOI: 10.1016/j.plaphy.2017.05.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 05/16/2017] [Accepted: 05/16/2017] [Indexed: 06/07/2023]
Abstract
Exploration of new and promising anticancer compounds continues to be one of the main tasks of cancer research because of the drug resistance, high cytotoxicity and limitations of tumor selectivity. Natural products represent a better choice for cancer treatment in comparison with synthetic compounds because of their pharmacokinetic properties and lower side effects. In the current study, we isolated a steroidal saponin, named Cepa2, from the dry roots of shallot (Allium cepa L. Aggregatum group), and determined its structure by using two-dimensional nuclear manganic resonance (2D NMR). The 1H NMR and 13C NMR data revealed that the newly isolated Cepa2 compound is identical to alliospiroside A (C38H60O12) [(25S)-3β-hydroxyspirost-5-en-1β-yl-2-O-(6-deoxy-α-L-mannopyranosyl)-α-L-arabinopyranoside], whose anticancer activity remains elusive. Our in vitro examination of the cytotoxic activity of the identified Cepa2 against P3U1 myeloma cancer cell line showed its high efficiency as an anticancer with 91.13% reduction in P3U1 cell viability 12 h post-treatment. The reduction of cell viability was correlated with the increase in reactive oxygen species levels in Cepa2-treated P3U1 cells, as compared with untreated cells. Moreover, scanning electron microscope results demonstrated apoptosis of the Cepa2-treated P3U1 cells in a time course-dependent manner. The results of our study provide evidence for the anticancer properties of the natural Cepa2/alliospiroside A extracted from shallot plants, and a strong foundation for in-depth investigations to build theoretical bases for cell apoptosis and development of novel anticancer drugs.
Collapse
Affiliation(s)
- Mostafa Abdelrahman
- Graduate School of Life Sciences, Tohoku University 2-1-1, Katahira, Aoba-ku, Sendai, 980-8577, Japan; Botany Department Faculty of Science, Aswan University, Aswan 81528, Egypt
| | - Hassan Y A H Mahmoud
- Division of infectious Diseases, Animal Medicine Department, Faculty of Veterinary Medicine, South Valley University, Qena 83523, Egypt
| | - Magdi El-Sayed
- Botany Department Faculty of Science, Aswan University, Aswan 81528, Egypt
| | - Shuhei Tanaka
- Department of Biological and Environmental Sciences, Faculty of Agriculture, Yamaguchi University, Yamaguchi 753-8515, Japan
| | - L S Tran
- Plant Abiotic Stress Research Group & Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City 70000, Viet Nam; Signaling Pathway Research Unit, RIKEN Center for Sustainable Resource Science, 1-7-22, Suehiro-cho, Tsurumiku, Yokohama 230-0045, Japan.
| |
Collapse
|
15
|
Paris saponin-induced autophagy promotes breast cancer cell apoptosis via the Akt/mTOR signaling pathway. Chem Biol Interact 2017; 264:1-9. [DOI: 10.1016/j.cbi.2017.01.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 12/08/2016] [Accepted: 01/10/2017] [Indexed: 11/20/2022]
|
16
|
Zhang C, Jia X, Bao J, Chen S, Wang K, Zhang Y, Li P, Wan JB, Su H, Wang Y, Mei Z, He C. Polyphyllin VII induces apoptosis in HepG2 cells through ROS-mediated mitochondrial dysfunction and MAPK pathways. Altern Ther Health Med 2016; 16:58. [PMID: 26861252 PMCID: PMC4746894 DOI: 10.1186/s12906-016-1036-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 02/02/2016] [Indexed: 01/15/2023]
Abstract
Background Paris polyphylla is an oriental folk medicine that has anticancer activities both in vivo and in vitro. Polyphyllin VII (PP7), a pennogenyl saponin from P. polyphylla has been found to exert strong anticancer activity. However, the underlying mechanisms are poorly understood. In the present study, the anticancer effect of polyphyllin VII against human liver cancer cells and the molecular mechanisms were investigated. Methods Cellular viability was measured by MTT assay. Apoptosis, intracellular reactive oxygen species (ROS) and mitochondrial membrane potential levels were evaluated using the InCell 2000 confocal microscope. The expression levels of apoptotic-related proteins were evaluated by Western blotting. Results PP7 strongly inhibited the cell growth and induced apoptosis and necrosis in hepatocellular carcinoma HepG2 cells. Meanwhile, PP7 up-regulated the levels of Bax/Bcl-2, cytochrome c, the cleaved forms of caspases-3, -8, -9, and poly (ADP-ribose) polymerase in a dose- and time-dependent manner, indicating that PP7 induced apoptosis in HepG2 cells through both intrinsic and extrinsic pathways. Moreover, PP7 provoked the production of intracellular ROS and the depolarization of mitochondrial membrane potential. Further analysis showed that PP7 significantly augmented the phosphorylation of JNK, ERK and p38, the major components of mitogen-activated protein kinase (MAPK) pathways, and the expressions of tumor suppressor proteins p53 and PTEN. In addition, PP7-induced apoptosis was remarkably attenuated by MAPK inhibitors and ROS inhibitor. Conclusions These results demonstrated that PP7 induced apoptotic cell death in HepG2 cells through both intrinsic and extrinsic pathways by promoting the generation of mitochondrial-mediated ROS and activating MAPK and PTEN/p53 pathways. Electronic supplementary material The online version of this article (doi:10.1186/s12906-016-1036-x) contains supplementary material, which is available to authorized users.
