1
|
Laksana C, Sophiphun O, Nualkaew S, Chanprame S. In vitro propagation of Stephania pierrei diels and exploration of its potential as sustainable phytochemical production from tuber and callus. BMC PLANT BIOLOGY 2025; 25:377. [PMID: 40133844 PMCID: PMC11934495 DOI: 10.1186/s12870-025-06422-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND The tuber of Stephania pierrei Diels has been used for treating cardiovascular disease, migraine, and body edema and may exhibit antimalarial, anticancer, and anticholinesterase properties. It is also a popular ornamental plant. Consequently, plant tubers have been extensively harvested from the wild, posing a high risk of extinction. We assessed the in vitro propagation and essential phytochemical production from the calli of S. pierrei. RESULTS The highest callus weight (1.52-1.59 g) induced from the tuber flesh without peel occurred when using Murashige and Skoog (MS) medium with basal salts supplemented with 3-4 mg/L benzyladenine (BA) and 0.1 or 0.5 mg/L naphthaleneacetic acid. MS medium with a concentration of 3 mg/L BA was the most efficient medium for shoot regeneration, producing an average of 7.40 ± 1.140 shoots. Similarly, MS medium with 1 mg/L meta-topolin (mT) was most efficient for multiple shoot production (with an average of 13.40 ± 2.30 shoots). Root induction was successfully performed using the ½ MS medium. HPLC analysis revealed that calluses, tubers without peel, tubers with peels, and peels contained fangchinoline, cepharantine, and tetrandrine. Peels had the highest amounts of the first two alkaloids and a high amount of tetrandrine, which was related to the relative expression levels of three genes involved in the isoquinoline alkaloid biosynthesis pathway: coclaurine N-methyltransferase (CNMT), norcoclaurine synthase (NCS), and 6-O-methyltransferase (6OMT). GC-MS was employed for phytochemical identification of callus and tuber with peel, which revealed that out of the 24 phytochemicals identified, 13 were only found in callus and 5 were present only in tubers. DPPH scavenging percentage, ferric reducing antioxidant power, and ABTS radical cation scavenging activity assays revealed that the extracts from the four tissues showed antioxidant activities. The peel showed significantly higher total flavonoid and phenolic content, whereas the callus displayed the highest total alkaloid concentration. CONCLUSIONS Producing secondary metabolites such as cepharantine, tetrandrine, and fangchinoline by culturing callus holds potential as a low-cost and sustainable method for producing pharmaceutical phytochemicals.
Collapse
Affiliation(s)
- Chanakan Laksana
- Faculty of Agricultural Technology, Burapha University Sakaeo Campus, Sakaeo, 27160, Thailand
| | - Onsulang Sophiphun
- Faculty of Agricultural Technology, Burapha University Sakaeo Campus, Sakaeo, 27160, Thailand
| | - Somsak Nualkaew
- Pharmaceutical Chemistry and Natural Product Research Unit, Faculty of Pharmacy, Mahasarakham University, Mahasarakham, 44150, Thailand
| | - Sontichai Chanprame
- Department of Agronomy, Faculty of Agriculture at Kamphaeng Saen, Kasetsart University, Nakhon Pathom, 73140, Thailand.
| |
Collapse
|
2
|
Li Q, Ye Z, Wang G, Chen Y, Deng J, Wang D, Wang Y. Natural Products as Novel Therapeutic Agents for Triple-Negative Breast Cancer: Current Evidence, Mechanisms, Challenges, and Opportunities. Molecules 2025; 30:1201. [PMID: 40141978 PMCID: PMC11944566 DOI: 10.3390/molecules30061201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/23/2025] [Accepted: 03/02/2025] [Indexed: 03/28/2025] Open
Abstract
Breast cancer (BC) tops the list of causes for female fatalities globally, with the elusive triple-negative breast cancer (TNBC) constituting 10-20% of all cases. Current clinical strategies for combating TNBC encompass a multifaceted approach, including surgical intervention, radiation therapy, chemotherapy, and advanced targeted drugs and immunotherapies. While these modalities have catalyzed significant advancements in TNBC management, lingering limitations continue to pose formidable challenges. There is an acute need for novel therapeutics in the realm of TNBC treatment. Natural products (NPs) have emerged as a rich reservoir for pharmaceutical innovation, owing to their extraordinary range of structures and physicochemical properties. Scholars have reported diverse evidence of NPs' efficacy against TNBC. This review aims to comprehensively explore the bioactive constituents, specifics and commonalities of chemical structure, and pharmacological mechanisms of NPs, specifically examining their multifaceted roles in impeding TNBC. NPs, which have recently garnered significant interest, are intriguing in terms of their capacity to combat TNBC through multifaceted mechanisms, including the suppression of tumor cell proliferation, the induction of apoptosis, and the inhibition of tumor metastasis. These natural agents primarily encompass a range of compounds, including terpenoids, glycosides, phenolic compounds, and alkaloids. An in-depth exploration has unveiled their involvement in key signaling pathways, including the transforming growth factor-beta (TGF-β), vascular endothelial growth factor A (VEGFA), phosphoinositide 3-kinase/protein kinase B (PI3K/AKT), Wingless/Int-1 (Wnt) /β-catenin, and mitogen-activated protein kinase (MAPK) pathways. Meanwhile, this review also looks at the challenges and opportunities that arise from harnessing natural compounds to influence TNBC, while outlining the prospective trajectory for future research in the field of NPs.
Collapse
Affiliation(s)
- Qingzhou Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China;
| | - Zhen Ye
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Z.Y.); (G.W.); (Y.C.); (J.D.)
| | - Guilin Wang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Z.Y.); (G.W.); (Y.C.); (J.D.)
| | - Yuhui Chen
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Z.Y.); (G.W.); (Y.C.); (J.D.)
| | - Jinghong Deng
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Z.Y.); (G.W.); (Y.C.); (J.D.)
| | - Dong Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China;
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Z.Y.); (G.W.); (Y.C.); (J.D.)
| | - Yumei Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China;
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Z.Y.); (G.W.); (Y.C.); (J.D.)
| |
Collapse
|
3
|
Valsan A, Omanakuttan VK, Radhakrishnan KV, Maiti KK. A Comprehensive Appraisal of Bisbenzylisoquinoline Alkaloids Isolated From Genus Cyclea for Anticancer Potential. J Biochem Mol Toxicol 2025; 39:e70137. [PMID: 39835479 DOI: 10.1002/jbt.70137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/17/2024] [Accepted: 12/29/2024] [Indexed: 01/22/2025]
Abstract
The pharmaceutical industry and academia are continuously searching for novel and effective anticancer lead compounds to ensure patient safety, provide a cure, and surpass all other obstacles. Given the indeterminate nature of cancer etiology, the importance of drugs capable of targeting multiple pathways cannot be overstated. Among naturally occurring compounds, bisbenzylisoquinoline (BBIQ) alkaloids, such as berberine, tetrandrine, chelidonine, and berbamine, have demonstrated significant anticancer potential by modulating diverse signaling pathways. Several of these compounds are currently in clinical trials, highlighting their relevance in cancer treatment. This review emphasizes the need for further investigation into the anticancer properties of BBIQ alkaloids, particularly those isolated from eight Cyclea species in India. With around 27 BBIQ alkaloids identified, these compounds hold promise, especially in combating multidrug resistance-a critical challenge in cancer therapy. Given the rising cancer incidence, these alkaloids warrant a deeper exploration of their therapeutic potential.
Collapse
Affiliation(s)
- Alisha Valsan
- Chemical Sciences and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Vishnu K Omanakuttan
- Chemical Sciences and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kokuvayil Vasu Radhakrishnan
- Chemical Sciences and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kaustabh Kumar Maiti
- Chemical Sciences and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
4
|
Bhutta ZA, Choi KC. Phytochemicals as Novel Therapeutics for Triple-Negative Breast Cancer: A Comprehensive Review of Current Knowledge. Phytother Res 2025; 39:364-396. [PMID: 39533509 DOI: 10.1002/ptr.8376] [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: 01/14/2024] [Revised: 05/10/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Triple-negative breast cancer is a characteristic subtype of breast cancer that lacks the estrogen receptor, human epidermal growth factor receptor 2, and progesterone receptor. Because of its highly diverse subtypes, increased metastasis capability, and poor prognosis, the risk of mortality for people with triple-negative breast cancers is high as compared with other cancers. Chemotherapy is currently playing a major role in treating triple-negative breast cancer patients; however, poor prognosis due to drug resistance is causing serious concern. Recent studies on several phytochemicals derived from various plants being used in Traditional Chinese Medicine, Traditional Korean Medicine, Ayurveda (Traditional Indian Medicine), and so on, have demonstrated to be a promising agent as a viable therapy against triple-negative breast cancer. Phytochemicals categorized as alkaloids, polyphenols, terpenoids, phytosterols, and organosulfur compounds have been demonstrated to reduce cancer cell proliferation and metastasis by activating various molecular pathways, thereby reducing the spread of triple-negative breast cancer. This review analyzes the molecular mechanisms by which various phytochemicals fight triple-negative breast cancer and offers a perspective on the difficulties and potential prospects for treating triple-negative breast cancer with various phytochemicals.
Collapse
Affiliation(s)
- Zeeshan Ahmad Bhutta
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|
5
|
Hu SY, Lin TH, Chen CY, He YH, Huang WC, Hsieh CY, Chen YH, Chang WC. Stephania tetrandra and Its Active Compound Coclaurine Sensitize NSCLC Cells to Cisplatin through EFHD2 Inhibition. Pharmaceuticals (Basel) 2024; 17:1356. [PMID: 39458997 PMCID: PMC11510146 DOI: 10.3390/ph17101356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/19/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Adjuvant chemotherapy, particularly cisplatin, is recommended for non-small cell lung carcinoma (NSCLC) patients at high risk of recurrence. EF-hand domain-containing protein D2 (EFHD2) has been recently shown to increase cisplatin resistance and is significantly associated with recurrence in early-stage NSCLC patients. Natural products, commonly used as phytonutrients, are also recognized for their potential as pharmaceutical anticancer agents. RESULT In this study, a range of Chinese herbs known for their antitumor or chemotherapy-enhancing properties were evaluated for their ability to inhibit EFHD2 expression in NSCLC cells. Among the herbs tested, Stephania tetrandra (S. tetrandra) exhibited the highest efficacy in inhibiting EFHD2 and sensitizing cells to cisplatin. Through LC-MS identification and functional assays, coclaurine was identified as a key molecule in S. tetrandra responsible for EFHD2 inhibition. Coclaurine not only downregulated EFHD2-related NOX4-ABCC1 signaling and enhanced cisplatin sensitivity, but also suppressed the stemness and metastatic properties of NSCLC cells. Mechanistically, coclaurine disrupted the interaction between the transcription factor FOXG1 and the EFHD2 promoter, leading to a reduction in EFHD2 transcription. Silencing FOXG1 further inhibited EFHD2 expression and sensitized NSCLC cells to cisplatin. CONCLUSIONS S. tetrandra and its active compound coclaurine may serve as effective adjuvant therapies to improve cisplatin efficacy in the treatment of NSCLC.
