1
|
Liu Z, Lu T, Qian R, Wang Z, Qi R, Zhang Z. Exploiting Nanotechnology for Drug Delivery: Advancing the Anti-Cancer Effects of Autophagy-Modulating Compounds in Traditional Chinese Medicine. Int J Nanomedicine 2024; 19:2507-2528. [PMID: 38495752 PMCID: PMC10944250 DOI: 10.2147/ijn.s455407] [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: 12/17/2023] [Accepted: 03/06/2024] [Indexed: 03/19/2024] Open
Abstract
Background Cancer continues to be a prominent issue in the field of medicine, as demonstrated by recent studies emphasizing the significant role of autophagy in the development of cancer. Traditional Chinese Medicine (TCM) provides a variety of anti-tumor agents capable of regulating autophagy. However, the clinical application of autophagy-modulating compounds derived from TCM is impeded by their restricted water solubility and bioavailability. To overcome this challenge, the utilization of nanotechnology has been suggested as a potential solution. Nonetheless, the current body of literature on nanoparticles delivering TCM-derived autophagy-modulating anti-tumor compounds for cancer treatment is limited, lacking comprehensive summaries and detailed descriptions. Methods Up to November 2023, a comprehensive research study was conducted to gather relevant data using a variety of databases, including PubMed, ScienceDirect, Springer Link, Web of Science, and CNKI. The keywords utilized in this investigation included "autophagy", "nanoparticles", "traditional Chinese medicine" and "anticancer". Results This review provides a comprehensive analysis of the potential of nanotechnology in overcoming delivery challenges and enhancing the anti-cancer properties of autophagy-modulating compounds in TCM. The evaluation is based on a synthesis of different classes of autophagy-modulating compounds in TCM, their mechanisms of action in cancer treatment, and their potential benefits as reported in various scholarly sources. The findings indicate that nanotechnology shows potential in enhancing the availability of autophagy-modulating agents in TCM, thereby opening up a plethora of potential therapeutic avenues. Conclusion Nanotechnology has the potential to enhance the anti-tumor efficacy of autophagy-modulating compounds in traditional TCM, through regulation of autophagy.
Collapse
Affiliation(s)
- Zixian Liu
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Tianming Lu
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Ruoning Qian
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Zian Wang
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Ruogu Qi
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Zhengguang Zhang
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| |
Collapse
|
2
|
ZHANG YU, ZHOU XI, ZHANG CHUNYAN, LAI DENGNI, LIU DONGBO, WU YANYANG. Vitamin B3 inhibits apoptosis and promotes autophagy of islet β cells under high glucose stress. BIOCELL 2023. [DOI: 10.32604/biocell.2023.026429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
|
3
|
Evodiamine as an anticancer agent: a comprehensive review on its therapeutic application, pharmacokinetic, toxicity, and metabolism in various cancers. Cell Biol Toxicol 2022; 39:1-31. [PMID: 36138312 DOI: 10.1007/s10565-022-09772-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/07/2022] [Indexed: 11/02/2022]
Abstract
Evodiamine is a major alkaloid component found in the fruit of Evodia rutaecarpa. It shows the anti-proliferative potential against a wide range of cancers by suppressing cell growth, invasion, and metastasis and inducing apoptosis both in vitro and in vivo. Evodiamine shows its anticancer potential by modulating aberrant signaling pathways. Additionally, the review focuses on several therapeutic implications of evodiamine, such as epigenetic modification, cancer stem cells, and epithelial to mesenchymal transition. Moreover, combinatory drug therapeutics along with evodiamine enhances the anticancer efficacy of chemotherapeutic drugs in various cancers by overcoming the chemo resistance and radio resistance shown by cancer cells. It has been widely used in preclinical trials in animal models, exhibiting very negligible side effects against normal cells and effective against cancer cells. The pharmacokinetic and pharmacodynamics-based collaborations of evodiamine are also included. Due to its poor bioavailability, synthetic analogs of evodiamine and its nano capsule have been formulated to enhance its bioavailability and reduce toxicity. In addition, this review summarizes the ongoing research on the mechanisms behind the antitumor potential of evodiamine, which proposes an exciting future for such interests in cancer biology.
Collapse
|
4
|
Yang R, Ma J, Guo H, Meng Q, Wang Y, Yan H, Jin R, Li Z, Meng L. Synthesis and Antitumor Activity of Evodiamine Derivatives With Nitro, Amino, and Methoxy Groups. Nat Prod Commun 2022; 17. [DOI: 10.1177/1934578x211059645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024] Open
Abstract
MS, and IR, 1H NMR and 13C NMR spectroscopy were employed to elucidate 4 novel evodiamine (EVO) derivatives with nitro, amino, and methoxy groups, namely 2-NO2-EVO (7a), 10-OCH3-2-NO2-EVO (7b), 2-NH2-EVO (8a), and 10-OCH3-2-NH2-EVO (8b). The amino compounds (8a, 8b) were obtained by the reduction of nitro derivatives (7a, 7b) with SnCl2/HCl. The antiproliferative activities of these compounds were tested by Cell Counting Kit-8 assay for 48 h against the MDA-MB-231 and sw620 cancer cell lines, as well as the normal LO2 cells. The in vitro experiment showed that 8a possesses the most potent inhibitory activities against MDA-MB-231 and SW620 cells, with IC50 values of 0.79 and 1.28 μM, respectively. The cytotoxicity of 8a against the 2 cancer cell lines was higher than that of EVO.