Collapse
|
17
|
Antognelli C, Palumbo I, Aristei C, Talesa VN. Glyoxalase I inhibition induces apoptosis in irradiated MCF-7 cells via a novel mechanism involving Hsp27, p53 and NF-κB. Br J Cancer 2014; 111:395-406. [PMID: 24918814 PMCID: PMC4102940 DOI: 10.1038/bjc.2014.280] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 04/24/2014] [Accepted: 04/30/2014] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Glyoxalase I (GI) is a cellular defence enzyme involved in the detoxification of methylglyoxal (MG), a cytotoxic byproduct of glycolysis, and MG-derived advanced glycation end products (AGEs). Argpyrimidine (AP), one of the major AGEs coming from MG modifications of proteins arginines, is a pro-apoptotic agent. Radiotherapy is an important modality widely used in cancer treatment. Exposure of cells to ionising radiation (IR) results in a number of complex biological responses, including apoptosis. The present study was aimed at investigating whether, and through which mechanism, GI was involved in IR-induced apoptosis. METHODS Apoptosis, by TUNEL assay, transcript and protein levels or enzymatic activity, by RT-PCR, western blot and spectrophotometric methods, respectively, were evaluated in irradiated MCF-7 breast cancer cells, also in experiments with appropriate inhibitors or using small interfering RNA. RESULTS Ionising radiation induced a dramatic reactive oxygen species (ROS)-mediated inhibition of GI, leading to AP-modified Hsp27 protein accumulation that, in a mechanism involving p53 and NF-κB, triggered an apoptotic mitochondrial pathway. Inhibition of GI occurred at both functional and transcriptional levels, the latter occurring via ERK1/2 MAPK and ERα modulation. CONCLUSIONS Glyoxalase I is involved in the IR-induced MCF-7 cell mitochondrial apoptotic pathway via a novel mechanism involving Hsp27, p53 and NF-κB.
Collapse
Affiliation(s)
- C Antognelli
- Department of Experimental Medicine, University of Perugia, Sant'Andrea delle Fratte, 06132 Perugia, Italy
| | - I Palumbo
- Radiation Oncology Section, University of Perugia, Sant'Andrea delle Fratte, 06132 Perugia, Italy
| | - C Aristei
- Radiation Oncology Section, University of Perugia, Sant'Andrea delle Fratte, 06132 Perugia, Italy
| | - V N Talesa
- Department of Experimental Medicine, University of Perugia, Sant'Andrea delle Fratte, 06132 Perugia, Italy
| |
Collapse
|
18
|
Evidence in support of potential applications of lipid peroxidation products in cancer treatment. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:931251. [PMID: 24369491 PMCID: PMC3867858 DOI: 10.1155/2013/931251] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Revised: 11/02/2013] [Accepted: 11/08/2013] [Indexed: 01/10/2023]
Abstract
Cancer cells generate reactive oxygen species (ROS) resulting from mitochondrial dysfunction, stimulation of oncogenes, abnormal metabolism, and aggravated inflammatory activities. Available evidence also suggests that cancer cells depend on intrinsic ROS level for proliferation and survival. Both physiological and pathophysiological roles have been ascribed to ROS which cause lipid peroxidation. In spite of their injurious effects, the ROS and the resulting lipid peroxidation products could be beneficial in cancer treatment. This review presents research findings suggesting that ROS and the resulting lipid peroxidation products could be utilized to inhibit cancer growth or induce cancer cell death. It also underscores the potential of lipid peroxidation products to potentiate the antitumor effect of other anticancer agents. The review also highlights evidence demonstrating other potential applications of lipid peroxidation products in cancer treatment. These include the prospect of lipid peroxidation products as a diagnostic tool to predict the chances of cancer recurrence, to monitor treatment progress or how well cancer patients respond to therapy. Further and detailed research is required on how best to successfully, effectively, and selectively target cancer cells in humans using lipid peroxidation products. This may prove to be an important strategy to complement current treatment regimens for cancer patients.
Collapse
|