Collapse
Affiliation(s)
- Shu-Yu Hu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404333, Taiwan; (S.-Y.H.); (Y.-H.H.); (W.-C.H.)
| | - Tsai-Hui Lin
- Department of Chinese Medicine, China Medical University Hospital, Taichung 404327, Taiwan;
| | - Chung-Yu Chen
- Research Center for Cancer Biology, China Medical University, Taichung 406040, Taiwan;
| | - Yu-Hao He
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404333, Taiwan; (S.-Y.H.); (Y.-H.H.); (W.-C.H.)
- Center for Molecular Medicine, China Medical University Hospital, Taichung 406040, Taiwan
- Program for Cancer Biology and Drug Discovery, China Medical University, Taichung 404333, Taiwan
| | - Wei-Chien Huang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404333, Taiwan; (S.-Y.H.); (Y.-H.H.); (W.-C.H.)
- Center for Molecular Medicine, China Medical University Hospital, Taichung 406040, Taiwan
- Program for Cancer Biology and Drug Discovery, China Medical University, Taichung 404333, Taiwan
- School of Pharmacy, China Medical University, Taichung 404333, Taiwan
| | - Ching-Yun Hsieh
- Division of Hematology and Oncology, Department of internal medicine, China Medical University Hospital, Taichung 404327, Taiwan;
| | - Ya-Huey Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404333, Taiwan; (S.-Y.H.); (Y.-H.H.); (W.-C.H.)
- Center for Molecular Medicine, China Medical University Hospital, Taichung 406040, Taiwan
- Program for Cancer Biology and Drug Discovery, China Medical University, Taichung 404333, Taiwan
| | - Wei-Chao Chang
- Center for Molecular Medicine, China Medical University Hospital, Taichung 406040, Taiwan
| |
Collapse
|
6
|
Gao SC, Dong MZ, Zhao BW, Liu SL, Guo JN, Sun SM, Li YY, Xu YH, Wang ZB. Fangchinoline inhibits mouse oocyte meiosis by disturbing MPF activity. Toxicol In Vitro 2024; 99:105876. [PMID: 38876226 DOI: 10.1016/j.tiv.2024.105876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 06/01/2024] [Accepted: 06/10/2024] [Indexed: 06/16/2024]
Abstract
Fangchinoline (FA) is an alkaloid derived from the traditional Chinese medicine Fangji. Numerous studies have shown that FA has a toxic effect on various cancer cells, but little is known about its toxic effects on germ cells, especially oocytes. In this study, we investigated the effects of FA on mouse oocyte maturation and its potential mechanisms. Our results showed that FA did not affect meiosis resumption but inhibited the first polar body extrusion. This inhibition is not due to abnormalities at the organelle level, such as chromosomes and mitochondrial, which was proved by detection of DNA damage and reactive oxygen species. Further studies revealed that FA arrested the oocyte at the metaphase I stage, and this arrest was not caused by abnormal kinetochore-microtubule attachment or spindle assembly checkpoint activation. Instead, FA inhibits the activity of anaphase-promoting complexes (APC/C), as evidenced by the inhibition of CCNB1 degeneration. The decreased activity of APC/C may be due to a reduction in CDC25B activity as indicated by the high phosphorylation level of CDC25B (Ser323). This may further enhance Maturation-Promoting Factor (MPF) activity, which plays a critical role in meiosis. In conclusion, our study suggests that the metaphase I arrest caused by FA may be due to abnormalities in MPF and APC/C activity.
Collapse
Affiliation(s)
- Shi-Cai Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Ming-Zhe Dong
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Bing-Wang Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Sai-Li Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jia-Ni Guo
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Si-Min Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yuan-Yuan Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Yuan-Hong Xu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Zhen-Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
7
|
Zhang QY, Li JQ, Li Q, Zhang Y, Zhang ZR, Li XD, Zhang HQ, Deng CL, Yang FX, Xu Y, Zhang B. Identification of fangchinoline as a broad-spectrum enterovirus inhibitor through reporter virus based high-content screening. Virol Sin 2024; 39:301-308. [PMID: 38452856 DOI: 10.1016/j.virs.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 02/26/2024] [Indexed: 03/09/2024] Open
Abstract
Hand, foot, and mouth disease (HFMD) is a common pediatric illness mainly caused by enteroviruses, which are important human pathogens. Currently, there are no available antiviral agents for the therapy of enterovirus infection. In this study, an excellent high-content antiviral screening system utilizing the EV-A71-eGFP reporter virus was developed. Using this screening system, we screened a drug library containing 1042 natural compounds to identify potential EV-A71 inhibitors. Fangchinoline (FAN), a bis-benzylisoquinoline alkaloid, exhibits potential inhibitory effects against various enteroviruses that cause HFMD, such as EV-A71, CV-A10, CV-B3 and CV-A16. Further investigations revealed that FAN targets the early stage of the enterovirus life cycle. Through the selection of FAN-resistant EV-A71 viruses, we demonstrated that the VP1 protein could be a potential target of FAN, as two mutations in VP1 (E145G and V258I) resulted in viral resistance to FAN. Our research suggests that FAN is an efficient inhibitor of EV-A71 and has the potential to be a broad-spectrum antiviral drug against human enteroviruses.
Collapse
Affiliation(s)
- Qiu-Yan Zhang
- The Joint Center of Translational Precision Medicine, Department of Infections and Diseases, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou, 510623, China; Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jia-Qi Li
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Qi Li
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Yang Zhang
- University of Science and Technology of China, Department of Life Sciences and Medicine, Hefei, 230026, China
| | - Zhe-Rui Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xiao-Dan Li
- Hunan Normal University, School of Medicine, Changsha, 410081, China
| | - Hong-Qing Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Cheng-Lin Deng
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Feng-Xia Yang
- The Joint Center of Translational Precision Medicine, Department of Infections and Diseases, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou, 510623, China
| | - Yi Xu
- The Joint Center of Translational Precision Medicine, Department of Infections and Diseases, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou, 510623, China.
| | - Bo Zhang
- The Joint Center of Translational Precision Medicine, Department of Infections and Diseases, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou, 510623, China; Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China.
| |
Collapse
|
8
|
Sharma P, Gupta K, Khandai SK, Malik S, Thareja S. Phytometabolites as modulators of breast cancer: a comprehensive review of mechanistic insights. Med Oncol 2024; 41:45. [PMID: 38172452 DOI: 10.1007/s12032-023-02269-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 11/22/2023] [Indexed: 01/05/2024]
Abstract
Breast cancer (BC) is a highly debilitating malignancy affecting females globally and imposing a substantial burden on healthcare systems in both developed and developing nations. Despite the application of conventional therapeutic modalities such as chemotherapy, radiation therapy, and hormonal intervention, BC frequently exhibits resistance, necessitating the urgent development of novel, cost-effective, and accessible treatment strategies. In this context, there is a growing scientific interest in exploring the pharmacological potential of chemical compounds derived from botanical sources, which often exhibit notable biological activity. Extensive in vitro and in vivo investigations have revealed the capacity of these compounds, referred to as phytochemicals, to attenuate the metastatic cascade and reduce the risk of cancer dissemination. These phytochemicals exert their effects through modulation of key molecular and metabolic processes, including regulation of the cell cycle, induction of apoptotic cell death, inhibition of angiogenesis, and suppression of metastatic progression. To shed light on the latest advancements in this field, a comprehensive review of the scientific literature has been conducted, focusing on secondary metabolite agents that have recently been investigated and have demonstrated promising anticancer properties. This review aims to delineate their underlying mechanisms of action and elucidate the associated signaling pathways, thereby contributing to a deeper understanding of their therapeutic potential in the context of BC management.
Collapse
Affiliation(s)
- Priyanka Sharma
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Khushi Gupta
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Sumit Kumar Khandai
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Sonia Malik
- Laboratory of Woody Plants and Crops Biology, University of Orleans, Orleans, France
| | - Suresh Thareja
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India.
| |
Collapse
|
9
|
Li B, Chen J, He J, Peng J, Wang Y, Liu S, Jiang Y. Total alkaloids in Stephania tetrandra induce apoptosis by regulating BBC3 in human non-small cell lung cancer cells. Biomed Pharmacother 2023; 162:114635. [PMID: 37044023 DOI: 10.1016/j.biopha.2023.114635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/23/2023] [Accepted: 03/30/2023] [Indexed: 04/14/2023] Open
Abstract
PURPOSE This study investigated the effects of total alkaloids in Stephania tetrandra (TAS) and the main alkaloid components tetrandrine, fangchinoline and cepharanthine on the biological function of lung cancer cells and the mechanism underlying the synergistic antitumor effects of TAS and cisplatin. METHODS RNA sequencing analysis was performed on TAS-treated H1299 cells. Differentially expressed genes were identified and analyzed, and the regulatory pathway was identified by gene set enrichment analysis. The mRNA and protein expression levels of the differentially expressed genes in cells were determined using quantitative reverse transcription-polymerase chain reaction and western blotting, respectively. Cell viability and wound healing assays evaluated the biological function of TAS and the main alkaloid components in non-small cell lung cancer (NSCLC) cells. Flow cytometry was used to determine the apoptosis rate in NSCLC cells. RESULTS TAS inhibited the proliferation and migration of A549 and H1299 cells and increased the apoptosis rate in a time- and dose-dependent manner. When H1299 cells were treated with TAS (7.5 µg/ml), MGLL and BBC3 were identified as the possible differentially expressed genes. Pathways associated with cisplatin resistance were screened to investigate the effect of TAS on the apoptosis of NSCLC cells. TAS may regulate fatty acid metabolism and induce apoptosis through the upregulated expression of MGLL and BBC3. The combination of TAS at noncytotoxic concentrations (A549: 1.0 μg/ml; H1299: 3.0 μg/ml) and cisplatin significantly inhibited the viability of A549 and H1299 cells. CONCLUSION TAS and the main alkaloid components exert anticancer activity in NSCLC by regulating tumor cell proliferation and apoptosis. Therefore, TAS and the main alkaloid components have the potential to be used as multi-targeted drugs for lung cancer treatment.