Collapse
Affiliation(s)
- Ruolan Yang
- Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi, China
| | - Jingjing Ma
- Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi, China
| | - Hui Guo
- Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi, China
- Xi’an Jiao TongUniversity, Xi’an, People’s Republic of China
| | - Qinghua Meng
- Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi, China
| | - Yuwei Wang
- Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi, China
| | - Hao Yan
- Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi, China
| | - Ruyi Jin
- Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi, China
| | - Zhi Li
- Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi, China
| | - Lingjie Meng
- Xi’an Jiao TongUniversity, Xi’an, People’s Republic of China
| |
Collapse
|
5
|
Research Advances in Antitumor Mechanism of Evodiamine. J CHEM-NY 2022. [DOI: 10.1155/2022/2784257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Evodiamine is a natural alkaloid extracted from Fructus Evodia. This bioactive alkaloid has been reported to have a wide range of biological activities, including anti-injury, antiobesity, vasodilator, and anti-inflammatory effects. In recent years, it has been found that evodiamine has tumor-suppressive effects on a variety of tumors. There is growing evidence that evodiamine can inhibit the rapid proliferation of tumor cells, induce cell cycle arrest at a certain phase, increase the incidence of apoptosis, promote autophagy, inhibit microangiogenesis and migration, and regulate immunotherapy. Evodiamine can inhibit Wnt/β-catenin, mTOR, NF-κB, PI3K/AKT, JAK-STAT, and other signaling pathways in various cancer cells, and it can significantly downregulate the expression of many tumor markers, such as VEGF and COX-2. These facts partially explain the antitumor mechanism of evodiamine. In this article, the antitumor mechanism of evodiamine was reviewed to provide the basis for its clinical application and therapeutic development in the future.
Collapse
|
6
|
Li Y, Wang Y, Wang X, Jin L, Yang L, Zhu J, Wang H, Zheng F, Cui H, Li X, Jia Y. Evodiamine suppresses the progression of non-small cell lung carcinoma via endoplasmic reticulum stress-mediated apoptosis pathway in vivo and in vitro. Int J Immunopathol Pharmacol 2022; 36:3946320221086079. [PMID: 35388733 PMCID: PMC9003648 DOI: 10.1177/03946320221086079] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Evodiamine (EVO) is one of the major components isolated from Evodia rutaecarpa (Juss.). Recent studies have shown that EVO has an anti-cancer effect. However, the pharmacological mechanism by which EVO impacts cancer is still poorly understood. OBJECTIVES This study focused on asking the anti-cancer effect of EVO in human non-small cell lung carcinoma (NSCLC), and in particular to investigate whether EVO acts via modulating the endoplasmic reticulum stress (ERS)-mediated apoptosis pathway. MATERIALS AND METHODS A Lewis lung carcinoma (LLC) tumor-bearing mouse model was treated with low-dose EVO (5 mg/kg) and high-dose EVO (10 mg/kg) intraperitoneally for 14 d. The effects of EVO on tumor growth, apoptosis, and ERS were assessed. In addition, NSCLC A549 and LLC cells were treated with EVO in vitro. The effects of EVO on cell proliferation, apoptosis, and ERS were investigated. Finally, 4-phenylbutyric acid (4-PBA), an ERS inhibitor, was used to validate whether EVO induced apoptosis of NSCLC cells by modulating ERS. RESULTS EVO treatment significantly inhibited tumor growth in LLC tumor-bearing mice. H&E staining indicated that EVO treatment reduced the number of tumor cells and the nucleo-plasmic ratio. Immunostaining showed that EVO treatment significantly decreased the expression of Ki-67. TUNEL staining revealed that EVO induced apoptosis in the tumor. Likewise, EVO treatment up-regulated the expression of apoptosis-related genes and proteins and increased activation of the ERS pathway in the tumor. Additionally, EVO inhibited cell proliferation and increased cell apoptotic rates in A549 and LLC cells. EVO also increased the expression levels of genes and proteins associated with ERS-mediated apoptosis pathway in vitro. The effects of EVO on apoptosis were abolished by 4-PBA treatment. CONCLUSIONS Our study demonstrated that EVO suppresses the progression of NSCLC by modulating the ERS-mediated apoptosis pathway.
