1
|
Luo M, Shang L, Xie J, Zhou T, He C, Fisher D, Pronyuk K, Musabaev E, Hien NTT, Wang H, Zhao L. Current status and trend of global research on the pharmacological effects of emodin family: bibliometric study and visual analysis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03758-5. [PMID: 39792164 DOI: 10.1007/s00210-024-03758-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025]
Abstract
Emodin, as a natural active ingredient, has shown great application potential in the fields of medicine, food and cosmetics due to its unique pharmacological effects, such as anti-inflammatory, antioxidant, anti-cancer, etc. In recent years, with the development of science and technology and the increase of people's demand for natural medicine, emodin research has been paid more and more attention by the global scientific research community. The bibliometric analysis of emodin and the construction of knowledge map are still blank. We searched the publications of emodin related studies in the Web of Science Core Collection (WoSCC) database from 2004 to 2024 and conducted a bibliometric analysis. Data processing was done using the R packages Bibliometrix, VOSviewer and CiteSpace. The consensus identified 4,125 emodin related articles from multiple countries, with China being the main contributor. The number of publications in this field is increasing year by year. China Medical University, the Chinese Academy of Sciences, and Nanjing University of Traditional Chinese Medicine are all prominent research institutions in this field. The Journal of ethnopharmacology published the most articles on the subject. The total number of authors of these articles has reached 14,991, among which Yi Wang is the author with the most output and Xiaoxv Dong is the author with the most cited times. "emodin", "apoptosis", and "liver injury" were the main research focuses. Topics such as "pharmacology", "photodynamic therapy", "advancing drug discovery" and "gallbladder cancer cell" may represent emerging areas of research in medicine. The results of this study help to identify the latest research frontiers and hot topics, and provide a valuable reference for the study of emodin family.
Collapse
Affiliation(s)
- Miao Luo
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Luorui Shang
- Department of Clinical Nutrition, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiao Xie
- Health Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tao Zhou
- Department of Gastroenterology and Hepatology, Huanggang Hospital of Traditional Chinese Medicine, Huanggang, Hubei, China
| | - Chengyi He
- Department of Vascular Surgery, the Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - David Fisher
- Department of Medical Biosciences, Faculty of Natural Sciences, University of The Western Cape, Cape Town, South Africa
| | - Khrystyna Pronyuk
- Infectious Diseases Department, O.Bogomolets National Medical University, Kiev, Ukraine
| | - Erkin Musabaev
- The Research Institute of Virology, Ministry of Health, Tashkent, Uzbekistan
| | | | - Huan Wang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Lei Zhao
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
2
|
Varlı M, Ji M, Kim E, Kim SJ, Choi B, Ha HH, Kim KK, Paik MJ, Kim H. Emodin disrupts the KITENIN oncogenic complex by binding ErbB4 and suppresses colorectal cancer progression in dual blockade with KSRP-binding compound. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156247. [PMID: 39586126 DOI: 10.1016/j.phymed.2024.156247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 10/22/2024] [Accepted: 11/09/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND The KITENIN/ErbB4 complex has been reported to participate in metastasis, which is the principal reason of death in most colorectal cancer patients. PURPOSE New therapeutics need to be developed to suppress the malignant effects of the KITENIN/ErbB4 complex, which is related to drug resistance. The present study aimed to evaluate changes in cancer cell invasion capacity, transcriptional regulators, and cellular bioenergetics after targeting the KITENIN/ErbB4 complex with emodin. Moreover, we aimed to reveal the mechanistic effects of emodin and observe the dual blockade effects of ErbB4-targeted therapy with KH-type splicing regulatory protein (KSRP) and search for new alternative blockade pathways. METHODS Using in vitro, in vivo, molecular-docking, and metabolomics studies, we evaluated the anticancer effect of emodin alone or in combination with DKCC14S. RESULTS Emodin treatment decreased KITENIN and ErbB4 protein levels. The dysfunctional KITENIN/ErbB4 complex suppressed KITENIN-mediated cell invasion and downregulated AP-1 activity, aerobic glycolysis, and the levels of transcriptional regulators associated with cell metabolism. We conclude that emodin targets the KITENIN/ErbB4 complex and offering a novel mechanism by which it disrupts KITENIN-mediated signaling. Furthermore, we were demonstrated that the dual blocking effect of emodin and DKC-C14S on the KITENIN complex showed synergistic effects in suppressing colorectal cancer progression under in cell-based and animal assay. CONCLUSION The results suggest that co-treatment with ErbB4 and KSRP-binding compounds could constitute a potential strategy for controlling colorectal cancer progression by disrupting the KITENIN complex.
Collapse
Affiliation(s)
- Mücahit Varlı
- College of Pharmacy, Sunchon National University, Sunchon 57922, Republic of Korea.
| | - Moongi Ji
- College of Pharmacy, Sunchon National University, Sunchon 57922, Republic of Korea.
| | - Eunae Kim
- College of Pharmacy, Chosun University, 146 Chosundae-gil, Gwangju 61452, Republic of Korea.
| | - Sung Jin Kim
- Department of Pharmacology, Chonnam National University Medical School, 160 Baekseoro, Dong-gu, Gwangju, 61469, Republic of Korea.
| | - Byeongchan Choi
- College of Pharmacy, Sunchon National University, Sunchon 57922, Republic of Korea.
| | - Hyung-Ho Ha
- College of Pharmacy, Sunchon National University, Sunchon 57922, Republic of Korea.
| | - Kyung Keun Kim
- Department of Pharmacology, Chonnam National University Medical School, 160 Baekseoro, Dong-gu, Gwangju, 61469, Republic of Korea.
| | - Man-Jeong Paik
- College of Pharmacy, Sunchon National University, Sunchon 57922, Republic of Korea.
| | - Hangun Kim
- College of Pharmacy, Sunchon National University, Sunchon 57922, Republic of Korea.
| |
Collapse
|
3
|
Wu CC, Chen MS, Chen JY. The Application of Emodin Treatment on Nasopharyngeal Carcinoma Therapy. Biomedicines 2024; 12:486. [PMID: 38540100 PMCID: PMC10967729 DOI: 10.3390/biomedicines12030486] [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: 12/30/2023] [Revised: 02/02/2024] [Accepted: 02/08/2024] [Indexed: 01/03/2025] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a malignancy prevailing in Taiwan, Hong Kong, Southern China, Southeast Asia, and North Africa. Although early-stage NPC responds well to the primary treatment of radio-chemotherapy, the mortality rate of advanced NPC remains high. Therefore, developing new therapies for nasopharyngeal carcinoma is an urgent task. Emodin is an anthraquinone derivative mainly found in Rheum palmatum. Emodin has been found to possess many anti-cancer functions against various types of cancers, but they are less discussed in the treatment of NPC. This review organized the different studies about the anti-NPC activity of emodin and discussed the potential and challenges of emodin treatment in NPC therapy.