Collapse
Affiliation(s)
- Bichen Li
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Juan Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Jia He
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Jing Peng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yuxin Wang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Shao Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| | - Yueping Jiang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| |
Collapse
|
10
|
Li S, Ni N, Wu X, Lan T, Yu Y. Protective Effect of Fangchinoline Against Glaucoma and Neuroinflammation in Unilateral Ocular Hypertension in Mice. INT J PHARMACOL 2023. [DOI: 10.3923/ijp.2023.131.138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
|
11
|
Yang LJ, Yang ZD, Li ZJ, Yang SH, Shu ZM. Stephtetrandrine A-D, bisbenzylisoquinoline alkaloids from Stephania tetrandra. Nat Prod Res 2023; 37:204-215. [PMID: 34348525 DOI: 10.1080/14786419.2021.1961135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Four undescribed bisbenzylisoquinoline alkaloids, designated as Stephtetrandrine A-D, were isolated from the roots of Stephania tetrandra. Their structures were elucidated by IR, HRESIMS, ECD spectra, 1 D and 2 D NMR spectra and comparison with the literature data. Additional five known compounds (limacine, tetrandrine, N-trans-Feruloyltyramine, 2'-N-chloromethyltetrandrine, 2,2'-N-N-dichloromethyltetrandrine) were also isolated. N-trans-Feruloyltyramine was isolated from Stephania tetrandra for the first time. The isolated compounds were tested for monoamine oxidase, acetylcholinesterase, phosphoinositide 3-kinase α and human hepatoma cell HepG2 inhibitory activities. Stephtetrandrine C showed obvious inhibitory effect on human hepatoma HepG2, with IC50 value of 16.2 μM. Limacine and 2'-N-chloromethyltetrandrine showed moderate monoamine oxidase inhibitory effect with the IC50 values of 37.7 and 29.2 μM, respectively.
Collapse
Affiliation(s)
- Li-Jun Yang
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, P.R. China.,The Provincial Education Key Laboratory of Screening, Evaluation and Advanced Processing of Traditional Chinese Medicine and Tibetan Medicine, School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, P.R. China
| | - Zhong-Duo Yang
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, P.R. China
| | - Zhi-Jie Li
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, P.R. China
| | - Shu-Hong Yang
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, P.R. China
| | - Zong-Mei Shu
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, P.R. China
| |
Collapse
|
12
|
Jung YY, Um JY, Sethi G, Ahn KS. Fangchinoline abrogates growth and survival of hepatocellular carcinoma by negative regulation of c-met/HGF and its associated downstream signaling pathways. Phytother Res 2022; 36:4542-4557. [PMID: 35867025 DOI: 10.1002/ptr.7573] [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: 01/04/2022] [Revised: 06/21/2022] [Accepted: 07/09/2022] [Indexed: 12/13/2022]
Abstract
Among all cancers, hepatocellular carcinoma (HCC) remains a lethal disease with limited treatment options. In this study, we have analyzed the possible inhibitory effects of Fangchinoline (FCN) on c-Met, a protein known to regulate the rapid phosphorylation of downstream signals, as well as mediate aberrant growth, metastasis, survival, and motility in cancer. FCN inhibited the activation of c-Met and its downstream signals PI3K, AKT, mTOR, MEK, and ERK under in vitro settings. Moreover, c-Met gene silencing lead to suppression of PI3K/AKT/mTOR and MEK/ERK signaling pathways, and induced apoptotic cell death upon exposure to FCN. In addition, FCN markedly inhibited the expression of the various oncogenic proteins such as Bcl-2/xl, survivin, IAP-1/2, cyclin D1, and COX-2. In vivo studies in HepG2 cells xenograft mouse model showed that FCN could significantly attenuate the tumor volume and weight, without affecting significant loss in the body weight. Similar to in vitro studies, expression level of c-Met and PI3K/AKT/mTOR, MEK/ERK signals was also suppressed by FCN in the tissues obtained from mice. Therefore, the novel findings of this study suggest that FCN can potentially function as a potent anticancer agent against HCC.
Collapse
Affiliation(s)
- Young Yun Jung
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Jae-Young Um
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
13
|
Phytotherapeutic applications of alkaloids in treating breast cancer. Biomed Pharmacother 2022; 155:113760. [DOI: 10.1016/j.biopha.2022.113760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 09/12/2022] [Accepted: 09/26/2022] [Indexed: 11/23/2022] Open
|
14
|
Benot-Dominguez R, Cimini A, Barone D, Giordano A, Pentimalli F. The Emerging Role of Cyclin-Dependent Kinase Inhibitors in Treating Diet-Induced Obesity: New Opportunities for Breast and Ovarian Cancers? Cancers (Basel) 2022; 14:2709. [PMID: 35681689 PMCID: PMC9179653 DOI: 10.3390/cancers14112709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/26/2022] [Accepted: 05/26/2022] [Indexed: 12/24/2022] Open
Abstract
Overweight and obesity constitute the most impactful lifestyle-dependent risk factors for cancer and have been tightly linked to a higher number of tumor-related deaths nowadays. The excessive accumulation of energy can lead to an imbalance in the level of essential cellular biomolecules that may result in inflammation and cell-cycle dysregulation. Nutritional strategies and phytochemicals are gaining interest in the management of obesity-related cancers, with several ongoing and completed clinical studies that support their effectiveness. At the same time, cyclin-dependent kinases (CDKs) are becoming an important target in breast and ovarian cancer treatment, with various FDA-approved CDK4/6 inhibitors that have recently received more attention for their potential role in diet-induced obesity (DIO). Here we provide an overview of the most recent studies involving nutraceuticals and other dietary strategies affecting cell-cycle pathways, which might impact the management of breast and ovarian cancers, as well as the repurposing of already commercialized chemotherapeutic options to treat DIO.
Collapse
Affiliation(s)
- Reyes Benot-Dominguez
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA; (R.B.-D.); (A.G.)
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Daniela Barone
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, 80131 Napoli, Italy;
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA; (R.B.-D.); (A.G.)
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | | |
Collapse
|
15
|
Jung YY, Chinnathambi A, Alahmadi TA, Alharbi SA, Kumar AP, Sethi G, Ahn KS. Fangchinoline targets epithelial-mesenchymal transition process by modulating activation of multiple cell-signaling pathways. J Cell Biochem 2022; 123:1222-1236. [PMID: 35621239 DOI: 10.1002/jcb.30279] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 12/15/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is a key process, which can promote the transition of tumor cells into other organs by weakening the cell-cell junctions. Tumor cell invasion and metastasis arising because of EMT can determine the prognosis of cancer. EMT can be induced by several growth factors including transforming growth factor-β (TGF-β), which can exert their effects by affecting several cell-signaling pathways. Fangchinoline (FCN), a kind of bisbenzylisoquinoline, belongs to the family Menispermaceae. FCN can display substantial antitumor effects against various malignant cell lines but its possible impact on EMT has not been explored. We examined the potential impact of FCN in affecting the activation of EMT in human colon cancer cells. We evaluated the influence of FCN on EMT in colon cancer cells by using Western blot analysis and reverse transcription-polymerase chain reaction assays. The cellular invasion and migration were observed by Boyden chamber and wound healing assays. Thereafter, the effect of the drug on proliferation and invasion was also evaluated by real-time cell analysis. FCN suppressed the levels of TGF-β-induced mesenchymal markers, such as fibronectin, vimentin, MMP-9, MMP-2, N-cadherin, Twist, and Snail. However, FCN markedly enhanced the expression of epithelial markers such as occludin and E-cadherin. These results imply that FCN can potentially inhibit tumor metastasis through abrogating EMT. In addition, FCN downregulated c-Met/PI3K/Akt/mTOR and Wnt/β-catenin cell signaling pathways and mitigated tumor migration as well as invasion. Overall, our study suggests a potential novel role of FCN as an antimetastatic agent against human colon cancer cells.
Collapse
Affiliation(s)
- Young Y Jung
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Tahani A Alahmadi
- Department of Pediatrics, King Khalid University Hospital [Medical City], King Saud University, Riyadh, Saudi Arabia
| | - Sulaiman A Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Alan P Kumar
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kwang S Ahn
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
16
|
Fangchinoline induces gallbladder cancer cell apoptosis by suppressing PI3K/Akt/XIAP axis. PLoS One 2022; 17:e0266738. [PMID: 35446864 PMCID: PMC9022853 DOI: 10.1371/journal.pone.0266738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/25/2022] [Indexed: 11/19/2022] Open
Abstract
Gallbladder cancer (GBC) is the most common biliary tract malignancy with a dismal prognosis. The development of new drugs may help to improve prognosis. This study found that fangchinoline, a bisbenzylisoquinoline alkaloids, inhibited the proliferation and clone formation of GBC cells in a dose-dependent manner. Moreover, Hoechst staining, TUNEL assays, and flow cytometry demonstrated that fangchinoline effectively induced apoptosis in GBC cells. Further studies found that an anti-apoptotic pathway, the PI3K/Akt/XIAP axis, was significantly inhibited in GBC cells after treating with fangchinoline. Finally, we confirmed that fangchinoline restrained xenograft tumor growth in vivo. Our findings indicate that fangchinoline can be considered a potential drug for GBC treatment.
Collapse
|
17
|
Fangchinoline diminishes STAT3 activation by stimulating oxidative stress and targeting SHP-1 protein in multiple myeloma model. J Adv Res 2022; 35:245-257. [PMID: 35024200 PMCID: PMC8721253 DOI: 10.1016/j.jare.2021.03.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/11/2022] Open
Abstract
Aberrant STAT3 activation can promote neoplastic transformation by affecting cellular proliferation, invasion, metastasis, angiogenesis, and anti-apoptosis induction. Fangchinoline abrogated protein expression levels of STAT3 and upstream signals (JAK1/2 and Src) in different tumor cells. Fangchinoline inhibited the levels of various tumorigenic markers and promoted marked apoptosis through degradation of PARP and caspase-3. Fangchinoline attenuated the level of STAT3 and upstream signals and suppressed the level of anti- apoptotic proteins in xenograft mice model.
Introduction The development of cancer generally occurs as a result of various deregulated molecular mechanisms affecting the genes that can control normal cellular growth. Signal transducer and activator of transcription 3 (STAT3) pathway, once aberrantly activated can promote carcinogenesis by regulating the transcription of a number of oncogenic genes. Objectives Here, we evaluated the impact of fangchinoline (FCN) to attenuate tumor growth and survival through modulation of oncogenic STAT3 signaling pathway using diverse tumor cell lines and a xenograft mouse model. Methods To evaluate the action of FCN on STAT3 cascade, protein levels were analyzed by Western blot analysis and electrophoretic mobility shift assay (EMSA). Translocation of STAT3 was detected by immunocytochemistry. Thereafter, FCN-induced ROS was measured by GSH/GSSG assay and H2DCF-DA. FCN-induced apoptosis was analyzed using Western blot analysis and flow cytometry for various assays. Finally, anti-cancer effects of FCN in vivo was evaluated in a myeloma model. Results We noted that FCN abrogated protein expression levels of STAT3 and upstream signals (JAK1/2 and Src). In addition, FCN also attenuated DNA binding ability of STAT3 and its translocation into the nucleus. It altered the levels of upstream signaling proteins, increased SHP-1 levels, and induced substantial apoptosis in U266 cells. FCN also promoted an increased production of reactive oxygen species (ROS) and altered GSSG/GSH ratio in tumor cells. Moreover, FCN effectively abrogated tumor progression and STAT3 activation in a preclinical myeloma model. Conclusion Overall, this study suggests that FCN may have a tremendous potential to alter abnormal STAT3 activation and induce cell death in malignant cells along with causing the suppression of pathogenesis and growth of cancer through a pro-oxidant dependent molecular mechanism.