Collapse
Affiliation(s)
- Yuting Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuming Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoqun Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lulu Jin
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lu Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinli Zhu
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hongwu Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Fang Zheng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huantian Cui
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Xiaojiang Li
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingjie Jia
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
7
|
Luo C, Ai J, Ren E, Li J, Feng C, Li X, Luo X. Research progress on evodiamine, a bioactive alkaloid of Evodiae fructus: Focus on its anti-cancer activity and bioavailability (Review). Exp Ther Med 2021; 22:1327. [PMID: 34630681 DOI: 10.3892/etm.2021.10762] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 08/17/2021] [Indexed: 12/24/2022] Open
Abstract
Evodiae fructus (Wu-Zhu-Yu in Chinese) can be isolated from the dried, unripe fruits of Tetradium ruticarpum and is a well-known traditional Chinese medicine that is applied extensively in China, Japan and Korea. Evodiae fructus has been traditionally used to treat headaches, abdominal pain and menorrhalgia. In addition, it is widely used as a dietary supplement to provide carboxylic acids, essential oils and flavonoids. Evodiamine (EVO) is one of the major bioactive components contained within Evodiae fructus and is considered to be a potential candidate anti-cancer agent. EVO has been reported to exert anti-cancer effects by inhibiting cell proliferation, invasion and metastasis, whilst inducing apoptosis in numerous types of cancer cells. However, EVO is susceptible to metabolism and may inhibit the activities of metabolizing enzymes, such as cytochrome P450. Clinical application of EVO in the treatment of cancers may prove difficult due to poor bioavailability and potential toxicity due to metabolism. Currently, novel drug carriers involving the use of solid dispersion techniques, phospholipids and nanocomplexes to deliver EVO to improve its bioavailability and mitigate side effects have been tested. The present review aims to summarize the reported anti-cancer effects of EVO whilst discussing the pharmacokinetic behaviors, characteristics and effective delivery systems of EVO.
Collapse
Affiliation(s)
- Chaodan Luo
- Subtropical Agricultural Products Processing Engineering Technology Center, Guangxi Institute of Subtropical Agricultural Products Processing, Guangxi Subtropical Crops Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, Guangxi 530001, P.R. China
| | - Jingwen Ai
- Subtropical Agricultural Products Processing Engineering Technology Center, Guangxi Institute of Subtropical Agricultural Products Processing, Guangxi Subtropical Crops Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, Guangxi 530001, P.R. China
| | - Erfang Ren
- Subtropical Agricultural Products Processing Engineering Technology Center, Guangxi Institute of Subtropical Agricultural Products Processing, Guangxi Subtropical Crops Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, Guangxi 530001, P.R. China
| | - Jianqiang Li
- Subtropical Agricultural Products Processing Engineering Technology Center, Guangxi Institute of Subtropical Agricultural Products Processing, Guangxi Subtropical Crops Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, Guangxi 530001, P.R. China
| | - Chunmei Feng
- Subtropical Agricultural Products Processing Engineering Technology Center, Guangxi Institute of Subtropical Agricultural Products Processing, Guangxi Subtropical Crops Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, Guangxi 530001, P.R. China
| | - Xinrong Li
- Subtropical Agricultural Products Processing Engineering Technology Center, Guangxi Institute of Subtropical Agricultural Products Processing, Guangxi Subtropical Crops Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, Guangxi 530001, P.R. China
| | - Xiaojie Luo
- Subtropical Agricultural Products Processing Engineering Technology Center, Guangxi Institute of Subtropical Agricultural Products Processing, Guangxi Subtropical Crops Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, Guangxi 530001, P.R. China
| |
Collapse
|
8
|
Sun Q, Xie L, Song J, Li X. Evodiamine: A review of its pharmacology, toxicity, pharmacokinetics and preparation researches. JOURNAL OF ETHNOPHARMACOLOGY 2020; 262:113164. [PMID: 32738391 DOI: 10.1016/j.jep.2020.113164] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/17/2020] [Accepted: 07/06/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Evodia rutaecarpa, a well-known herb medicine in China, is extensively applied in traditional Chinese medicine (TCM). The plant has the effects of dispersing cold and relieving pain, arresting vomiting, and helping Yang and stopping diarrhea. Modern research demonstrates that evodiamine, the main component of Evodia rutaecarpa, is the material basis for its efficacy. AIMS OF THE REVIEW This paper is primarily addressed to summarize the current studies on evodiamine. The progress in research on the pharmacology, toxicology, pharmacokinetics, preparation researches and clinical application are reviewed. Moreover, outlooks and directions for possible future studies concerning it are also discussed. MATERIALS AND METHODS The information of this systematic review was conducted with resources of multiple literature databases including PubMed, Google scholar, Web of Science and Wiley Online Library and so on, with employing a combination of keywords including "pharmacology", "toxicology", "pharmacokinetics" and "clinical application", etc. RESULTS: As the main component of Evodia rutaecarpa, evodiamine shows considerable pharmacological activities, such as analgesic, anti-inflammatory, anti-tumor, anti-microbial, heart protection and metabolic disease regulation. However, it is also found that it has significant hepatotoxicity and cardiotoxicity, thereby it should be monitored in clinical. In addition, available data demonstrate that the evodiamine has a needy solubility in aqueous medium. Scientific and reasonable pharmaceutical strategies should be introduced to improve the above defects. Meanwhile, more efforts should be made to develop novel efficient and low toxic derivatives. CONCLUSIONS This review summarizes the results from current studies of evodiamine, which is one of the valuable medicinal ingredients from Evodia rutaecarpa. With the assistance of relevant pharmacological investigation, some conventional application and problems in pharmaceutical field have been researched in recent years. In addition, unresolved issues include toxic mechanisms, pharmacokinetics, novel pharmaceutical researches and relationship between residues and intestinal environment, which are still being explored and excavate before achieving integration into clinical practice.