Collapse
Affiliation(s)
- Chung-Chun Wu
- Translational Cell Therapy Center, Department of Medical Research, China Medical University Hospital, Taichung City 404447, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Zhunan 350401, Taiwan;
| | - Mei-Shu Chen
- National Institute of Cancer Research, National Health Research Institutes, Zhunan 350401, Taiwan;
| | - Jen-Yang Chen
- National Institute of Cancer Research, National Health Research Institutes, Zhunan 350401, Taiwan;
| |
Collapse
|
4
|
Zarezadeh SM, Sharafi AM, Erabi G, Tabashiri A, Teymouri N, Mehrabi H, Golzan SA, Faridzadeh A, Abdollahifar Z, Sami N, Arabpour J, Rahimi Z, Ansari A, Abbasi MR, Azizi N, Tamimi A, Poudineh M, Deravi N. Natural STAT3 Inhibitors for Cancer Treatment: A Comprehensive Literature Review. Recent Pat Anticancer Drug Discov 2024; 19:403-502. [PMID: 37534488 DOI: 10.2174/1574892818666230803100554] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 06/05/2023] [Accepted: 06/09/2023] [Indexed: 08/04/2023]
Abstract
Cancer is one of the leading causes of mortality and morbidity worldwide, affecting millions of people physically and financially every year. Over time, many anticancer treatments have been proposed and studied, including synthetic compound consumption, surgical procedures, or grueling chemotherapy. Although these treatments have improved the daily life quality of patients and increased their survival rate and life expectancy, they have also shown significant drawbacks, including staggering costs, multiple side effects, and difficulty in compliance and adherence to treatment. Therefore, natural compounds have been considered a possible key to overcoming these problems in recent years, and thorough research has been done to assess their effectiveness. In these studies, scientists have discovered a meaningful interaction between several natural materials and signal transducer and activator of transcription 3 molecules. STAT3 is a transcriptional protein that is vital for cell growth and survival. Mechanistic studies have established that activated STAT3 can increase cancer cell proliferation and invasion while reducing anticancer immunity. Thus, inhibiting STAT3 signaling by natural compounds has become one of the favorite research topics and an attractive target for developing novel cancer treatments. In the present article, we intend to comprehensively review the latest knowledge about the effects of various organic compounds on inhibiting the STAT3 signaling pathway to cure different cancer diseases.
Collapse
Affiliation(s)
- Seyed Mahdi Zarezadeh
- Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Mohammad Sharafi
- Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gisou Erabi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Arefeh Tabashiri
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Navid Teymouri
- Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hoda Mehrabi
- Student Research Committee, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Seyyed Amirhossein Golzan
- Student Research Committee, Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arezoo Faridzadeh
- Department of Immunology and Allergy, Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Abdollahifar
- Student Research Committee, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Nafiseh Sami
- Student Research Committee, Tehran Medical Sciences, Islamic Azad University Medical Branch of Tehran, Tehran, Iran
| | - Javad Arabpour
- Department of Microbiology, Faculty of New Sciences, Islamic Azad University Medical Branch of Tehran, Tehran, Iran
| | - Zahra Rahimi
- School of Medicine, Zanjan University of Medical Sciences Zanjan, Iran
| | - Arina Ansari
- Student Research Committee, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | | | - Nima Azizi
- Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Niloofar Deravi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Yu L, Zhao Y, Zhao Y. Advances in the pharmacological effects and molecular mechanisms of emodin in the treatment of metabolic diseases. Front Pharmacol 2023; 14:1240820. [PMID: 38027005 PMCID: PMC10644045 DOI: 10.3389/fphar.2023.1240820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 10/04/2023] [Indexed: 12/01/2023] Open
Abstract
Rhubarb palmatum L., Polygonum multijiorum Thunb., and Polygonum cuspidatum Sieb. Et Zucc. are traditional Chinese medicines that have been used for thousands of years. They are formulated into various preparations and are widely used. Emodin is a traditional Chinese medicine monomer and the main active ingredient in Rhubarb palmatum L., Polygonum multijiorum Thunb., and Polygonum cuspidatum Sieb. Et Zucc. Modern research shows that it has a variety of pharmacological effects, including promoting lipid and glucose metabolism, osteogenesis, and anti-inflammatory and anti-autophagy effects. Research on the toxicity and pharmacokinetics of emodin can promote its clinical application. This review aims to provide a basis for further development and clinical research of emodin in the treatment of metabolic diseases. We performed a comprehensive summary of the pharmacology and molecular mechanisms of emodin in treating metabolic diseases by searching databases such as Web of Science, PubMed, ScienceDirect, and CNKI up to 2023. In addition, this review also analyzes the toxicity and pharmacokinetics of emodin. The results show that emodin mainly regulates AMPK, PPAR, and inflammation-related signaling pathways, and has a good therapeutic effect on obesity, hyperlipidemia, non-alcoholic fatty liver disease, diabetes and its complications, and osteoporosis. In addition, controlling toxic factors and improving bioavailability are of great significance for its clinical application.
Collapse
Affiliation(s)
- Linyuan Yu
- Department of Traditional Chinese Medicine, Chengdu Integrated TCM and Western Medicine Hospital, Chengdu, China
- Department of Pharmacy, Sichuan Second Hospital of TCM, Chengdu, China
| | - Yongliang Zhao
- Nursing Department, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yongli Zhao
- Department of Traditional Chinese Medicine, Chengdu Integrated TCM and Western Medicine Hospital, Chengdu, China
| |
Collapse
|
6
|
LI X, DUAN Z, YUE J, ZHANG Y, LI Y, LIU S, NIE Q, YANG D, ZHANG L. Bornyl acetate extracted from Sharen () inhibits proliferation, invasion and induces apoptosis by suppressing phosphatidylinositol-3-kinase/protein kinase B signaling in colorectal cancer. J TRADIT CHIN MED 2023; 43:1081-1091. [PMID: 37946470 PMCID: PMC10623251 DOI: 10.19852/j.cnki.jtcm.20231018.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 11/15/2022] [Indexed: 11/12/2023]
Abstract
OBJECTIVE To investigate the antitumor effects of bornyl acetate (BA) isolated from Sharen (Fructus Amomi) in colorectal cancer (CRC) and the underlying mechanisms. METHODS SW480 and HT29 cells were treated with increasing doses of BA in order to determine its antitumor effects in vitro. Cell viability, colony formation, cell cycle, and apoptosis as well as migration and invasion were assessed using various assays. In addition, the in vivo antitumor effects of BA were assessed using a xenograft mouse model. We then assessed the mechanism of action of BA by conducting pathway activator-mediated rescue experiments and assessed the protein levels by Western blot analysis. RESULTS BA showed anti-CRC tumor activities in vitro by suppressing cell proliferation and colony formation, inducing apoptosis, blocking cell cycle, and inhibiting migration and invasion. These effects were mediated via suppression of the phosphatidylinositol-3-kinase/protein kinase B (PI3K/AKT) pathway. In the tumor xenograft experiment, BA was found to repress tumor growth in vivo with low toxicity. CONCLUSIONS The results demonstrated that BA exerts antitumor effects by suppressing the PI3K/AKT pathway, with low toxicity. Thus, BA might be a potential novel therapeutic agent for CRC.