Collapse
Key Words
- Apoptosis
- DAPI, 4′,6-Diamidino-2-Phenylindole, Dihydrochloride
- DMEM, Dulbecco’s Modified Eagle Medium
- FBS, Fetal bovine serum
- FCN, Fangchinoline
- Fangchinoline
- GAPDH, Glyceraldehyde 3-phosphate dehydrogenase
- GSH
- HRP, Horseradish peroxidase
- ICC, Immunocytochemistry
- IHC, Immunohistochemistry
- JAK, Janus kinase
- MMP, Matrix metalloproteinase
- Multiple myeloma
- NT, Non treat
- P/S, Penicillin-streptomycin
- PARP, Poly (ADP-ribose) polymerase
- ROS
- RT-PCR, Reverse transcription polymerase chain reaction
- RTCA, Real-time cell analysis
- SHP-1, Src homology 2 domain-containing protein tyrosine phosphatase-1
- STAT3
- STAT3, signal transducer and activator of transcription 3
- VEGF, vascular endothelial growth factor
- c/w, Cell per well
- ip, Intraperitoneal injection
Collapse
|
18
|
Nandi S, Dey R, Dey S, Samadder A, Saxena A. Naturally Sourced CDK Inhibitors and Current Trends in Structure-Based Synthetic Anticancer Drug Design by Crystallography. Anticancer Agents Med Chem 2021; 22:485-498. [PMID: 34503422 DOI: 10.2174/1871520621666210908101751] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 06/12/2021] [Accepted: 07/12/2021] [Indexed: 11/22/2022]
Abstract
Cyclin-dependent kinases (CDKs) are the chief regulators in cell proliferation; the kinase activities are largely regulated by their interactions with CDK inhibitors (CKIs) and Cyclins. The association of different CDKs with CDKIs and Cyclins at the cell-cycle checkpoints of different stages of mitotic cell cycle function act more likely as the molecular switches that regulate different transcriptional events required for progression through the cell cycle. A fine balance in response to extracellular and intracellular signals is highly maintained in the orchestrated function of CDKs along with Cyclins and CDKIs for normal cell proliferation. This fine-tuning in mitotic cell cycle progression sometimes gets lost due to dysregulation of CDKs. The aberrant functioning of the CDKIs is therefore studied for its contributions as a vital hallmark of cancers. It has attracted our focus to maneuver cancer therapy. Hence, several synthetic CDKIs and their crystallography-based drug design have been explained to understand their mode of action with CDKs. Since most of the synthetic drugs function by inhibiting the CDK4/6 kinases by competitively binding to their ATP binding cleft, these synthetic drugs are reported to attack the normal, healthy growing cells adjacent to the cancer cells leading to the decrease in the life span of the cancer patients. The quest for traditional natural medicines may have a great impact on the treatment of cancer. Therefore, in the present studies, a search for naturally sourced CDK inhibitors has been briefly focused. Additionally, some synthetic crystallography-based drug design has been explained to elucidate different avenues to develop better anticancer chemotherapeutics, converting natural scaffolds into inhibitors of the CDK mediated abnormal signal transduction with lesser side effects.
Collapse
Affiliation(s)
- Sisir Nandi
- Global Institute of Pharmaceutical Education and Research (Affiliated to Uttarakhand Technical University), Kashipur-244713. India
| | - Rishita Dey
- Global Institute of Pharmaceutical Education and Research (Affiliated to Uttarakhand Technical University), Kashipur-244713. India
| | - Sudatta Dey
- Cytogenetics and Molecular Biology Lab., Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235. India
| | - Asmita Samadder
- Cytogenetics and Molecular Biology Lab., Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235. India
| | - Anil Saxena
- Global Institute of Pharmaceutical Education and Research (Affiliated to Uttarakhand Technical University), Kashipur-244713. India
| |
Collapse
|
19
|
Xia J, Huang W, Zhou F, Chen Q. Effect of fangchinoline on oxidant status in male albino rats with streptozotocin-induced diabetes. ELECTRON J BIOTECHN 2021. [DOI: 10.1016/j.ejbt.2021.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
20
|
Zhang Y, Wang S, Chen Y, Zhang J, Yang J, Xian J, Li L, Zhao H, Hoffman RM, Zhang Y, Jia L. Fangchinoline Inhibits Human Esophageal Cancer by Transactivating ATF4 to Trigger Both Noxa-Dependent Intrinsic and DR5-Dependent Extrinsic Apoptosis. Front Oncol 2021; 11:666549. [PMID: 34195076 PMCID: PMC8236818 DOI: 10.3389/fonc.2021.666549] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/12/2021] [Indexed: 12/20/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a recalcitrant cancer. The Chinese herbal monomer fangchinoline (FCL) has been reported to have anti-tumor activity in several human cancer cell types. However, the therapeutic efficacy and underlying mechanism on ESCC remain to be elucidated. In the present study, for the first time, we demonstrated that FCL significantly suppressed the growth of ESCC both in vitro and in vivo. Mechanistic studies revealed that FCL-induced G1 phase cell-cycle arrest in ESCC which is dependent on p21 and p27. Moreover, we found that FCL coordinatively triggered Noxa-dependent intrinsic apoptosis and DR5-dependent extrinsic apoptosis by transactivating ATF4, which is a novel mechanism. Our findings elucidated the tumor-suppressive efficacy and mechanisms of FCL and demonstrated FCL is a potential anti-ESCC agent.
Collapse
Affiliation(s)
- Yunjing Zhang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shiwen Wang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Laboratory Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Yukun Chen
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Junqian Zhang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Yang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingrong Xian
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Laboratory Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Lihui Li
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hu Zhao
- Department of Laboratory Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Robert M Hoffman
- Department of Surgery, University of California, San Diego, San Diego, CA, United States.,Anticancer Inc., San Diego, CA, United States
| | - Yanmei Zhang
- Department of Laboratory Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Lijun Jia
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
21
|
Wang WH, Chen SK, Huang HC, Juan HF. Proteomic Analysis Reveals That Metformin Suppresses PSMD2, STIP1, and CAP1 for Preventing Gastric Cancer AGS Cell Proliferation and Migration. ACS OMEGA 2021; 6:14208-14219. [PMID: 34124444 PMCID: PMC8190800 DOI: 10.1021/acsomega.1c00894] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 05/14/2021] [Indexed: 05/04/2023]
Abstract
Metformin is one of the most widely used anti-diabetic drugs in type-II diabetes treatment. The mechanism of decreasing blood glucose is believed to suppress hepatic gluconeogenesis by increasing muscular glucose uptake and insulin sensitivity. Recent studies suggest that metformin may reduce cancer risk; however, its anticancer mechanism in gastric cancers remains unclear. Here, we aim to evaluate the anticancer effects of metformin on human gastric adenocarcinoma (AGS) cells. Our results showed that metformin inhibited AGS cell proliferation in a dose-dependent manner. Using small-scale quantitative proteomics, we identified 177 differentially expressed proteins upon metformin treatment; among these, nine proteins such as 26S proteasome non-ATPase regulatory subunit 2 (PSMD2), stress-induced phosphoprotein 1 (STIP1), and adenylyl cyclase-associated protein 1 (CAP1) were significantly altered. We found that metformin induced cell cycle arrest at the G0/G1 phase, suppressed cell migration, and affected cytoskeleton distribution. Additionally, patients with highly expressed PSMD2, STIP1, and CAP1 have a poor clinical outcome. Our study provides a novel view of developing therapies for gastric cancer.
Collapse
Affiliation(s)
- Wei-Hsuan Wang
- Genome
and Systems Biology Degree Program, Academia
Sinica and National Taiwan University, Taipei 10617, Taiwan
| | - Szu-Kai Chen
- Department
of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Hsuan-Cheng Huang
- Institute
of Biomedical Informatics, National Yang
Ming Chiao Tung University, Taipei 11221, Taiwan
- . Phone: +886-2-2826-7357
| | - Hsueh-Fen Juan
- Genome
and Systems Biology Degree Program, Academia
Sinica and National Taiwan University, Taipei 10617, Taiwan
- Department
of Life Science, National Taiwan University, Taipei 10617, Taiwan
- Graduate
Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 10617, Taiwan
- . Phone: +886-2-3366-4536
| |
Collapse
|
22
|
Velmurugan D, Pachaiappan R, Ramakrishnan C. Recent Trends in Drug Design and Discovery. Curr Top Med Chem 2021; 20:1761-1770. [PMID: 32568020 DOI: 10.2174/1568026620666200622150003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/25/2019] [Accepted: 01/10/2020] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Structure-based drug design is a wide area of identification of selective inhibitors of a target of interest. From the time of the availability of three dimensional structure of the drug targets, mostly the proteins, many computational methods had emerged to address the challenges associated with drug design process. Particularly, drug-likeness, druggability of the target protein, specificity, off-target binding, etc., are the important factors to determine the efficacy of new chemical inhibitors. OBJECTIVE The aim of the present research was to improve the drug design strategies in field of design of novel inhibitors with respect to specific target protein in disease pathology. Recent statistical machine learning methods applied for structural and chemical data analysis had been elaborated in current drug design field. METHODS As the size of the biological data shows a continuous growth, new computational algorithms and analytical methods are being developed with different objectives. It covers a wide area, from protein structure prediction to drug toxicity prediction. Moreover, the computational methods are available to analyze the structural data of varying types and sizes of which, most of the semi-empirical force field and quantum mechanics based molecular modeling methods showed a proven accuracy towards analysing small structural data sets while statistics based methods such as machine learning, QSAR and other specific data analytics methods are robust for large scale data analysis. RESULTS In this present study, the background has been reviewed for new drug lead development with respect specific drug targets of interest. Overall approach of both the extreme methods were also used to demonstrate with the plausible outcome. CONCLUSION In this chapter, we focus on the recent developments in the structure-based drug design using advanced molecular modeling techniques in conjunction with machine learning and other data analytics methods. Natural products based drug discovery is also discussed.