Collapse
Affiliation(s)
- Qiang Sun
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Long Xie
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Jiawen Song
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Xiaofang Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China.
| |
Collapse
|
9
|
Hong Z, Wang Z, Zhou B, Wang J, Tong H, Liao Y, Zheng P, Jamshed MB, Zhang Q, Chen H. Effects of evodiamine on PI3K/Akt and MAPK/ERK signaling pathways in pancreatic cancer cells. Int J Oncol 2020; 56:783-793. [PMID: 31922213 PMCID: PMC7010218 DOI: 10.3892/ijo.2020.4956] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 07/30/2019] [Indexed: 02/07/2023] Open
Abstract
The effective antitumor drug evodiamine (EVO) is attracting increased attention. Therefore, the present study aimed to investigate the effects of EVO on the proliferation, apoptosis and autophagy of human pancreatic cancer (PC) cell lines in vitro and in vivo. Human PANC-1 and SW1990 PC cell lines were treated with different concentrations of EVO and proliferation was detected using a Cell Counting Kit (CCK)-8 assay. Colony formation and wound-healing assays showed that EVO inhibited PC cell viability and migration, and apoptosis was detected using flow cytometry. Western blotting and immunofluorescence detected the expression of proteins in PANC-1 and SW1990 cells. The PANC-1 cells were used to establish an orthotopic pancreatic tumor model in nude mice. Tumor-bearing nude mice were administered with different concentrations of EVO, and growth was monitored. High-resolution positron emission tomography and fluorine-18-labeled fluorodeoxyglucose were used to monitor the tumor/non-tumor (T/NT) ratio and standard uptake value (SUV) of the mice, which were subsequently sacrificed to measure the transplanted tumor weight. Apoptosis increased with increasing EVO concentration. The EVO-treated PC cells exhibited significantly higher expression of LC3II than the controls cells. EVO decreased LC3II, enhanced P62 and inhibited the expression of Akt, extracellular-signal-regulated protein kinase (ERK)1/2 and p38. Compared with the control group, the T/NT ratio, SUV and tumor weight decreased more markedly in the EVO-treated group. The tumor expression of phosphorylated AKT, detected using immunohistochemistry, decreased with increasing EVO doses in vivo. EVO induced PC cell apoptosis by inhibiting phosphoinositide 3-kinase/AKT and mitogen-activated protein kinase/ERK and inhibiting the phosphorylation of signal transducer and activator of transcription activator 3 in PC cells to inhibit autophagy, suggesting that EVO may be considered as a novel PC treatment.
Collapse
Affiliation(s)
- Zhong Hong
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Zhaohong Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Bin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Jisheng Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Hongfei Tong
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Yi Liao
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Peng Zheng
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Muhammad Babar Jamshed
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Qiyu Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Hui Chen
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| |
Collapse
|
10
|
Efferth T, Oesch F. Repurposing of plant alkaloids for cancer therapy: Pharmacology and toxicology. Semin Cancer Biol 2019; 68:143-163. [PMID: 31883912 DOI: 10.1016/j.semcancer.2019.12.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/15/2019] [Indexed: 02/08/2023]
Abstract
Drug repurposing (or repositioning) is an emerging concept to use old drugs for new treatment indications. Phytochemicals isolated from medicinal plants have been largely neglected in this context, although their pharmacological activities have been well investigated in the past, and they may have considerable potentials for repositioning. A grand number of plant alkaloids inhibit syngeneic or xenograft tumor growth in vivo. Molecular modes of action in cancer cells include induction of cell cycle arrest, intrinsic and extrinsic apoptosis, autophagy, inhibition of angiogenesis and glycolysis, stress and anti-inflammatory responses, regulation of immune functions, cellular differentiation, and inhibition of invasion and metastasis. Numerous underlying signaling processes are affected by plant alkaloids. Furthermore, plant alkaloids suppress carcinogenesis, indicating chemopreventive properties. Some plant alkaloids reveal toxicities such as hepato-, nephro- or genotoxicity, which disqualifies them for repositioning purposes. Others even protect from hepatotoxicity or cardiotoxicity of xenobiotics and established anticancer drugs. The present survey of the published literature clearly demonstrates that plant alkaloids have the potential for repositioning in cancer therapy. Exploitation of the chemical diversity of natural alkaloids may enrich the candidate pool of compounds for cancer chemotherapy and -prevention. Their further preclinical and clinical development should follow the same stringent rules as for any other synthetic drug as well. Prospective randomized, placebo-controlled clinical phase I and II trials should be initiated to unravel the full potential of plant alkaloids for drug repositioning.