Collapse
Affiliation(s)
- Xiaohua LI
- 1 School of Traditional Dai-Thai Medicine, West Yunnan University of Apllied Science, Jinghong 666100, China
| | - Zhihang DUAN
- 1 School of Traditional Dai-Thai Medicine, West Yunnan University of Apllied Science, Jinghong 666100, China
| | - Jianjun YUE
- 1 School of Traditional Dai-Thai Medicine, West Yunnan University of Apllied Science, Jinghong 666100, China
| | - Yongyu ZHANG
- 1 School of Traditional Dai-Thai Medicine, West Yunnan University of Apllied Science, Jinghong 666100, China
| | - Yihang LI
- 2 Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Jinghong 666100, China
| | - Shifang LIU
- 2 Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Jinghong 666100, China
| | - Qu NIE
- 1 School of Traditional Dai-Thai Medicine, West Yunnan University of Apllied Science, Jinghong 666100, China
| | - Depo YANG
- 2 Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Jinghong 666100, China
| | - Lixia ZHANG
- 3 Yunnan Branch, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Yunnan Key Laboratory of Southern Medicine Utilization, Jinghong 666100, China
| |
Collapse
|
7
|
Lu H, Xie D, Qu B, Li M, He Y, Liu W. Emodin prevents renal ischemia-reperfusion injury via suppression of p53-mediated cell apoptosis based on network pharmacology. Heliyon 2023; 9:e15682. [PMID: 37215853 PMCID: PMC10195913 DOI: 10.1016/j.heliyon.2023.e15682] [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: 08/31/2022] [Revised: 04/16/2023] [Accepted: 04/18/2023] [Indexed: 05/24/2023] Open
Abstract
Background Previous evidence indicated that emodin has significant advantages for preventing acute kidney injury (AKI). However, the mechanisms responsible for these effects of emodin have yet to be elucidated. Methods We first used network pharmacology and molecular docking to identify the core targets of emodin for AKI and performed a range of experiments to validate this result. Pretreatment with emodin for 7 days, the rats were treated with bilateral renal artery clipping for 45 min to identify the prevention effect. Hypoxia/reoxygenation (H/R), and vancomycin - induced renal tubular epithelial cells (HK-2 cells) were treated with emodin to explore the related molecular mechanism. Results Network pharmacology and molecular docking showed that anti-apoptosis might be the core mechanism responsible for the action of emodin on AKI; this anti-apoptotic effect appears to because by regulation p53-related signaling pathway. Our data showed that pretreatment with emodin significantly improved renal function and renal tubular injury in renal I/R model rats (P < 0.05. The prevention effect of emodin was proved to be related to anti - apoptosis of HK-2 cells, possibly by downregulating the levels of p53, cleaved-caspase-3, pro-caspase-9, and upregulated the levels of Bcl-2. The efficacy and mechanism of emodin on anti - apoptosis was also confirmed in vancomycin - induced HK-2 cells. Meanwhile, the data also showed that emodin promoted angiogenesis in I/R damaged kidneys and H/R-induced HK-2 cells, which was associated with decreasing HIF-1α levels and increasing VEGF levels. Conclusions Our findings indicated that the preventive effect of emodin on AKI is probably attributable to anti-apoptosis response and promoting angiogenesis effect.
Collapse
Affiliation(s)
- Hongmei Lu
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100700, China
- Department of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Beijing, 100700, China
| | - Dengpiao Xie
- Department of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, China
| | - Bo Qu
- Department of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, China
| | - Mingquan Li
- Department of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, China
| | - Yuhua He
- Department of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, China
| | - Weijing Liu
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100700, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Beijing, 100700, China
| |
Collapse
|
8
|
Kim YJ, Song J, Lee DH, Um SH, Bhang SH. Suppressing cancer by damaging cancer cell DNA using LED irradiation. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2023; 243:112714. [PMID: 37084656 DOI: 10.1016/j.jphotobiol.2023.112714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 04/09/2023] [Accepted: 04/13/2023] [Indexed: 04/23/2023]
Abstract
BACKGROUND High-energy irradiation eliminates cancer cells by destroying their genetic components. However, there are several side effects from doing this, such as fatigue, dermatitis, and hair loss, which remain obstacles to this treatment. Here, we propose a moderate method that uses low-energy white light from a light-emitting diode (LED) to selectively inhibit cancer cell proliferation without affecting normal cells. METHODS The association between LED irradiation and cancer cell growth arrest was evaluated based on cell proliferation, viability, and apoptotic activity. Immunofluorescence, polymerase chain reaction, and western blotting were performed in vitro and in vivo to identify the metabolism related to the inhibition of HeLa cell proliferation. RESULTS LED irradiation aggravated the defective p53 signaling pathway and induced cell growth arrest in cancer cells. Consequently, cancer cell apoptosis was induced by the increased DNA damage. Additionally, LED irradiation inhibited the proliferation of cancer cells by suppressing the MAPK pathway. Furthermore, the suppression of cancer growth by the regulation of p53 and MAPK was observed in cancer-bearing mice irradiated with LED. CONCLUSIONS Our findings suggest that LED irradiation can suppress cancer cell activity and may contribute to preventing the proliferation of cancer cells after medical surgery without causing side effects.
Collapse
Affiliation(s)
- Yu-Jin Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea; Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Jihun Song
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Dong-Hyun Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Soong Ho Um
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
9
|
Current Trends in Toxicity Assessment of Herbal Medicines: A Narrative Review. Processes (Basel) 2022. [DOI: 10.3390/pr11010083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Even in modern times, the popularity level of medicinal plants and herbal medicines in therapy is still high. The World Health Organization estimates that 80% of the population in developing countries uses these types of remedies. Even though herbal medicine products are usually perceived as low risk, their potential health risks should be carefully assessed. Several factors can cause the toxicity of herbal medicine products: plant components or metabolites with a toxic potential, adulteration, environmental pollutants (heavy metals, pesticides), or contamination of microorganisms (toxigenic fungi). Their correct evaluation is essential for the patient’s safety. The toxicity assessment of herbal medicine combines in vitro and in vivo methods, but in the past decades, several new techniques emerged besides conventional methods. The use of omics has become a valuable research tool for prediction and toxicity evaluation, while DNA sequencing can be used successfully to detect contaminants and adulteration. The use of invertebrate models (Danio renio or Galleria mellonella) became popular due to the ethical issues associated with vertebrate models. The aim of the present article is to provide an overview of the current trends and methods used to investigate the toxic potential of herbal medicinal products and the challenges in this research field.
Collapse
|
10
|
Ahmad W, Ansari MA, Alsayari A, Almaghaslah D, Wahab S, Alomary MN, Jamal QMS, Khan FA, Ali A, Alam P, Elderdery AY. In Vitro, Molecular Docking and In Silico ADME/Tox Studies of Emodin and Chrysophanol against Human Colorectal and Cervical Carcinoma. Pharmaceuticals (Basel) 2022; 15:1348. [PMID: 36355520 PMCID: PMC9697597 DOI: 10.3390/ph15111348] [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: 10/02/2022] [Revised: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 08/11/2023] Open
Abstract
Anthraquinones (AQs) are present in foods, dietary supplements, pharmaceuticals, and traditional treatments and have a wide spectrum of pharmacological activities. In the search for anti-cancer drugs, AQ derivatives are an important class. In this study, anthraquinone aglycons chrysophanol (Chr), emodin (EM) and FDA-approved anticancer drug fluorouracil were analyzed by molecular docking studies against receptor molecules caspase-3, apoptosis regulator Bcl-2, TRAF2 and NCK-interacting protein kinase (TNIK) and cyclin-dependent protein kinase 2 (CDK2) as novel candidates for future anticancer therapeutic development. The ADMET SAR database was used to predict the toxicity profile and pharmacokinetics of the Chr and EM. Furthermore, in silico results were validated by the in vitro anticancer activity against HCT-116 and HeLa cell lines to determine the anticancer effect. According to the docking studies simulated by the docking program AutoDock Vina 4.0, Chr and EM had good binding energies against the target proteins. It has been observed that Chr and EM show stronger molecular interaction than that of the FDA-approved anticancer drug fluorouracil. In the in vitro results, Chr and EM demonstrated promising anticancer activity in HCT-116 and HeLa cells. These findings lay the groundwork for the potential use of Chr and EM in the treatment of human colorectal and cervical carcinomas.