Collapse
Affiliation(s)
- Devadasan Velmurugan
- CAS in Crystallography and Biophysics, University of Madras, Guindy Campus, Chennai - 600025, India
| | - R Pachaiappan
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur - 603203, Kanchipuram District, Tamilnadu, India
| | - Chandrasekaran Ramakrishnan
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology (IIT) Madras, Chennai - 600036, India
| |
Collapse
|
23
|
Xiang X, Tian Y, Hu J, Xiong R, Bautista M, Deng L, Yue Q, Li Y, Kuang W, Li J, Liu K, Yu C, Feng G. Fangchinoline exerts anticancer effects on colorectal cancer by inducing autophagy via regulation AMPK/mTOR/ULK1 pathway. Biochem Pharmacol 2021; 186:114475. [PMID: 33609560 DOI: 10.1016/j.bcp.2021.114475] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 12/24/2022]
Abstract
Autophagy has become a promising target for cancer therapy. Fangchinoline (Fan) has been shown to exert anticancer effects in some types of cancers. However, the anticancer effects on colorectal cancer (CRC) and the underlying mechanisms have never been elucidated. More specifically, regulation of autophagy in CRC by Fan has never been reported before. In the present study, Fan was found to induce apoptosis and autophagic flux in the CRC cell lines HT29 and HCT116, which was reflected by the enhanced levels of LC3-II protein and p62 degradation, and the increased formation of autophagosomes and puncta formation by LC3-II. Meanwhile, combination with the early-stage autophagy inhibitor 3-methyladenine (3-MA) but not the late-stage autophagy inhibitor chloroquine (CQ) further increased Fan-induced cell death, which suggested the cytoprotective function of autophagy induced by Fan in both HT29 and HCT116 cells. Moreover, Fan treatment demonstrated a dose- and time-dependently increase in the phosphorylation of AMPK and decrease in the phosphorylation of mammalian target of rapamycin (mTOR) and ULK1, leading to the activation of the AMPK/mTOR/ULK1 signaling pathway. Furthermore, in the HT29 xenograft model, Fan inhibited tumor growth in vivo. These results indicate that Fan inhibited CRC cell growth both in vitro and in vivo and revealed a new molecular mechanism involved in the anticancer effect of Fan on CRC, suggesting that Fan is a potent autophagy inducer and might be a promising anticancer agent.
Collapse
Affiliation(s)
- Xiaocong Xiang
- Institute of Tissue Engineering and Stem Cells, Cancer Biotherapy Key Laboratory of Nanchong Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong 637000, China
| | - Yunhong Tian
- Department of General Surgery, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong 637000, China
| | - Jiani Hu
- Department of Radiology, Wayne State University, Detroit, MI 48201, USA
| | - Rong Xiong
- Institute of Tissue Engineering and Stem Cells, Cancer Biotherapy Key Laboratory of Nanchong Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong 637000, China
| | - Matthew Bautista
- Department of Radiology, Wayne State University, Detroit, MI 48201, USA
| | - Li Deng
- Institute of Tissue Engineering and Stem Cells, Cancer Biotherapy Key Laboratory of Nanchong Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong 637000, China
| | - Qiuju Yue
- Institute of Tissue Engineering and Stem Cells, Cancer Biotherapy Key Laboratory of Nanchong Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong 637000, China
| | - Yuqi Li
- Institute of Materia Medica, School of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Wei Kuang
- Institute of Materia Medica, School of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Junfeng Li
- Departments of Cardiothoracic Surgery, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong 637000, China
| | - Kang Liu
- Institute of Tissue Engineering and Stem Cells, Cancer Biotherapy Key Laboratory of Nanchong Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong 637000, China
| | - Chunlei Yu
- Institute of Materia Medica, School of Pharmacy, North Sichuan Medical College, Nanchong 637000, China.
| | - Gang Feng
- Institute of Tissue Engineering and Stem Cells, Cancer Biotherapy Key Laboratory of Nanchong Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong 637000, China.
| |
Collapse
|
24
|
Chan EWC, Wong SK, Chan HT. An overview on the chemistry, pharmacology and anticancer properties of tetrandrine and fangchinoline (alkaloids) from Stephania tetrandra roots. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2021; 19:311-316. [PMID: 33583757 DOI: 10.1016/j.joim.2021.01.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 12/07/2020] [Indexed: 01/26/2023]
Abstract
Tetrandrine (TET) and fangchinoline (FAN) are dominant bisbenzylisoquinoline (BBIQ) alkaloids from the roots of Stephania tetrandra of the family Menispermaceae. BBIQ alkaloids comprise two benzylisoquinoline units linked by oxygen bridges. The molecular structures of TET and FAN are exactly the same, except that TET has a methoxy (-OCH3) group, while FAN has a hydroxyl (-OH) group at C7. In this overview, the current knowledge on the chemistry, pharmacology and anticancer properties of TET and FAN have been updated. The focus is on colon and breast cancer cells, because they are most susceptible to TET and FAN, respectively. Against colon cancer cells, TET inhibits cell proliferation and tumor growth by inducing apoptosis and G1 cell cycle arrest, and suppresses adhesion, migration and invasion of cells. Against breast cancer cells, FAN inhibits cell proliferation by inducing apoptosis, G1-phase cell cycle arrest and inhibits cell migration. The processes involve various molecular mechanisms and signaling pathways. Some insights on the ability of TET and FAN to reverse multi-drug resistance in cancer cells and suggestions for future research are provided.
Collapse
Affiliation(s)
| | - Siu Kuin Wong
- School of Foundation Studies, Xiamen University Malaysia, Selangor 43900, Malaysia
| | - Hung Tuck Chan
- Secretariat of the International Society for Mangrove Ecosystems (ISME), Faculty of Agriculture, University of the Ryukyus, Okinawa 903-0129, Japan
| |
Collapse
|
25
|
Wang Q, Tang B, Cao M. Synthesis, characterization, and fungicidal activity of novel Fangchinoline derivatives. Bioorg Med Chem 2020; 28:115778. [DOI: 10.1016/j.bmc.2020.115778] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 01/04/2023]
|
26
|
Oh I, Raymundo B, Jung SA, Kim HJ, Park J, Kim C. Extremely
Low‐Frequency
Electromagnetic Field Altered
PPARγ
and
CCL2
Levels and Suppressed
CD44
+
/
CD24
−
Breast Cancer Cells Characteristics. B KOREAN CHEM SOC 2020. [DOI: 10.1002/bkcs.12072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- In‐Rok Oh
- College of Life Sciences and BiotechnologyKorea University Seoul 136‐701 Korea
| | - Bernardo Raymundo
- College of Life Sciences and BiotechnologyKorea University Seoul 136‐701 Korea
| | - Sung A Jung
- College of Life Sciences and BiotechnologyKorea University Seoul 136‐701 Korea
| | - Hyun Jung Kim
- College of Life Sciences and BiotechnologyKorea University Seoul 136‐701 Korea
| | - Jung‐Keug Park
- Dongguk University Biomedi CampusDongguk University Goyang Korea
| | - Chan‐Wha Kim
- College of Life Sciences and BiotechnologyKorea University Seoul 136‐701 Korea
| |
Collapse
|
27
|
Guo L, Yang T. Oxymatrine Inhibits the Proliferation and Invasion of Breast Cancer Cells via the PI3K Pathway. Cancer Manag Res 2019; 11:10499-10508. [PMID: 31853201 PMCID: PMC6916680 DOI: 10.2147/cmar.s221950] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 11/04/2019] [Indexed: 01/12/2023] Open
Abstract
Purpose Oxymatrine has been reported to possess anti-cancer activity, but its role in breast cancer (BC) is weakly defined. We investigated the anti-cancer effects of oxymatrine in human BC cells, and the underlying molecular mechanisms of these effects. Methods BC lines were treated with oxymatrine. The MTT assay was conducted to evaluate cell viability. The cell cycle and apoptosis of BC cells were analyzed using flow cytometry and Hoechst 33258 staining. Transwell™ assays were undertaken to measure the migratory and invasive abilities of MCF-7 or MDA-MB-231 cells. Expression of phosphatidylinositol 3-kinase (PI3K), Akt, cyclin D1, cluster of differentiation (CD)K2, PARP, Gsk3β, caspase-3, matrix metalloproteinase (MMP)2 and Bax at protein and RNA levels was measured by Western blotting and quantitative real-time polymerase chain reaction. Results Oxymatrine inhibited the proliferation of BC cells in a time-dependent manner. It induced apoptosis in a dose- and time-dependent way according to Annexin V and Hoechst 33258 staining. Oxymatrine could inhibit the invasion of BC cells as shown by the Transwell assay. Oxymatrine inhibited expression of B-cell lymphoma-2 while increasing that of Bax as well as increasing expression of caspase-3 and caspase-9. Addition of oxymatrine to BC cells attenuated the PI3K/Akt signaling pathway cascade, as evidenced by dephosphorylation of P13K and Akt. Conclusion Oxymatrine exerts its anti-tumor effects in BC cells by abolishing the PI3K pathway. Oxymatrine may be a new compound for BC treatment.
Collapse
Affiliation(s)
- Lin Guo
- Department of Gastrointestinal and Nutriology Surgery, Shengjing Hospital of China Medical University, Shenyang, LiaoNing 110004, People's Republic of China
| | - Tengfei Yang
- The Department of Social Service, Shengjing Hospital of China Medical University, Shenyang, LiaoNing 110004, People's Republic of China
| |
Collapse
|
28
|
Niu Y, Xu J, Sun T. Cyclin-Dependent Kinases 4/6 Inhibitors in Breast Cancer: Current Status, Resistance, and Combination Strategies. J Cancer 2019; 10:5504-5517. [PMID: 31632494 PMCID: PMC6775706 DOI: 10.7150/jca.32628] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 08/06/2019] [Indexed: 12/26/2022] Open
Abstract
Dysregulated activation of the cyclin-dependent kinases (CDKs) 4/6, leading to uncontrolled cell division, is hallmark of cancers. Further study of the cell cycle will advance the cancer treatment. As powerful and effective drugs, inhibitors of CDK 4/6 have been widely used in clinical practice for several malignancies, particularly against breast cancers driven by the estrogen receptor (ER). Three CDK4/6 inhibitors, including palbociclib (PD0332991), ribociclib (LEE011) and abemaciclib (LY2835219), have been approved by the US Food and Drug Administration (FDA) for the treatment of hormone receptor-positive, human epidermal growth factor receptor 2-negative advanced or metastatic breast cancer. However, CDK4/6 inhibitors act downstream of many mitogenic signaling pathways, and this has implications for resistance. It is worth to note that the mechanisms of resistance are not very clear. Up to now, a small number of preclinical and clinical studies have explored potential mechanisms of CDK4/6 inhibitors resistance in breast cancer. On this basis, rational and effective combination therapy is under development. Here we review the current knowledge about the mechanisms and efficacy of CDK4/6 inhibitors, and summarize data on resistance mechanisms to make future combination therapies more accurate and reasonable.