Collapse
Affiliation(s)
- Thomas Efferth
- Department of Pharmaceutical Biology, Johannes Gutenberg University, Mainz, Germany.
| | - Franz Oesch
- Institute of Toxicology, Medical Center, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
11
|
Antiproliferative Effects of Alkaloid Evodiamine and Its Derivatives. Int J Mol Sci 2018; 19:ijms19113403. [PMID: 30380774 PMCID: PMC6274956 DOI: 10.3390/ijms19113403] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 10/20/2018] [Accepted: 10/24/2018] [Indexed: 12/18/2022] Open
Abstract
Alkaloids, a category of natural products with ring structures and nitrogen atoms, include most U.S. Food and Drug Administration approved plant derived anti-cancer agents. Evodiamine is an alkaloid with attractive multitargeting antiproliferative activity. Its high content in the natural source ensures its adequate supply on the market and guarantees further medicinal study. To the best of our knowledge, there is no systematic review about the antiproliferative effects of evodiamine derivatives. Therefore, in this article the review of the antiproliferative activities of evodiamine will be updated. More importantly, the antiproliferative activities of structurally modified new analogues of evodiamine will be summarized for the first time.
Collapse
|
12
|
Wu Y, Hu Y, Yuan Y, Luo Y, lai D, Zhou H, Tong Z, Liu D. Gymnemic acid I triggers mechanistic target of rapamycin‐mediated β cells cytoprotection through the promotion of autophagy under high glucose stress. J Cell Physiol 2018; 234:9370-9377. [DOI: 10.1002/jcp.27621] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 09/24/2018] [Indexed: 12/30/2022]
Affiliation(s)
- Yanyang Wu
- Hunan Agricultural University Changsha China
- Horticulture and Landscape College, Hunan Agricultural University Changsha China
- College of Food Science and Technology, Hunan Agricultural University Changsha China
| | - Yongquan Hu
- Horticulture and Landscape College, Hunan Agricultural University Changsha China
- Hunan Co‐Innovation Center for Utilization of Botanical Functional Ingredients Changsha China
- State Key Laboratory of Subhealth Intervention Technology Changsha China
| | - Yuju Yuan
- Hunan Agricultural University Changsha China
- Horticulture and Landscape College, Hunan Agricultural University Changsha China
- College of Food Science and Technology, Hunan Agricultural University Changsha China
| | - Yushuang Luo
- Horticulture and Landscape College, Hunan Agricultural University Changsha China
- Hunan Co‐Innovation Center for Utilization of Botanical Functional Ingredients Changsha China
- State Key Laboratory of Subhealth Intervention Technology Changsha China
| | - Dengni lai
- Hunan Agricultural University Changsha China
- Horticulture and Landscape College, Hunan Agricultural University Changsha China
- College of Food Science and Technology, Hunan Agricultural University Changsha China
| | - Haiyan Zhou
- Horticulture and Landscape College, Hunan Agricultural University Changsha China
- Hunan Co‐Innovation Center for Utilization of Botanical Functional Ingredients Changsha China
- State Key Laboratory of Subhealth Intervention Technology Changsha China
| | - Zhongyi Tong
- Department of Pathology The Second Xiangya Hospital of Central South University Changsha China
| | - Dongbo Liu
- Horticulture and Landscape College, Hunan Agricultural University Changsha China
- Hunan Co‐Innovation Center for Utilization of Botanical Functional Ingredients Changsha China
- State Key Laboratory of Subhealth Intervention Technology Changsha China
| |
Collapse
|
13
|
Moosavi MA, Haghi A, Rahmati M, Taniguchi H, Mocan A, Echeverría J, Gupta VK, Tzvetkov NT, Atanasov AG. Phytochemicals as potent modulators of autophagy for cancer therapy. Cancer Lett 2018; 424:46-69. [PMID: 29474859 DOI: 10.1016/j.canlet.2018.02.030] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 02/18/2018] [Accepted: 02/19/2018] [Indexed: 02/07/2023]
Abstract
The dysregulation of autophagy is involved in the pathogenesis of a broad range of diseases, and accordingly universal research efforts have focused on exploring novel compounds with autophagy-modulating properties. While a number of synthetic autophagy modulators have been identified as promising cancer therapy candidates, autophagy-modulating phytochemicals have also attracted attention as potential treatments with minimal side effects. In this review, we firstly highlight the importance of autophagy and its relevance in the pathogenesis and treatment of cancer. Subsequently, we present the data on common phytochemicals and their mechanism of action as autophagy modulators. Finally, we discuss the challenges associated with harnessing the autophagic potential of phytochemicals for cancer therapy.