Collapse
Affiliation(s)
- Wasim Ahmad
- Department of Pharmacy, Mohammed Al-Mana College for Medical Sciences, Dammam 34222, Saudi Arabia
| | - Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Abdulrhman Alsayari
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
- Complementary and Alternative Medicine Unit, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Dalia Almaghaslah
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
- Complementary and Alternative Medicine Unit, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Mohammad N. Alomary
- National Centre for Biotechnology, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Qazi Mohammad Sajid Jamal
- Department of Health Informatics, College of Public Health and Health Informatics, Qassim University, Al Bukayriyah 52741, Saudi Arabia
| | - Firdos Alam Khan
- Department of Stem Cell Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Abuzer Ali
- Department of Pharmacognosy, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia
| | - Prawez Alam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11941, Saudi Arabia
| | - Abozer Y. Elderdery
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia
| |
Collapse
|
11
|
Talib WH, Awajan D, Hamed RA, Azzam AO, Mahmod AI, AL-Yasari IH. Combination Anticancer Therapies Using Selected Phytochemicals. Molecules 2022; 27:5452. [PMID: 36080219 PMCID: PMC9458090 DOI: 10.3390/molecules27175452] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer is still one of the most widespread diseases globally, it is considered a vital health challenge worldwide and one of the main barriers to long life expectancy. Due to the potential toxicity and lack of selectivity of conventional chemotherapeutic agents, discovering alternative treatments is a top priority. Plant-derived natural products have high potential in cancer treatment due to their multiple mechanisms of action, diversity in structure, availability in nature, and relatively low toxicity. In this review, the anticancer mechanisms of the most common phytochemicals were analyzed. Furthermore, a detailed discussion of the anticancer effect of combinations consisting of natural product or natural products with chemotherapeutic drugs was provided. This review should provide a strong platform for researchers and clinicians to improve basic and clinical research in the development of alternative anticancer medicines.
Collapse
Affiliation(s)
- Wamidh H. Talib
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan
| | - Dima Awajan
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan
| | - Reem Ali Hamed
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan
| | - Aya O. Azzam
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan
| | - Asma Ismail Mahmod
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan
| | - Intisar Hadi AL-Yasari
- Department of Genetic Engineering, College of Biotechnology, Al-Qasim Green University, Babylon 964, Iraq
| |
Collapse
|
12
|
Talib WH, Daoud S, Mahmod AI, Hamed RA, Awajan D, Abuarab SF, Odeh LH, Khater S, Al Kury LT. Plants as a Source of Anticancer Agents: From Bench to Bedside. Molecules 2022; 27:molecules27154818. [PMID: 35956766 PMCID: PMC9369847 DOI: 10.3390/molecules27154818] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 12/03/2022] Open
Abstract
Cancer is the second leading cause of death after cardiovascular diseases. Conventional anticancer therapies are associated with lack of selectivity and serious side effects. Cancer hallmarks are biological capabilities acquired by cancer cells during neoplastic transformation. Targeting multiple cancer hallmarks is a promising strategy to treat cancer. The diversity in chemical structure and the relatively low toxicity make plant-derived natural products a promising source for the development of new and more effective anticancer therapies that have the capacity to target multiple hallmarks in cancer. In this review, we discussed the anticancer activities of ten natural products extracted from plants. The majority of these products inhibit cancer by targeting multiple cancer hallmarks, and many of these chemicals have reached clinical applications. Studies discussed in this review provide a solid ground for researchers and physicians to design more effective combination anticancer therapies using plant-derived natural products.
Collapse
Affiliation(s)
- Wamidh H. Talib
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931, Jordan; (A.I.M.); (R.A.H.); (D.A.); (S.F.A.); (L.H.O.); (S.K.)
- Correspondence:
| | - Safa Daoud
- Department Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmacy, Applied Science Private University, Amman 11931, Jordan;
| | - Asma Ismail Mahmod
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931, Jordan; (A.I.M.); (R.A.H.); (D.A.); (S.F.A.); (L.H.O.); (S.K.)
| | - Reem Ali Hamed
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931, Jordan; (A.I.M.); (R.A.H.); (D.A.); (S.F.A.); (L.H.O.); (S.K.)
| | - Dima Awajan
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931, Jordan; (A.I.M.); (R.A.H.); (D.A.); (S.F.A.); (L.H.O.); (S.K.)
| | - Sara Feras Abuarab
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931, Jordan; (A.I.M.); (R.A.H.); (D.A.); (S.F.A.); (L.H.O.); (S.K.)
| | - Lena Hisham Odeh
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931, Jordan; (A.I.M.); (R.A.H.); (D.A.); (S.F.A.); (L.H.O.); (S.K.)
| | - Samar Khater
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931, Jordan; (A.I.M.); (R.A.H.); (D.A.); (S.F.A.); (L.H.O.); (S.K.)
| | - Lina T. Al Kury
- Department of Health Sciences, College of Natural and Health Sciences, Zayed University, Abu Dhabi 144534, United Arab Emirates;
| |
Collapse
|
13
|
Dai G, Wang D, Ma S, Hong S, Ding K, Tan X, Ju W. ACSL4 promotes colorectal cancer and is a potential therapeutic target of emodin. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 102:154149. [PMID: 35567995 DOI: 10.1016/j.phymed.2022.154149] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 04/11/2022] [Accepted: 05/02/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is an important death-related disease in the world and new therapeutic strategies are urgently needed to reduce mortality. Several studies have demonstrated that emodin, the main ingredient of Rheum palmatum, fights cancer but its potential anti-tumor effect on CRC is still unknown. PURPOSE The present study is aimed to explore the potential anti-tumor effects of emodin against CRC and the underlying molecular mechanism. METHODS CRC-related datasets were screened according to filter criteria in the GEO database and TCGA database. By using screened differentially expressed genes, GO, KEGG and survival analysis were carried out. The expressions of ACSL4, VEGFR1, and VEGFR2 were examined by immunohistochemistry and western blot. Then, pcDNA-ACSL4, pcDNA-VEGFR1, and pcDNA-VEGFR2 were used to overexpress ACSL4, VEGFR1, and VEGFR2, while ACSL4 siRNA was used to silence ACSL4 expression in HCT116 cells. CCK-8 assay and transwell migration assay were used to detect the cell proliferation and invasion. A docking simulation assay and an MST assay were performed to explore the potential mode of emodin binding to ACSL4. The HCT116 cells and CRC mouse model were established to investigate the effects of emodin on CRC. RESULTS The ACSL4, VEGFR1, and VEGFR2 expression were upregulated in CRC tissues and ACSL4 was associated with a shorter survival time in CRC patients. ACSL4 downregulation reduced cell proliferation and invasion, while ACSL4 exhibited a positive correlation with the levels of VEGFR1, VEGFR2, and VEGF. In HCT116 cells, emodin reduced cell proliferation and invasion by inhibiting ACSL4, VEGFR1, and VEGFR2 expression and VEGF secretion. Docking simulation and MST assay confirmed that emodin can directly bind to ACSL4 target. Moreover, ACSL4 overexpression abolished the inhibitory effect of emodin on VEGF secretion and VEGFR1 and VEGFR2 expression, but VEGFR1 and VEGFR2 overexpression did not affect the inhibitory effect of emodin on ACSL4 expression and VEGF secretion. Furthermore, emodin reduced the mortality and tumorigenesis of CRC mice and reduced ACSL4, VEGFR1, VEGFR2 expression, and VEGF content. CONCLUSION Our findings indicate that emodin inhibits proliferation and invasion of CRC cells and reduces VEGF secretion and VEGFR1 and VEGFR2 expression by inhibiting ACSL4. This emodin-induced pathway offers insights into the molecular mechanism of its antitumor effect and provides a potential therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Guoliang Dai
- Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Dong Wang
- Department of Acupuncture and Rehabilitation, Jiangsu Second Chinese Medicine Hospital, Nanjing 210017, China
| | - Shitang Ma
- Life and Health College, Anhui Science and Technology University, Fengyang 233100, China
| | - Shengwei Hong
- Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Kang Ding
- National Center of Colorectal Surgery, Jiangsu Integrate Colorectal Oncology Center, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing 210022, China
| | - Xiying Tan
- Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Wenzheng Ju
- Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China.