Collapse
Affiliation(s)
- Ying Niu
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, No. 44 Xiaoheyan Road, Dadong, Shenyang, Liaoning 110042, P.R. China
| | - Junnan Xu
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, No. 44 Xiaoheyan Road, Dadong, Shenyang, Liaoning 110042, P.R. China.,Key Laboratory of Liaoning Breast Cancer Research, Shenyang, Liaoning 110042, P.R. China
| | - Tao Sun
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, No. 44 Xiaoheyan Road, Dadong, Shenyang, Liaoning 110042, P.R. China
| |
Collapse
|
29
|
Fangchinoline, a Bisbenzylisoquinoline Alkaloid can Modulate Cytokine-Impelled Apoptosis via the Dual Regulation of NF-κB and AP-1 Pathways. Molecules 2019; 24:molecules24173127. [PMID: 31466313 PMCID: PMC6749215 DOI: 10.3390/molecules24173127] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/24/2019] [Accepted: 08/27/2019] [Indexed: 12/25/2022] Open
Abstract
Fangchinoline (FCN) derived from Stephaniae tetrandrine S. Moore can be employed to treat fever, inflammation, rheumatism arthralgia, edema, dysuria, athlete’s foot, and swollen wet sores. FCN can exhibit a plethora of anti-neoplastic effects although its precise mode of action still remains to be deciphered. Nuclear factor-κB (NF-κB) and activator protein-1 (AP-1) can closely regulate carcinogenesis and thus we analyzed the possible action of FCN may have on these two signaling cascades in tumor cells. The effect of FCN on NF-κB and AP-1 signaling cascades and its downstream functions was deciphered using diverse assays in both human chronic myeloid leukemia (KBM5) and multiple myeloma (U266). FCN attenuated growth of both leukemic and multiple myeloma cells and repressed NF-κB, and AP-1 activation through diverse mechanisms, including attenuation of phosphorylation of IκB kinase (IKK) and p65. Furthermore, FCN could also cause significant enhancement in TNFα-driven apoptosis as studied by various molecular techniques. Thus, FCN may exhibit potent anti-neoplastic effects by affecting diverse oncogenic pathways and may be employed as pro-apoptotic agent against various malignancies.
Collapse
|
30
|
Tetrandrine inhibits colon carcinoma HT-29 cells growth via the Bcl-2/Caspase 3/PARP pathway and G1/S phase. Biosci Rep 2019; 39:BSR20182109. [PMID: 31040202 PMCID: PMC6522708 DOI: 10.1042/bsr20182109] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 04/28/2019] [Accepted: 04/30/2019] [Indexed: 12/27/2022] Open
Abstract
Tetrandrine (Tet) bisbenzylisoquinoline alkaloids isolated from Stephania tetrandra and other related species of Menispermaceae. It has been demonstrated to have positive therapeutic effects on cardiovascular disease, hypertension, silicosis, autoimmune diseases. In recent years, some reports have shown that Tet has anticancer activity in human cancers. To explore the pharmacological activity and mechanism of Tet on colon cancer and its unique advantages as a natural product. In the present study, analyses of the cell cycle, apoptosis, targets prediction, molecular docking, and alterations in protein levels were performed to elucidate how Tet functions in colon cancer. We found that Tet robustly induced arrest at the G1 phase in colon cancer cell line HT-29. It induced HT-29 cell apoptosis in a dose-dependent manner. Similarly, analysis of protein expression levels in HT-29 cells showed down-regulation of Bcl-2, pro-caspase 3, pro-caspase 8, PARP, cyclin D1 (CCND1), cyclin-dependent kinase 4 (CDK 4), and up-regulation of Bax, active caspase 3, and active caspase 8. These results indicate that Tet induces apoptosis of colon cancer cells through the mitochondrial pathway and caspase family pathway. Molecular docking showed interaction effects and binding energy. Comparing with the CDK4 inhibitors ribociclib and palbociclib, the docking energy is similar to the docked amino acid residues. Therefore, we conclude that Tet and the CCND1/CDK4 compound could form hydrogen bonds and a stable compound structure, which can inhibit colon cancer cells proliferation by regulating CCND1/CDK4 compound and its downstream proteins phosphorylated Rb (p-Rb). In summary, Tet may be a potential drug for colon cancer therapy.
Collapse
|
31
|
Fangchinoline supplementation attenuates inflammatory markers in experimental rheumatoid arthritis-induced rats. Biomed Pharmacother 2019; 111:142-150. [DOI: 10.1016/j.biopha.2018.12.043] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 11/30/2018] [Accepted: 12/07/2018] [Indexed: 12/13/2022] Open
|
32
|
Wang SQ, Hou HL, Bie LY, Nie CY, Wang LN, Gao S, Hu TT, Chen XB. Mechanistic studies of the apoptosis induced by the macrocyclic natural product tetrandrine in MGC 803 cells. Med Chem Res 2018. [DOI: 10.1007/s00044-018-2268-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
33
|
Molecular Targets Modulated by Fangchinoline in Tumor Cells and Preclinical Models. Molecules 2018; 23:molecules23102538. [PMID: 30301146 PMCID: PMC6222742 DOI: 10.3390/molecules23102538] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 09/29/2018] [Accepted: 10/04/2018] [Indexed: 01/24/2023] Open
Abstract
Despite tremendous progress made during the last few decades in the treatment options for cancer, compounds isolated from Mother Nature remain the mainstay for therapy of various malignancies. Fangchinoline, initially isolated from the dried root of Stephaniae tetrandrine, has been found to exhibit diverse pharmacological effects including significant anticancer activities both in tumor cell lines and selected preclinical models. This alkaloid appears to act by modulating the activation of various important oncogenic molecules involved in tumorigenesis leading to a significant decrease in aberrant proliferation, survival and metastasis of tumor cells. This mini-review briefly describes the potential effects of fangchinoline on important hallmarks of cancer and highlights the molecular targets modulated by this alkaloid in various tumor cell lines and preclinical models.
Collapse
|
34
|
Liu T, Liu X, Li W. Tetrandrine, a Chinese plant-derived alkaloid, is a potential candidate for cancer chemotherapy. Oncotarget 2018; 7:40800-40815. [PMID: 27027348 PMCID: PMC5130046 DOI: 10.18632/oncotarget.8315] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 03/10/2016] [Indexed: 12/19/2022] Open
Abstract
Cancer is a disease caused by the abnormal proliferation and differentiation of cells governed by tumorigenic factors. Chemotherapy is one of the major cancer treatment strategies, and it functions by targeting the physiological capabilities of cancer cells, including sustained proliferation and angiogenesis, the evasion of programmed cell death, tissue invasion and metastasis. Remarkably, natural products have garnered increased attention in the chemotherapy drug discovery field because they are biologically friendly and have high therapeutic effects. Tetrandrine, isolated from the root of Stephania tetrandra S Moore, is a traditional Chinese clinical agent for silicosis, autoimmune disorders, inflammatory pulmonary diseases, cardiovascular diseases and hypertension. Recently, the novel anti-tumor effects of tetrandrine have been widely investigated. More impressive is that tetrandrine affects multiple biological activities of cancer cells, including the inhibition of proliferation, angiogenesis, migration, and invasion; the induction of apoptosis and autophagy; the reversal of multidrug resistance (MDR); and the enhancement of radiation sensitization. This review focuses on introducing the latest information about the anti-tumor effects of tetrandrine on various cancers and its underlying mechanism. Moreover, we discuss the nanoparticle delivery system being developed for tetrandrine and the anti-tumor effects of other bisbenzylisoquinoline alkaloid derivatives on cancer cells. All current evidence demonstrates that tetrandrine is a promising candidate as a cancer chemotherapeutic.
Collapse
Affiliation(s)
- Ting Liu
- College of Life Sciences, Wuhan University, Wuhan, P. R. China
| | - Xin Liu
- Ministry of Education Laboratory of Combinatorial Biosynthesis and Drug Discovery, College of Pharmacy, Wuhan University, Wuhan, P. R. China
| | - Wenhua Li
- College of Life Sciences, Wuhan University, Wuhan, P. R. China
| |
Collapse
|
35
|
Liu Y, Xia B, Lan J, Hu S, Huang L, Chen C, Zeng X, Lou H, Lin C, Pan W. Design, Synthesis and Anticancer Evaluation of Fangchinoline Derivatives. Molecules 2017; 22:E1923. [PMID: 29117113 PMCID: PMC6150242 DOI: 10.3390/molecules22111923] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/01/2017] [Accepted: 11/05/2017] [Indexed: 01/08/2023] Open
Abstract
Twenty fangchinoline derivatives were synthesized from the natural product fangchinoline, and their anticancer activities on human breast cancer MDA-MB-231 cell line, human prostate cancer PC3 cell line, human melanoma WM9 cell line and human leukaemia HEL and K562 cell lines were evaluated. The biological result showed that those derivatives exhibited potent activities on inhibiting cancer cell growth, and the structure-activity relationships were investigated. Among them, compound 4g, which was protected by benzoyl group in 7-phenolic position and nitrified in 14-position, showed impressive inhibition on all 5 cancer cell lines, especially WM9 cell line, with an IC50 value of 1.07 µM. Further mechanistic studies demonstrated that compound 4g may induce cancer cell death by apoptotic means. These research results suggested that compound 4g could be a lead for the further development toward an anticancer agent against human melanoma WM9 in the future.
Collapse
Affiliation(s)
- Yazhou Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, 3491 Baijin Road, Guiyang 550014, China.
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, 3491 Baijin Road, Guiyang 550014, China.
| | - Bin Xia
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, 3491 Baijin Road, Guiyang 550014, China.
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, 3491 Baijin Road, Guiyang 550014, China.
| | - Junjie Lan
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, 3491 Baijin Road, Guiyang 550014, China.
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, 3491 Baijin Road, Guiyang 550014, China.
| | - Shengcao Hu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, 3491 Baijin Road, Guiyang 550014, China.
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, 3491 Baijin Road, Guiyang 550014, China.
| | - Lan Huang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, 3491 Baijin Road, Guiyang 550014, China.
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, 3491 Baijin Road, Guiyang 550014, China.
| | - Chao Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, 3491 Baijin Road, Guiyang 550014, China.
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, 3491 Baijin Road, Guiyang 550014, China.
| | - Xueyi Zeng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, 3491 Baijin Road, Guiyang 550014, China.