Collapse
Affiliation(s)
- Mohammad Amin Moosavi
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, P.O Box:14965/161, Tehran, Iran.
| | - Atousa Haghi
- Young Researchers & Elite Club, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Marveh Rahmati
- Cancer Biology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hiroaki Taniguchi
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, 05-552 Jastrzebiec, Poland
| | - Andrei Mocan
- Department of Pharmaceutical Botany, "Iuliu Haţieganu" University of Medicine and Pharmacy, Gheorghe Marinescu 23 Street, 400337 Cluj-Napoca, Romania
| | - Javier Echeverría
- Facultad de Química y Biología, Universidad de Santiago de Chile, Casilla 40, Correo 33, Santiago 9170022, Chile
| | - Vijai K Gupta
- Department of Chemistry and Biotechnology, ERA Chair of Green Chemistry, Tallinn University of Technology, 12618 Tallinn, Estonia
| | - Nikolay T Tzvetkov
- Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany; NTZ Lab Ltd., Krasno Selo 198, Sofia 1618, Bulgaria
| | - Atanas G Atanasov
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, 05-552 Jastrzebiec, Poland; Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria.
| |
Collapse
|
14
|
Shin D, Kim EH, Lee J, Roh JL. RITA plus 3-MA overcomes chemoresistance of head and neck cancer cells via dual inhibition of autophagy and antioxidant systems. Redox Biol 2017; 13:219-227. [PMID: 28582730 PMCID: PMC5925444 DOI: 10.1016/j.redox.2017.05.025] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 05/28/2017] [Accepted: 05/30/2017] [Indexed: 12/25/2022] Open
Abstract
Reactivation of p53 and induction of tumor cell apoptosis (RITA) is a small molecule that blocks p53–MDM2 interaction, thereby reactivating p53 in tumors. RITA can induce exclusive apoptosis in cancer cells independently of the p53 pathway; however, the resistance of cancer cells remains a major drawback. Here, we found a novel resistance mechanism of RITA treatment and an effective combined treatment to overcome RITA resistance in head and neck cancer (HNC) cells. The effects of RITA and 3-methyladenine (3-MA) were tested in different HNC cell lines, including cisplatin-resistant and acquired RITA-resistant HNC cells. The effects of each drug alone and in combination were assessed by measuring cell viability, apoptosis, cell cycle, glutathione, reactive oxygen species, protein expression, genetic inhibition of p62 and Nrf2, and a mouse xenograft model of cisplatin-resistant HNC. RITA induced apoptosis of HNC cells at different levels without significantly inhibiting normal cell viability. Following RITA treatment, RITA-resistant HNC cells exhibited a sustained expression of other autophagy-related proteins, overexpressed p62, and displayed activation of the Keap1-Nrf2 antioxidant pathway. The autophagy inhibitor 3-MA sensitized resistant HNC cells to RITA treatment via the dual inhibition of molecules related to the autophagy and antioxidant systems. Silencing of the p62 gene augmented the combined effects. The effective antitumor activity of RITA plus 3-MA was also confirmed in vivo in mouse xenograft models transplanted with resistant HNC cells, showing increased oxidative stress and DNA damage. The results indicate that RITA plus 3-MA can help overcome RITA resistance in HNC cells. Condensed abstract This study revealed a novel RITA resistant mechanism associated with the sustained induction of autophagy, p62 overexpression, and Keap1-Nrf2 antioxidant system activation. The combined treatment of RITA with the autophagy inhibitor 3-methyladenine overcomes RITA resistance via dual inhibition of autophagy and antioxidant systems in vitro and in vivo. RITA induces apoptosis of HNC cells at different levels. RITA resistance is related to the sustained expression of autophagy proteins and p62. Keap1-Nrf2 antioxidant system is also engaged in the RITA resistance mechanism. The autophagy inhibitor 3-MA sensitizes resistant HNC cells to RITA treatment. RITA plus 3-MA induces apoptosis of resistant HNC cells via dual inhibition of autophagy and Nrf2 system in vitro and in vivo.