| |
Collapse
|
14
|
Automatic Segmentation of Magnetic Resonance Images of Severe Patients with Advanced Liver Cancer and the Molecular Mechanism of Emodin-Induced Apoptosis of HepG2 Cells under the Deep Learning. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:3951112. [PMID: 35295165 PMCID: PMC8920667 DOI: 10.1155/2022/3951112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 01/26/2022] [Indexed: 11/17/2022]
Abstract
To improve the accuracy of clinical diagnosis of severe patients with advanced liver cancer and enhance the effect of chemotherapy treatment, the U-Net model was optimized by introducing the batch normalization (BN) layer and the dropout layer, and the segmentation training and verification of the optimized model were realized by the magnetic resonance (MR) image data. Subsequently, HepG2 cells were taken as the research objects and treated with 0, 10, 20, 40, 60, 80, and 100 μmol/L emodin (EMO), respectively. The methyl thiazolyl tetrazolium (MTT) method was used to explore the changes in cell viability, the acridine orange (AO)/ethidium bromide (EB) and 4',6-diamidino-2-phenylindole (DAPI) were used for staining, the Annexin V fluorescein isothiocyanate (FITC)/propidium iodide (PI) (Annexin V-FITC/PI) was adopted to detect the apoptosis after EMO treatment, and the Western blot (WB) method was used with the purpose of exploring the changes in protein expression levels of PARP, Bcl-2, and p53 in the cells after treatment. It was found that compared with the original U-Net model, the introduction of the BN layer and the dropout layer can improve the robustness of the U-Net model, and the optimized U-Net model had the highest dice similarity coefficient (DSC) (98.45%) and mean average precision (MAP) (0.88) for the liver tumor segmentation.
Collapse
|
15
|
Role of Plant-Derived Active Constituents in Cancer Treatment and Their Mechanisms of Action. Cells 2022; 11:cells11081326. [PMID: 35456005 PMCID: PMC9031068 DOI: 10.3390/cells11081326] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 03/31/2022] [Accepted: 04/11/2022] [Indexed: 02/07/2023] Open
Abstract
Despite significant technological advancements in conventional therapies, cancer remains one of the main causes of death worldwide. Although substantial progress has been made in the control and treatment of cancer, several limitations still exist, and there is scope for further advancements. Several adverse effects are associated with modern chemotherapy that hinder cancer treatment and lead to other critical disorders. Since ancient times, plant-based medicines have been employed in clinical practice and have yielded good results with few side effects. The modern research system and advanced screening techniques for plants’ bioactive constituents have enabled phytochemical discovery for the prevention and treatment of challenging diseases such as cancer. Phytochemicals such as vincristine, vinblastine, paclitaxel, curcumin, colchicine, and lycopene have shown promising anticancer effects. Discovery of more plant-derived bioactive compounds should be encouraged via the exploitation of advanced and innovative research techniques, to prevent and treat advanced-stage cancers without causing significant adverse effects. This review highlights numerous plant-derived bioactive molecules that have shown potential as anticancer agents and their probable mechanisms of action and provides an overview of in vitro, in vivo and clinical trial studies on anticancer phytochemicals.
Collapse
|
16
|
Gao M, Yang Y, Gao Y, Liu T, Guan Q, Zhou T, Shi Y, Hao M, Li Z, Zuo D, Zhang W, Wu Y. The anti-MDR efficacy of YAN against A549/Taxol cells is associated with its inhibition on glycolysis and is further enhanced by 2-deoxy-d-glucose. Chem Biol Interact 2022; 354:109843. [PMID: 35122754 DOI: 10.1016/j.cbi.2022.109843] [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: 07/11/2021] [Revised: 01/11/2022] [Accepted: 01/31/2022] [Indexed: 12/09/2022]
Abstract
Aerobic glycolysis is a hallmark of malignant tumor. Here, the hyperactive glycolysis in multidrug-resistant A549/Taxol cells was demonstrated to be essential for maintaining the vigorous cell viability and drug resistance. 5-(4-ethoxyphenyl)-1-(3,4,5-trimethoxyphenyl)-1H-1,2,4-triazol-3-amine (YAN), a newly synthesized tubulin inhibitor, could not only inhibit the glycolysis in A549 and A549/Taxol cells through down-regulating the glycolysis-related proteins, but also disrupt the mitochondrial localization of hexokinase-2 (HK-2) which is related with the apoptosis resistance. The effects of YAN above were relevant to the down-regulation of PI3K-Akt-c-Myc/HIF-1α pathway. Moreover, YAN induced the reactive oxygen species generation in A549 and A549/Taxol cells, which only mediated the apoptosis in A549 cells. We also showed that 2-DG, the glycolysis inhibitor, synergistically enhanced YAN-triggered apoptosis in A549/Taxol cells via further suppressing glycolysis and reducing mitotic slippage. Collectively, we illustrate the inhibition effect of YAN on the glycolysis in A549 and A549/Taxol cells, and provide a fresh insight into the mechanism for the development of YAN as a candidate for multidrug resistant cancer treatment. The finding that 2-DG improved the anti-tumor efficacy of YAN against A549/Taxol cells, offers a reference for solving mitotic slippage-mediated drug resistance.
Collapse
Affiliation(s)
- Minghuan Gao
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Yuying Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Ying Gao
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Tong Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Qi Guan
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Tianhao Zhou
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Yani Shi
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Mingjing Hao
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Zengqiang Li
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Daiying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| | - Weige Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| | - Yingliang Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| |
Collapse
|
17
|
Antiangiogenic Phytochemicals Constituent of Diet as Promising Candidates for Chemoprevention of Cancer. Antioxidants (Basel) 2022; 11:antiox11020302. [PMID: 35204185 PMCID: PMC8868078 DOI: 10.3390/antiox11020302] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 12/04/2022] Open
Abstract
Despite the extensive knowledge on cancer nature acquired over the last years, the high incidence of this disease evidences a need for new approaches that complement the clinical intervention of tumors. Interestingly, many types of cancer are closely related to dietary habits associated with the Western lifestyle, such as low fruit and vegetable intake. Recent advances around the old-conceived term of chemoprevention highlight the important role of phytochemicals as good candidates for the prevention or treatment of cancer. The potential to inhibit angiogenesis exhibited by many natural compounds constituent of plant foods makes them especially interesting for their use as chemopreventive agents. Here, we review the antitumoral potential, with a focus on the antiangiogenic effects, of phenolic and polyphenolic compounds, such as quercetin or myricetin; terpenoids, such as ursolic acid or kahweol; and anthraquinones from Aloe vera, in different in vitro and in vivo assays, and the available clinical data. Although clinical trials have failed to assess the preventive role of many of these compounds, encouraging preclinical data support the efficacy of phytochemicals constituent of diet in the prevention and treatment of cancer, but a deeper understanding of their mechanisms of action and better designed clinical trials are urgently needed.