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, 3491 Baijin Road, Guiyang 550014, China.
| | - Huayong Lou
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, 3491 Baijin Road, Guiyang 550014, China.
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, 3491 Baijin Road, Guiyang 550014, China.
| | - Changhu Lin
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, 3491 Baijin Road, Guiyang 550014, China.
| | - Weidong Pan
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, 3491 Baijin Road, Guiyang 550014, China.
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, 3491 Baijin Road, Guiyang 550014, China.
| |
Collapse
|
36
|
Shi J, Guo B, Hui Q, Chang P, Tao K. Fangchinoline suppresses growth and metastasis of melanoma cells by inhibiting the phosphorylation of FAK. Oncol Rep 2017; 38:63-70. [PMID: 28560386 PMCID: PMC5492563 DOI: 10.3892/or.2017.5678] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 05/22/2017] [Indexed: 11/07/2022] Open
Abstract
Melanoma is a malignant tumor with high degree of malignancy, metastasis and high mortality. The etiology of melanoma has not been fully elucidated, and there is no effective drug for the complete treatment of melanoma. In recent years, many traditional Chinese herbal medicines have played an important role in clinical treatment and experimental research on cancer. As a natural product, fangchinoline has the characteristics of enhancing immune function, low toxicity and good liver protection features, so it is considered to be a new type of anticancer drug. In the present study, we found that fangchinoline has inhibitory effects on the proliferation and metastasis of A375 and A875 cells in a concentration-dependent manner. Fangchinoline inhibited the proliferation of A375 and A875 cell activity with IC50 values of 12.41 and 16.20 µM. We also found that fangchinoline could significantly reduce the phosphorylation of Focal adhesion kinase (FAK). In summary, we demonstrated that fangchinoline inhibits the proliferation and metastasis of melanoma cells by suppressing FAK and its downstream signaling pathway. More importantly, we provide a novel mechanism that fangchinoline could be an effective candidate for the treatment of melanoma.
Collapse
Affiliation(s)
- Jie Shi
- Department of Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, Shenyang, Liaoning 110840, P.R. China
| | - Bingyu Guo
- Department of Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, Shenyang, Liaoning 110840, P.R. China
| | - Qiang Hui
- Department of Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, Shenyang, Liaoning 110840, P.R. China
| | - Peng Chang
- Department of Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, Shenyang, Liaoning 110840, P.R. China
| | - Kai Tao
- Department of Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, Shenyang, Liaoning 110840, P.R. China
| |
Collapse
|
37
|
Li X, Yang Z, Han W, Lu X, Jin S, Yang W, Li J, He W, Qian Y. Fangchinoline suppresses the proliferation, invasion and tumorigenesis of human osteosarcoma cells through the inhibition of PI3K and downstream signaling pathways. Int J Mol Med 2017; 40:311-318. [PMID: 28586029 PMCID: PMC5504998 DOI: 10.3892/ijmm.2017.3013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 05/23/2017] [Indexed: 12/15/2022] Open
Abstract
Osteosarcoma is the most common malignant bone tumor. Most patients diagnosed with osteosarcoma are less than 20 years of age. Osteosarcoma cells proliferate rapidly and invade other tissues. At present, neoadjuvant chemotherapy is the primary pharmacodynamic strategy to prevent the progression of osteosarcoma. However, adverse effects of this strategy limit its long-term application. Previous research has shown that fangchinoline exerts antitumor effects on several types of tumor cells; however, its effect on osteosarcoma cells remains unknown. The present study evaluated the effects of fangchinoline on the proliferation, apoptosis, migration and invasion of osteosarcoma cells in vitro and on their tumorigenesis in vivo and determined the possible underlying mechanism of action. Fangchinoline-treated MG63 and U20S cells showed significantly decreased proliferation and significantly increased apoptosis. Fangchinoline markedly suppressed the migration and invasion of the MG63 cells. Fangchinoline-treated MG63 cells showed significantly decreased expression of phosphoinositide 3-kinase (PI3K) and Aktp-Thr308. Moreover, fangchinoline-treated MG63 cells showed downregulated expression of cyclin D1 and matrix metalloproteinase 2 and 9, which act downstream of PI3K, and upregulated expression of caspase-3 and caspase-8. Furthermore, fangchinoline suppressed the growth of subcutaneous osteosarcoma tumors in Balb/c mice subcutaneously injected with osteosarcoma cells. These findings suggest that fangchinoline inhibits the progression of osteosarcoma by suppressing the proliferation, migration and invasion and by accelerating the apoptosis of osteosarcoma cells. In addition, our results suggest that the mechanism underlying the antitumor effects of fangchinoline involve the inhibition of PI3K and its downstream signaling pathways.
Collapse
Affiliation(s)
- Xiucheng Li
- Department of Orthopaedics, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, Zhejiang 312000, P.R. China
| | - Zhifan Yang
- Department of Orthopaedics, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, Zhejiang 312000, P.R. China
| | - Weiqi Han
- Department of Orthopaedics, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, Zhejiang 312000, P.R. China
| | - Xuanyuan Lu
- Department of Orthopaedics, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, Zhejiang 312000, P.R. China
| | - Songtao Jin
- Department of Orthopaedics, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, Zhejiang 312000, P.R. China
| | - Wanlei Yang
- Department of Orthopaedics, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, Zhejiang 312000, P.R. China
| | - Jianlei Li
- Department of Orthopaedics, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, Zhejiang 312000, P.R. China
| | - Wei He
- Department of Orthopaedics, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, Zhejiang 312000, P.R. China
| | - Yu Qian
- Department of Orthopaedics, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, Zhejiang 312000, P.R. China
| |
Collapse
|
38
|
Bailon-Moscoso N, Cevallos-Solorzano G, Romero-Benavides JC, Orellana MIR. Natural Compounds as Modulators of Cell Cycle Arrest: Application for Anticancer Chemotherapies. Curr Genomics 2017; 18:106-131. [PMID: 28367072 PMCID: PMC5345333 DOI: 10.2174/1389202917666160808125645] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 11/13/2015] [Accepted: 11/20/2015] [Indexed: 12/22/2022] Open
Abstract
Natural compounds from various plants, microorganisms and marine species play an important role in the discovery novel components that can be successfully used in numerous biomedical applications, including anticancer therapeutics. Since uncontrolled and rapid cell division is a hallmark of cancer, unraveling the molecular mechanisms underlying mitosis is key to understanding how various natural compounds might function as inhibitors of cell cycle progression. A number of natural compounds that inhibit the cell cycle arrest have proven effective for killing cancer cells in vitro, in vivo and in clinical settings. Significant advances that have been recently made in the understanding of molecular mechanisms underlying the cell cycle regulation using the chemotherapeutic agents is of great importance for improving the efficacy of targeted therapeutics and overcoming resistance to anticancer drugs, especially of natural origin, which inhibit the activities of cyclins and cyclin-dependent kinases, as well as other proteins and enzymes involved in proper regulation of cell cycle leading to controlled cell proliferation.
Collapse
|
39
|
Chen Z, He T, Zhao K, Xing C. Anti-metastatic activity of fangchinoline in human gastric cancer AGS cells. Oncol Lett 2016; 13:655-660. [PMID: 28356942 PMCID: PMC5351403 DOI: 10.3892/ol.2016.5457] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 11/10/2016] [Indexed: 12/27/2022] Open
Abstract
Fangchinoline (FCL) is an active component isolated from the traditional medicinal plant Stephania tetrandra S. Moore, and has been reported to possess anti-cancer functions in several types of cancers; however, the effect of FCL on gastric cancer metastasis and its underlying molecular mechanisms remain unknown. The current study aimed to investigate the effect of FCL on the cell migration and invasion of human metastatic gastric cancer AGS cells and its mechanisms. Our study demonstrates that FCL dosage dependently suppressed the adhesion, migration and invasion capacities of human gastric cancer AGS cells without obvious cytotoxic effects. Reverse transcription-polymerase chain reaction and western blot assays demonstrated that FCL greatly inhibited the expression of matrix metalloproteinase (MMP)-2 and MMP-9 at both the mRNA and protein levels, while it significantly increased the expression of tissue inhibitor of metalloproteinase (TIMP) 1 and TIMP2 messenger RNAs. Our results also indicated that FCL repressed the phosphorylation of AKT in gastric cancer AGS cells. In summary, FCL may exert its anti-metastatic property in human gastric cancer cells in vitro by suppression of MMP-2 and MMP-9, increase of TIMP1 and TIMP2 genes, and inhibition of AKT phosphorylation. FCL may be a drug candidate for the treatment of gastric cancer metastasis.
Collapse
Affiliation(s)
- Zhengrong Chen
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215007, P.R. China
| | - Tengfei He
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215007, P.R. China
| | - Kui Zhao
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215007, P.R. China
| | - Chungen Xing
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215007, P.R. China
| |
Collapse
|
40
|
Luo X, Peng JM, Su LDI, Wang DY, Yu YJ. Fangchinoline inhibits the proliferation of SPC-A-1 lung cancer cells by blocking cell cycle progression. Exp Ther Med 2015; 11:613-618. [PMID: 26893655 DOI: 10.3892/etm.2015.2915] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 09/24/2015] [Indexed: 01/19/2023] Open
Abstract
Fangchinoline (Fan) is a bioactive compound isolated from the Chinese herb Stephania tetrandra S. Moore (Fen Fang Ji). The aim of the present study was to investigate the effect of Fan on the proliferation of SPC-A-1 lung cancer cells, and to define the associated molecular mechanisms. Following treatment with Fan, Cell Counting Kit-8, phase contrast imaging and Giemsa staining assays were used to detect cell viability; flow cytometry was performed to analyze the cell cycle distribution; and reverse transcription-quantitative polymerase chain reaction and western blot assays were used to investigate changes in the expression levels of cell cycle-associated genes and proteins. In the present study, treatment with Fan markedly inhibited the proliferation of SPC-A-1 lung cancer cells and significantly increased the percentage of cells in the G0/G1 phase of the cell cycle in a dose-dependent manner (P<0.05 for 2.5-5 µm; P<0.01 for 10 µm), whereas the percentage of cells in the S and G2/M phases were significantly reduced following treatment (P<0.05 for 5 µm; P<0.01 for 10 µm). Mechanistically, Fan significantly reduced the mRNA expression levels of cyclin D1, cyclin-dependent kinase 4 (CDK4) and CDK6 (P<0.05 for 2.5-5 µm; P<0.01 for 10 µm), which are key genes in the regulation of the G0/G1 phase of the cell cycle. Furthermore, treatment with Fan also decreased the expression of phosphorylated retinoblastoma (Rb) and E2F transcription factor-1 (E2F-1) proteins (P<0.05 for 5 µm; P<0.01 for 10 µm). In summary, the present study demonstrated that Fan inhibited the proliferation of SPC-A-1 lung cancer cells and induced cell cycle arrest at the G0/G1 phase. These effects may be mediated by the downregulation of cellular CDK4, CDK6 and cyclin D1 levels, thus leading to hypophosphorylation of Rb and subsequent suppression of E2F-1 activity. Therefore, the present results suggest that Fan may be a potential drug candidate for the prevention of lung cancer.