Collapse
Affiliation(s)
- Daiha Shin
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eun Hye Kim
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jaewang Lee
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jong-Lyel Roh
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Li J, Yang D, Wang W, Piao S, Zhou J, Saiyin W, Zheng C, Sun H, Li Y. Inhibition of autophagy by 3-MA enhances IL-24-induced apoptosis in human oral squamous cell carcinoma cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:97. [PMID: 26361755 PMCID: PMC4567787 DOI: 10.1186/s13046-015-0211-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 08/25/2015] [Indexed: 12/21/2022]
Abstract
Background Interleukin-24(IL-24), also referred to as melanoma differentiation-associated gene-7(mda-7), is a unique member of the IL-10 gene family, which displays nearly ubiquitous cancer-specific toxicity. The most notable feature of IL-24 is selectively induced growth suppression and apoptosis in various cancer cells, with no harmful effects toward normal cells. Autophagy is a self-protective mechanism in many kinds of tumor cells that respond to anticancer treatment. It is reported that autophagy inhibition could enhance the effects of many kinds of anticancer treatments, including gene therapy. However, whether IL-24 is effective to treat oral squamous cell carcinomas (OSCC) and if autophagy inhibition could improve the anticancer effect of IL-24 towards OSCC is has not been detected. Methods MTT assays were carried out to determine the cell proliferation; Transfection was used to gene transfer; Western Blot was performed to detect the protein level of LC3II, P62, Beclin 1, Cleaved caspase-3, β-Tubulin and β-actin; Apoptosis rates and cell cycle alteration were analyzed using flow cytometry; Autophagy induction was confirmed by MDC staining, GFP-LC3 staining and transmission electron microscopy. Amount of IL-24 in the culture medium was quantified by ELISA. Apoptosis in vivo was analyzed by TUNEL assay. HE staining was used to observe the morphology of the samples. Results In the present study, we proved that IL-24 have a novel anticancer effect towards KB cells and that autophagy inhibition could improve the anticancer effect of IL-24. IL-24 treated cells showed autophagy characteristics and autophagy inhibition by 3-methyladenine (3-MA) significantly enhanced IL-24-induced apoptosis. Similar results were obtained in the KB cells xenograft tumor model. Conclusions These results suggest that the combination of autophagy inhibitors and IL-24 based on the AdLTR2EF1α-mediated gene transfer could be a promising way to cure OSCC.
Collapse
Affiliation(s)
- Jichen Li
- School of Life Science and Technology, Harbin Institute of Technology, 2 Yikuang Street, Harbin, 150001, People's Republic of China.
| | - Dezhao Yang
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Harbin Medical University, 141 Yiman Street, Nangang District, Harbin, 150001, People's Republic of China.
| | - Wei Wang
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Harbin Medical University, 141 Yiman Street, Nangang District, Harbin, 150001, People's Republic of China.
| | - Songlin Piao
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Harbin Medical University, 141 Yiman Street, Nangang District, Harbin, 150001, People's Republic of China.
| | - Jianyu Zhou
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Harbin Medical University, 141 Yiman Street, Nangang District, Harbin, 150001, People's Republic of China.
| | - Wuliji Saiyin
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Harbin Medical University, 141 Yiman Street, Nangang District, Harbin, 150001, People's Republic of China.
| | - Changyu Zheng
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA.
| | - Hongchen Sun
- Department of Oral Pathology, School of Stomatology, Jilin University, 1500 Qinghua Road, Changchun, 130021, People's Republic of China.
| | - Yu Li
- School of Life Science and Technology, Harbin Institute of Technology, 2 Yikuang Street, Harbin, 150001, People's Republic of China.
| |
Collapse
|
16
|
KHAN MUHAMMAD, BI YANYING, QAZI JAVEDIQBAL, FAN LIMEI, GAO HONGWEN. Evodiamine sensitizes U87 glioblastoma cells to TRAIL via the death receptor pathway. Mol Med Rep 2014; 11:257-62. [DOI: 10.3892/mmr.2014.2705] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 05/02/2014] [Indexed: 11/05/2022] Open
|
17
|
De Petrocellis L, Schiano Moriello A, Fontana G, Sacchetti A, Passarella D, Appendino G, Di Marzo V. Effect of chirality and lipophilicity in the functional activity of evodiamine and its analogues at TRPV1 channels. Br J Pharmacol 2014; 171:2608-20. [PMID: 23902373 PMCID: PMC4009003 DOI: 10.1111/bph.12320] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 07/18/2013] [Accepted: 07/26/2013] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND AND PURPOSE Evodiamine, a racemic quinazolinocarboline alkaloid isolated from the traditional Chinese medicine Evodiae fructus, has been reported to act as an agonist of the transient receptor potential vanilloid type-1 (TRPV1) cation channel both in vitro and in vivo. Evodiamine is structurally different from all known TRPV1 activators, and has significant clinical potential as a thermogenic agent. Nevertheless, the molecular bases for its actions are still poorly understood. EXPERIMENTAL APPROACH To investigate the structure-activity relationships of evodiamine, the natural racemate was resolved, and a series of 23 synthetic analogues was prepared, using as the end point the intracellular Ca(2+) elevation in HEK-293 cells stably overexpressing either the human or the rat recombinant TRPV1. KEY RESULTS S-(+) evodiamine was more efficacious and potent than R-(-) evodiamine, and a new potent lead (Evo30) was identified, more potent than the reference TRPV1 agonist, capsaicin. In general, potency and efficacy correlated with the lipophilicity of the analogues. Like other TRPV1 agonists, several synthetic analogues could efficiently desensitize TRPV1 to activation by capsaicin. CONCLUSIONS AND IMPLICATIONS Evodiamine qualifies as structurally unique lead structure to develop new potent TRPV1 agonists/desensitizers.