Collapse
|
18
|
The interplay of pineal hormones and socioeconomic status leading to colorectal cancer disparity. Transl Oncol 2022; 16:101330. [PMID: 34990909 PMCID: PMC8741600 DOI: 10.1016/j.tranon.2021.101330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer-related deaths in the United States. Despite increased screening options and state-of-art treatments offered in clinics, racial differences remain in CRC. African Americans (AAs) are disproportionately affected by the disease; the incidence and mortality are higher in AAs than Caucasian Americans (CAs). At the time of diagnosis, AAs more often present with advanced stages and aggressive CRCs, primarily accounting for the racial differences in therapeutic outcomes and mortality. The early incidence of CRC in AAs could be attributed to race-specific gene polymorphisms and lifestyle choices associated with socioeconomic status (SES). Altered melatonin-serotonin signaling, besides the established CRC risk factors (age, diet, obesity, alcoholism, and tobacco use), steered by SES, glucocorticoid, and Vitamin D status in AAs could also account for the early incidence in this racial group. This review focuses on how the lifestyle factors, diet, allelic variants, and altered expression of specific genes could lead to atypical serotonin and melatonin signaling by modulating the synthesis, secretion, and signaling of these pineal hormones in AAs and predisposing them to develop more aggressive CRC earlier than CAs. Crosstalk between gut microbiota and pineal hormones and its impact on CRC pathobiology is addressed from a race-specific perspective. Lastly, the status of melatonin-focused CRC treatments, the need to better understand the perturbed melatonin signaling, and the potential of pineal hormone-directed therapeutic interventions to reduce CRC-associated disparity are discussed.
Collapse
|
19
|
McDonald SJ, VanderVeen BN, Velazquez KT, Enos RT, Fairman CM, Cardaci TD, Fan D, Murphy EA. Therapeutic Potential of Emodin for Gastrointestinal Cancers. Integr Cancer Ther 2022; 21:15347354211067469. [PMID: 34984952 PMCID: PMC8738880 DOI: 10.1177/15347354211067469] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 01/12/2023] Open
Abstract
Gastrointestinal (GI) cancers cause one-third of all cancer-related deaths worldwide. Natural compounds are emerging as alternative or adjuvant cancer therapies given their distinct advantage of manipulating multiple pathways to both suppress tumor growth and alleviate cancer comorbidities; however, concerns regarding efficacy, bioavailability, and safety are barriers to their development for clinical use. Emodin (1,3,8-trihydroxy-6-methylanthraquinone), a Chinese herb-derived anthraquinone, has been shown to exert anti-tumor effects in colon, liver, and pancreatic cancers. While the mechanisms underlying emodin's tumoricidal effects continue to be unearthed, recent evidence highlights a role for mitochondrial mediated apoptosis, modulated stress and inflammatory signaling pathways, and blunted angiogenesis. The goals of this review are to (1) highlight emodin's anti-cancer properties within GI cancers, (2) discuss the known anti-cancer mechanisms of action of emodin, (3) address emodin's potential as a treatment complementary to standard chemotherapeutics, (4) assess the efficacy and bioavailability of emodin derivatives as they relate to cancer, and (5) evaluate the safety of emodin.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Daping Fan
- University of South Carolina, Columbia, SC, USA
- AcePre, LLC, Columbia, SC, USA
| | - E. Angela Murphy
- University of South Carolina, Columbia, SC, USA
- AcePre, LLC, Columbia, SC, USA
| |
Collapse
|
20
|
Zhang FY, Li RZ, Xu C, Fan XX, Li JX, Meng WY, Wang XR, Liang TL, Guan XX, Pan HD, Liu L, Yao XJ, Wu QB, Leung ELH. Emodin induces apoptosis and suppresses non-small-cell lung cancer growth via downregulation of sPLA2-IIa. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 95:153786. [PMID: 34785104 DOI: 10.1016/j.phymed.2021.153786] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/22/2021] [Accepted: 09/29/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Lung cancer has become the principal cause of cancer-related deaths. Emodin is a Chinese herb-derived compound extracted from the roots of Rheum officinale that exhibits numerous pharmacological characteristics. Secretory phospholipase A2-IIa (sPLA2-IIa) is overexpressed in cancers and plays an important role in cancer development. PURPOSE This study aims to investigate the anti-tumor mechanism of emodin in non-small-cell lung cancer (NSCLC). METHODS MTT assay was applied to detect the sensitivity of emodin to NSCLC cell line. Flow cytometry was used to examine the effect of emodin on cell cycle distribution and evaluate ROS level and apoptosis. Western blot analysis was utilised to examine the expression levels of sPLA2-IIa, PKM2, and AMPK and its downstream pathways induced by emodin. Enzyme inhibition assay was applied to investigate the inhibitory effect of emodin on sPLA2-IIa. The anticancer effect of emodin was also detected using an in vivo model. RESULTS Emodin significantly inhibited NSCLC proliferation in vivo and in vitro and was relatively less cytotoxic to normal lung cell lines. Most importantly, emodin inhibited the proliferation of KRAS mutant cell lines by decreasing the expression of sPLA2-IIa and NF-κB pathways. Emodin also inhibited mTOR and AKT and activated the AMPK pathway. Furthermore, emodin induced apoptosis, increased the reactive oxygen species (ROS) level, and arrested the cell cycle. CONCLUSION Emodin exhibited a novel anti-tumor mechanism of inhibiting the proliferation of KRAS mutant cell lines by decreasing the expression levels of sPLA2-IIa and NF-κB pathways. Hence, emodin can potentially serve as a therapeutic target in NSCLC.
Collapse
Affiliation(s)
- Fang-Yuan Zhang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Avenida Wai Long, Macao, Taipa Macau (SAR), China; State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Run-Ze Li
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Avenida Wai Long, Macao, Taipa Macau (SAR), China; State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Cong Xu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Avenida Wai Long, Macao, Taipa Macau (SAR), China; State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Xing-Xing Fan
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Avenida Wai Long, Macao, Taipa Macau (SAR), China; State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Jia-Xin Li
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Avenida Wai Long, Macao, Taipa Macau (SAR), China; State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Wei-Yu Meng
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Avenida Wai Long, Macao, Taipa Macau (SAR), China; State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Xuan-Run Wang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Avenida Wai Long, Macao, Taipa Macau (SAR), China; State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Tu-Liang Liang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Avenida Wai Long, Macao, Taipa Macau (SAR), China; State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Xiao-Xiang Guan
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Avenida Wai Long, Macao, Taipa Macau (SAR), China; State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Hu-Dan Pan
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Avenida Wai Long, Macao, Taipa Macau (SAR), China; State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Liang Liu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Avenida Wai Long, Macao, Taipa Macau (SAR), China; State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Xiao-Jun Yao
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Avenida Wai Long, Macao, Taipa Macau (SAR), China; State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Qi Biao Wu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Avenida Wai Long, Macao, Taipa Macau (SAR), China; State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China.
| | - Elaine Lai-Han Leung
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Avenida Wai Long, Macao, Taipa Macau (SAR), China; State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China; Zhuhai Hospital of Traditional Chinese and Western Medicine, Zhuhai City, Guangdong, PR China.