Collapse
Affiliation(s)
- Xue Luo
- Medical College of Yangzhou Polytechnic College, Yangzhou, Jiangsu 225009, P.R. China
| | - Jian-Ming Peng
- Medical College of Yangzhou Polytechnic College, Yangzhou, Jiangsu 225009, P.R. China; Key Laboratory of Pain, Basic Research and Clinical Therapy, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Lan-DI Su
- Medical College of Yangzhou Polytechnic College, Yangzhou, Jiangsu 225009, P.R. China
| | - Dong-Yan Wang
- Medical College of Yangzhou Polytechnic College, Yangzhou, Jiangsu 225009, P.R. China
| | - You-Jiang Yu
- Medical College of Yangzhou Polytechnic College, Yangzhou, Jiangsu 225009, P.R. China
| |
Collapse
|
41
|
Li D, Lu Y, Sun P, Feng LX, Liu M, Hu LH, Wu WY, Jiang BH, Yang M, Qu XB, Guo DA, Liu X. Inhibition on Proteasome β1 Subunit Might Contribute to the Anti-Cancer Effects of Fangchinoline in Human Prostate Cancer Cells. PLoS One 2015; 10:e0141681. [PMID: 26512898 PMCID: PMC4626104 DOI: 10.1371/journal.pone.0141681] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 10/11/2015] [Indexed: 11/28/2022] Open
Abstract
Fangchinoline is a bisbenzylisoquinoline alkaloid isolated from Radix Stephaniae tetrandrae S. Moore. Fangchinoline and its structure analogue, tetrandrine, exhibited direct binding affinity with recombinant human proteasome β1 subunit and also inhibited its activity in vitro. In cultured prostate PC-3 cells and LnCap cells, fangchinoline could dose-dependently inhibit cell proliferation and caspase-like activity of cellular proteasome which was mediated by proteasome β1 subunit. The inhibitive effect of fangchinoline on caspase-like activity of proteasome was also observed in purified human erythrocyte 20S proteasome. In PC-3 cells, fangchinoline induced cell cycle arrest at G0/G1 phase and apoptosis. Treatment of PC-3 tumor-bearing nude mice with fangchinoline inhibited tumor growth, induced apoptosis and also caused decrease in proteasome activities in tumor xenografts. Dose-dependent and time-dependent accumulation of ubiquitinated proteins and important proteasome substrates such as p27, Bax and IκB-α were observed in fangchinoline-treated cells. Over-expression of proteasome β1 subunit by plasmid transfection increased sensitivity of cells to the cytotoxicity of fangchinoline while knockdown of proteasome β1 subunit ameliorated cytotoxicity of fangchinoline in PC-3 cells. Results of the present study suggested that proteasome inhibition was involved in the anti-cancer effects of fangchinoline. Fangchinoline and its structure analogues might be new natural proteasome inhibitors targeting β1 subunit.
Collapse
Affiliation(s)
- Dong Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
- Changchun University of Chinese Medicine, Changchun 130117, P.R. China
| | - Yu Lu
- Nanjing Tianyi Bioscience Co. Ltd, Nanjing 210061, P.R. China
| | - Peng Sun
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Li-Xing Feng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Miao Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Li-Hong Hu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Wan-Ying Wu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Bao-Hong Jiang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Min Yang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Xiao-Bo Qu
- Changchun University of Chinese Medicine, Changchun 130117, P.R. China
| | - De-An Guo
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
- * E-mail: (DG); (XL)
| | - Xuan Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
- * E-mail: (DG); (XL)
| |
Collapse
|
42
|
Fangchinoline suppresses the growth and invasion of human glioblastoma cells by inhibiting the kinase activity of Akt and Akt-mediated signaling cascades. Tumour Biol 2015; 37:2709-19. [PMID: 26408176 DOI: 10.1007/s13277-015-3990-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 08/25/2015] [Indexed: 12/19/2022] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most palindromic and malignant central nervous system neoplasms, and the current treatment is not effectual for GBM. Research of specific medicine for GBM is significant. Fangchinoline possesses a wide range of pharmacological activities and attracts more attentions due to its anti-tumor effects. In this study, two WHO grade IV human GBM cell lines (U87 MG and U118 MG) were exposed to fangchinoline, and we found that fangchinoline specifically inhibits the kinase activity of Akt and markedly suppresses the phosphorylation of Thr308 and Ser473 of Akt in human GBM cells. We also observed that fangchinoline inhibits tumor cell proliferation and invasiveness and induces apoptosis through suppressing the Akt-mediated signaling cascades, including Akt/p21, Akt/Bad, and Akt/matrix metalloproteinases (MMPs). These data demonstrated that fangchinoline exerts its anti-tumor effects in human glioblastoma cells, at least partly by inhibiting the kinase activity of Akt and suppressing Akt-mediated signaling cascades.
Collapse
|
43
|
Tian F, Ding D, Li D. Fangchinoline targets PI3K and suppresses PI3K/AKT signaling pathway in SGC7901 cells. Int J Oncol 2015; 46:2355-63. [PMID: 25872479 PMCID: PMC4441295 DOI: 10.3892/ijo.2015.2959] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 03/12/2015] [Indexed: 01/17/2023] Open
Abstract
Fangchinoline, an important compound in Stephania tetrandra S. Moore, as a novel antitumor agent, has been implicated in several types of cancers cells except gastric cancer. To investigate whether fangchinoline affects gastric cancer cells, we detected the signaling pathway by which fangchinoline plays a role in different human gastric cancer cells lines. We found that fangchinoline effectively suppressed proliferation and invasion of SGC7901 cell lines, but not MKN45 cell lines by inhibiting the expression of PI3K and its downstream pathway. All of the Akt/MMP2/MMP9 pathway, Akt/Bad pathway, and Akt/Gsk3β/CDK2 pathway could be inhibited by fangchinoline through inhibition of PI3K. Taken together, these results suggest that fangchinoline targets PI3K in tumor cells that express PI3K abundantly and inhibits the growth and invasive ability of the tumor cells.
Collapse
Affiliation(s)
- Feng Tian
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Ding Ding
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Dandan Li
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
44
|
Guo B, Su J, Zhang T, Wang K, Li X. Fangchinoline as a kinase inhibitor targets FAK and suppresses FAK-mediated signaling pathway in A549. J Drug Target 2014; 23:266-74. [PMID: 25539072 DOI: 10.3109/1061186x.2014.992898] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Fangchinoline as a novel anti-tumor agent has been paid attention in several types of cancers cells except lung cancer. Here we have investigated the effect of fangchinoline on A549 cells and its underlying mechanism. PURPOSE The purpose of this work was to study the effect of fangchinoline on A549 cells. METHODS Four lung cancer cell lines (A549, NCI-H292, NCI-H446, and NCI-H460) were exposed to varying concentrations (10-40 μmol/l) of fangchinoline to observe the effect of fangchinoline on the four lung cancer cell lines and to observe the changes of the lung cancer cell on proliferation, apoptosis, and invasion. RESULTS Fangchinoline effectively suppressed proliferation and invasion of A549 cell line but not NCI-H292, NCI-H446, and NCI-H460 cell lines by inhibiting the phosphorylation of FAK (Tyr397) and its downstream pathways, due to the significant differences of Fak expression between A549 and the other three cell lines. And all FAK-paxillin/MMP2/MMP9 pathway, FAK-Akt pathway, and FAK-MEK-ERK1/2 pathway could be inhibited by fangchinoline. DISCUSSION Fangchinoline effectively suppressed proliferation and invasion of A549 cell line by inhibiting the phosphorylation of FAK (Tyr397) and its downstream pathways. CONCLUSION Fangchinoline could inhibit the phosphorylation of FAK(p-Tyr397), at least partially. Fangchinoline as a kinase inhibitor targets FAK and suppresses FAK-mediated signaling pathway and inhibits the growth and the invasion in tumor cells which highly expressed FAK such as A549 cell line.
Collapse
Affiliation(s)
- Bingyu Guo
- Institute of Neurology, General Hospital of Shenyang Military Command , Shenyang, Liaoning , China
| | | | | | | | | |
Collapse
|
45
|
|
46
|
Feng CC, Chen LN, Chen MJ, Li W, Jia X, Zhou YY, He WM. Analysis of different activation statuses of human mammary epithelial cells from young and old groups. Asian Pac J Cancer Prev 2014; 15:3763-6. [PMID: 24870790 DOI: 10.7314/apjcp.2014.15.8.3763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Human mammary epithelial cells have different proliferative statuses and demonstrate a close relationship with age and cell proliferation. Research on this topic could help understand the occurrence, progression and prognosis of breast cancer. In this article, using significance analysis of a microarray algorithm, we analyzed gene expression profiles of human mammary epithelial cells of different proliferative statuses and different age groups. The results showed there were significant differences in gene expression in the same proliferation status between elderly and young groups. Three common differentially expressed genes were found to dynamically change with the proliferation status and to be closely related to tumorigenesis. We also found elderly group had less status-related differential genes from actively proliferating status to intermediate status and more status- related differential genes from intermediate status than the young group. Finally, functional enrichment analyses allowed evaluation of the detailed roles of these differentially-expressed genes in tumor progression.
Collapse
Affiliation(s)
- Chen-Chen Feng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China E-mail :
| | | | | | | | | | | | | |
Collapse
|
47
|
FTY720 for cancer therapy (Review). Oncol Rep 2013; 30:2571-8. [PMID: 24100923 DOI: 10.3892/or.2013.2765] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 09/16/2013] [Indexed: 02/04/2023] Open
Abstract
2-Amino-2-[2-(4-octylphenyl)]-1,3-propanediol hydrochloride (FTY720) is a potent immunosuppressant which has been approved by the Food and Drug Administration (FDA) as a new treatment for multiple sclerosis. As an immunosuppressant, it displays its anti-multiple sclerosis, immunosuppressive effects by activating sphingosine-1-phosphate receptors (S1PRs). In addition to the immunosuppressive effects, FTY720 also shows preclinical antitumor efficacy in several cancer models. In most cases, phosphorylation of FTY720 is not required for its cytotoxic effect, indicating the involvement of S1PR-independent mechanisms which are starkly different from the immunosuppressive property of FTY720. In the present study, we reviewed the rapidly advancing field of FTY720 in cancer therapy as well as some molecular targets of the unphosphorylated form of FTY720.
Collapse
|