Collapse
Affiliation(s)
- Luciano De Petrocellis
- Istituto di Cibernetica, Endocannabinoid Research Group, Consiglio Nazionale delle RicerchePozzuoli, Italy
| | - Aniello Schiano Moriello
- Istituto di Chimica Biomolecolare, Endocannabinoid Research Group, Consiglio Nazionale delle RicerchePozzuoli, Italy
| | | | | | - Daniele Passarella
- Dipartimento di Chimica, Materiali ed Ingegneria Chimica ‘G. Natta’, Politecnico di MilanoMilano, Italy
| | - Giovanni Appendino
- Dipartimento di Scienze Chimiche, Alimentari, Farmaceutiche e Farmacologiche, Università del Piemonte OrientaleNovara, Italy
| | - Vincenzo Di Marzo
- Istituto di Chimica Biomolecolare, Endocannabinoid Research Group, Consiglio Nazionale delle RicerchePozzuoli, Italy
| |
Collapse
|
18
|
Saiyin W, Wang D, Li L, Zhu L, Liu B, Sheng L, Li Y, Zhu B, Mao L, Li G, Zhu X. Sequential release of autophagy inhibitor and chemotherapeutic drug with polymeric delivery system for oral squamous cell carcinoma therapy. Mol Pharm 2014; 11:1662-75. [PMID: 24666011 DOI: 10.1021/mp5000423] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Autophagy inhibition is emerging as a new paradigm for efficient cancer therapy by overcoming multidrug resistance (MDR). Here, we developed an effective chemotherapeutic system for oral squamous cell carcinoma (OSCC) based on polymeric nanomicelles for codelivery of the anticancer drug doxorubicin (DOX) and the autophagy inhibitor LY294002 (LY). The hydrophobic DOX was conjugated onto a hydrophilic and pH-responsive hyperbranched polyacylhydrazone (HPAH), forming the DOX-conjugated HPAH (HPAH-DOX). Due to its amphiphilicity, HPAH-DOX self-assembled into nanomicelles in an aqueous solution and the autophagy inhibitor LY could be loaded into the HPAH-DOX micelles. The release of DOX and LY from the LY-loaded HPAH-DOX micelles was pH-dependent, whereas LY was released significantly faster than DOX at a mildly acidic condition. The in vitro evaluation demonstrated that the LY-loaded HPAH-DOX micelles could rapidly enter cancer cells and then release LY and DOX in response to an intracellular acidic environment. Compared to the HPAH-DOX micelles and the physical mixture of HPAH-DOX and LY, the LY-loaded HPAH-DOX micelles induced a higher proliferation inhibition of tumor cells, illustrating a synergistic effect of LY and DOX. The preferentially released LY inhibited the autophagy of tumor cells and made them more sensitive to the subsequent liberation of DOX. The polymeric codelivery system for programmable release of the chemotherapy drug and the autophagy inhibitor provides a new platform for combination of traditional chemotherapy and autophagy inhibition.
Collapse
Affiliation(s)
- Wuliji Saiyin
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Harbin Medical University , 141 Yiman Street, Nangang District, Harbin 150001, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Christodoulou MS, Sacchetti A, Ronchetti V, Caufin S, Silvani A, Lesma G, Fontana G, Minicone F, Riva B, Ventura M, Lahtela-Kakkonen M, Jarho E, Zuco V, Zunino F, Martinet N, Dapiaggi F, Pieraccini S, Sironi M, Dalla Via L, Gia OM, Passarella D. Quinazolinecarboline alkaloid evodiamine as scaffold for targeting topoisomerase I and sirtuins. Bioorg Med Chem 2013; 21:6920-8. [PMID: 24103429 DOI: 10.1016/j.bmc.2013.09.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 09/09/2013] [Accepted: 09/11/2013] [Indexed: 12/12/2022]
Abstract
This paper reports the synthesis of a series of evodiamine derivatives. We assayed the ability to inhibit cell growth on three human tumour cell lines (H460, MCF-7 and HepG2) and we evaluated the capacity to interfere with the catalytic activity of topoisomerase I both by the relaxation assay and the occurrence of the cleavable complex. Moreover, whose effect on sirtuins 1, 2 and 3 was investigated. Finally, molecular docking analyses were performed in an attempt to rationalize the biological results.
Collapse
Affiliation(s)
- Michael S Christodoulou
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi 19, 20133 Milano, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|