| |
Collapse
|
21
|
Emodin Improves Intestinal Health and Immunity through Modulation of Gut Microbiota in Mice Infected by Pathogenic Escherichia coli O 1. Animals (Basel) 2021; 11:ani11113314. [PMID: 34828045 PMCID: PMC8614316 DOI: 10.3390/ani11113314] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 02/07/2023] Open
Abstract
The effect of emodin on the intestinal mucosal barrier of a mouse E. coli O1-induced diarrhea model was observed. Following successful establishment of a diarrhea model, the mice were treated with drugs for seven days. Intestinal lesions and the shape and the number of goblet cells were assessed via hematoxylin-eosin and periodic-acid-Schiff staining, while changes in inflammatory factors, ultrastructure of the small intestine, expression of MUC-2, and changes in the intestinal microbiota were analyzed via RT-PCR, electron microscopy, immunofluorescence, and 16S rRNA sequencing. Examination showed that emodin ameliorated pathological damage to the intestines of diarrheic mice. RT-PCR indicated that emodin reduced TNF-α, IL-β, IL-6, MPO, and COX-2 mRNA levels in duodenal tissues and increased the levels of sIgA and MUC-2 and the number of goblet cells. Microbiome analysis revealed that Escherichia coli O1 reduced bacterial richness and altered the distribution pattern of bacterial communities at the phylum and order levels in cecum contents. Notably, pathogenic Clostridiales and Enterobacteriales were significantly increased in diarrheic mice. However, emodin reversed the trend. Thus, emodin protected against intestinal damage induced by E. coli O1 and improved intestinal mucosal barrier function in mice by increasing the abundance of beneficial intestinal microbiota and inhibiting the abundance of harmful bacteria, thereby alleviating diarrhea.
Collapse
|
22
|
Mani S, Swargiary G, Ralph SJ. Targeting the redox imbalance in mitochondria: A novel mode for cancer therapy. Mitochondrion 2021; 62:50-73. [PMID: 34758363 DOI: 10.1016/j.mito.2021.11.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 10/14/2021] [Accepted: 11/01/2021] [Indexed: 12/19/2022]
Abstract
Changes in reactive oxygen species (ROS) levels affect many aspects of cell behavior. During carcinogenesis, moderate ROS production modifies gene expression to alter cell function, elevating metabolic activity and ROS. To avoid extreme ROS-activated death, cancer cells increase antioxidative capacity, regulating sustained ROS levels that promote growth. Anticancer therapies are exploring inducing supranormal, cytotoxic oxidative stress levels either inhibiting antioxidative capacity or promoting excess ROS to selectively destroy cancer cells, triggering mechanisms such as apoptosis, autophagy, necrosis, or ferroptosis. This review exemplifies pro-oxidants (natural/synthetic/repurposed drugs) and their clinical significance as cancer therapies providing revolutionary approaches.
Collapse
Affiliation(s)
- Shalini Mani
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India.
| | - Geeta Swargiary
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Stephen J Ralph
- School of Medical Science, Griffith University, Southport, Australia.
| |
Collapse
|
23
|
Zheng Q, Li S, Li X, Liu R. Advances in the study of emodin: an update on pharmacological properties and mechanistic basis. Chin Med 2021; 16:102. [PMID: 34629100 PMCID: PMC8504117 DOI: 10.1186/s13020-021-00509-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/17/2021] [Indexed: 12/21/2022] Open
Abstract
Rhei Radix et Rhizoma, also known as rhubarb or Da Huang, has been widely used as a spice and as traditional herbal medicine for centuries, and is currently marketed in China as the principal herbs in various prescriptions, such as Da-Huang-Zhe-Chong pills and Da-Huang-Qing-Wei pills. Emodin, a major bioactive anthraquinone derivative extracted from rhubarb, represents multiple health benefits in the treatment of a host of diseases, such as immune-inflammatory abnormality, tumor progression, bacterial or viral infections, and metabolic syndrome. Emerging evidence has made great strides in clarifying the multi-targeting therapeutic mechanisms underlying the efficacious therapeutic potential of emodin, including anti-inflammatory, immunomodulatory, anti-fibrosis, anti-tumor, anti-viral, anti-bacterial, and anti-diabetic properties. This comprehensive review aims to provide an updated summary of recent developments on these pharmacological efficacies and molecular mechanisms of emodin, with a focus on the underlying molecular targets and signaling networks. We also reviewed recent attempts to improve the pharmacokinetic properties and biological activities of emodin by structural modification and novel material-based targeted delivery. In conclusion, emodin still has great potential to become promising therapeutic options to immune and inflammation abnormality, organ fibrosis, common malignancy, pathogenic bacteria or virus infections, and endocrine disease or disorder. Scientifically addressing concerns regarding the poor bioavailability and vague molecular targets would significantly contribute to the widespread acceptance of rhubarb not only as a dietary supplement in food flavorings and colorings but also as a health-promoting TCM in the coming years.
Collapse
Affiliation(s)
- Qi Zheng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Shuo Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Xiaojiaoyang Li
- School of Life Sciences, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Runping Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China.
| |
Collapse
|
24
|
The Health Benefits of Emodin, a Natural Anthraquinone Derived from Rhubarb-A Summary Update. Int J Mol Sci 2021; 22:ijms22179522. [PMID: 34502424 PMCID: PMC8431459 DOI: 10.3390/ijms22179522] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 08/27/2021] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
Emodin (6-methyl-1,3,8-trihydroxyanthraquinone) is a naturally occurring anthraquinone derivative found in roots and leaves of various plants, fungi and lichens. For a long time it has been used in traditional Chinese medicine as an active ingredient in herbs. Among other sources, it is isolated from the rhubarb Rheum palmatum or tuber fleece-flower Polygonam multiflorum. Emodin has a wide range of biological activities, including diuretic, antibacterial, antiulcer, anti-inflammatory, anticancer and antinociceptive. According to the most recent studies, emodin acts as an antimalarial and antiallergic agent, and can also reverse resistance to chemotherapy. In the present work the potential therapeutic role of emodin in treatment of inflammatory diseases, cancers and microbial infections is analysed.
Collapse
|
25
|
Wu FL, Chu PY, Chen GY, Wang K, Hsu WY, Ahmed A, Ma WL, Cheng WC, Wu YC, Yang JC. Natural anthraquinone compound emodin as a novel inhibitor of aurora A kinase: A pilot study. Chem Biol Drug Des 2021; 99:126-135. [PMID: 34411446 DOI: 10.1111/cbdd.13938] [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: 05/24/2021] [Revised: 07/08/2021] [Accepted: 07/24/2021] [Indexed: 12/14/2022]
Abstract
Aurora kinase A (AURKA) carries out an essential role in proliferation and involves in cisplatin resistance in various cancer cells. Overexpression of AURKA is associated with the poor prognosis of cancer patients. Thus, AURKA has been considered as a target for cancer therapy. Developing AURKA inhibitors became an important issue in cancer therapy. A natural compound emodin mainly extracted from rhubarbs possesses anti-cancer properties. However, the effect of emodin on AURKA has never been investigated. In the present study, molecular docking analysis indicated that emodin interacts with AURKA protein active site. We also found nine emodin analogues from Key Organic database by using ChemBioFinder software. Among that, one analogue 8L-902 showed a similar anti-cancer effect as emodin. The bindings of emodin and 8L-902 on AURKA protein were confirmed by cellular thermal shift assay. Furthermore, emodin inhibited the AURKA kinase activity in vitro and enhanced the cisplatin-DNA adduct level in a resistant ovarian cancer cell line. It seems that emodin may have the potential to inhibit cancer cell growth and enhance cisplatin therapy in cancer with resistance. Collectively, our finding reveals a novel AURKA inhibitor, emodin, which may be vulnerable to ovarian cancer therapy in the future.
Collapse
Affiliation(s)
- Fen-Lan Wu
- Department of Obstetrics and Gynecology, Suzhou BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Suzhou, China
| | - Pei-Yi Chu
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan
| | - Guan-Yu Chen
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan
| | - Ke Wang
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan.,Sex Hormone Research Center, Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
| | - Wei-Yu Hsu
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan
| | - Azaj Ahmed
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan.,Sex Hormone Research Center, Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
| | - Wen-Lung Ma
- Sex Hormone Research Center, Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
| | - Wei-Chung Cheng
- Sex Hormone Research Center, Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Biomedical Sciences, Graduate Institution of Cancer Biology, Graduate Institute of Public Health, China Medical University, Taichung, Taiwan
| | - Yang-Chang Wu
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Integrated Medicine, School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Juan-Cheng Yang
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Integrated Medicine, School of Chinese Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
26
|
Tuli HS, Aggarwal V, Tuorkey M, Aggarwal D, Parashar NC, Varol M, Savla R, Kaur G, Mittal S, Sak K. Emodin: A metabolite that exhibits anti-neoplastic activities by modulating multiple oncogenic targets. Toxicol In Vitro 2021; 73:105142. [PMID: 33722736 DOI: 10.1016/j.tiv.2021.105142] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/11/2021] [Accepted: 03/09/2021] [Indexed: 12/19/2022]
|
27
|
Is Emodin with Anticancer Effects Completely Innocent? Two Sides of the Coin. Cancers (Basel) 2021; 13:cancers13112733. [PMID: 34073059 PMCID: PMC8198870 DOI: 10.3390/cancers13112733] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 12/12/2022] Open
Abstract
Many anticancer active compounds are known to have the capacity to destroy pathologically proliferating cancer cells in the body, as well as to destroy rapidly proliferating normal cells. Despite remarkable advances in cancer research over the past few decades, the inclusion of natural compounds in researches as potential drug candidates is becoming increasingly important. However, the perception that the natural is reliable is an issue that needs to be clarified. Among the various chemical classes of natural products, anthraquinones have many biological activities and have also been proven to exhibit a unique anticancer activity. Emodin, an anthraquinone derivative, is a natural compound found in the roots and rhizomes of many plants. The anticancer property of emodin, a broad-spectrum inhibitory agent of cancer cells, has been detailed in many biological pathways. In cancer cells, these molecular mechanisms consist of suppressing cell growth and proliferation through the attenuation of oncogenic growth signaling, such as protein kinase B (AKT), mitogen-activated protein kinase (MAPK), HER-2 tyrosine kinase, Wnt/-catenin, and phosphatidylinositol 3-kinase (PI3K). However, it is known that emodin, which shows toxicity to cancer cells, may cause kidney toxicity, hepatotoxicity, and reproductive toxicity especially at high doses and long-term use. At the same time, studies of emodin, which has poor oral bioavailability, to transform this disadvantage into an advantage with nano-carrier systems reveal that natural compounds are not always directly usable compounds. Consequently, this review aimed to shed light on the anti-proliferative and anti-carcinogenic properties of emodin, as well as its potential toxicities and the advantages of drug delivery systems on bioavailability.
Collapse
|
28
|
Liu C, Chen L, Wang W, Qin D, Jia C, Yuan M, Wang H, Guo Y, Zhu J, Zhou Y, Zhao H, Liu T. Emodin Suppresses the Migration and Invasion of Melanoma Cells. Biol Pharm Bull 2021; 44:771-779. [PMID: 33731543 DOI: 10.1248/bpb.b20-00807] [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] [Indexed: 11/22/2022]
Abstract
Emodin (1,3,8-trihydroxy-6-methylanthraquinone), as an active ingredient in rhubarb roots and rhizomes, has been reported to possess various pharmacological properties including anti-tumor effects. Recent studies have confirmed that emodin inhibited cell proliferation and induced apoptosis of cancer cells. However, the inhibitory effect of emodin on the migration and invasion of melanoma cells and its underlying mechanism are still unclear. In the study, we observed the impercipient effects of emodin in B16F10 and A375 melanoma cells with strong metastatic abilities, focusing on the functions and mechanisms of migration and invasion of B16F10 and A375 melanoma cells. Cell counting kit-8 (CCK-8), colony formation test and Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) staining tests confirmed that emodin possessed anti-proliferative and pro-apoptotic activities in B16F10 and A375 cells. The inhibitory effects on the migration and invasion of B16F10 and A375 cells were proved by wound healing assay and Transwell methods. Moreover, immunofluorescence assay approved the decrease in protein expression of matrix metalloproteinas (MMP)-2/-9 by emodin, and Western blot analyses revealed that emodin could increase the Bax/Bcl-2 ratio and inhibit the MMP-2/-9 protein expression and Wnt/β-catenin pathway in a dose-depended manner. BML-284, as an agonist of Wnt/β-catenin signaling pathway, reversed the effects of emodin on cell growth, migration and invasion in B16F10 cells. These findings may suggest that emodin treatment can be a promising therapeutic strategy for melanoma with highly metastatic abilities.
Collapse
Affiliation(s)
- Chi Liu
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Hua Dong Hospital Affiliated to Fu Dan University
| | - Liang Chen
- Department of Plastic and Reconstructive Surgery, Hua Dong Hospital Affiliated to Fu Dan University
| | - Wanchen Wang
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Hua Dong Hospital Affiliated to Fu Dan University
| | - Dengke Qin
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Hua Dong Hospital Affiliated to Fu Dan University
| | - Chuanlong Jia
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Hua Dong Hospital Affiliated to Fu Dan University
| | - Mingjie Yuan
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Hua Dong Hospital Affiliated to Fu Dan University
| | - Heng Wang
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Hua Dong Hospital Affiliated to Fu Dan University
| | - Yu Guo
- Department of Plastic and Reconstructive Surgery, Hua Dong Hospital Affiliated to Fu Dan University
| | - Jingjing Zhu
- Department of Plastic and Reconstructive Surgery, Hua Dong Hospital Affiliated to Fu Dan University
| | - Yiqun Zhou
- Department of Plastic and Reconstructive Surgery, Hua Dong Hospital Affiliated to Fu Dan University
| | - Haiguang Zhao
- Department of Plastic and Reconstructive Surgery, Hua Dong Hospital Affiliated to Fu Dan University
| | - Tianyi Liu
- Department of Plastic and Reconstructive Surgery, Hua Dong Hospital Affiliated to Fu Dan University
| |
Collapse
|
29
|
Saeedifar AM, Mosayebi G, Ghazavi A, Bushehri RH, Ganji A. Macrophage polarization by phytotherapy in the tumor microenvironment. Phytother Res 2021; 35:3632-3648. [DOI: 10.1002/ptr.7058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/28/2020] [Accepted: 02/08/2021] [Indexed: 12/16/2022]
Affiliation(s)
- Amir Mohammad Saeedifar
- Department of Immunology & Microbiology, School of Medicine Arak University of Medical Sciences Arak Iran
| | - Ghasem Mosayebi
- Department of Immunology & Microbiology, School of Medicine Arak University of Medical Sciences Arak Iran
- Molecular and Medicine Research Center Arak University of Medical Sciences Arak Iran
| | - Ali Ghazavi
- Department of Immunology & Microbiology, School of Medicine Arak University of Medical Sciences Arak Iran
- Traditional and Complementary Medicine Research Center (TCMRC) Arak University of Medical Sciences Arak Iran
| | - Rouhollah Hemmati Bushehri
- Department of Immunology & Microbiology, School of Medicine Arak University of Medical Sciences Arak Iran
| | - Ali Ganji
- Department of Immunology & Microbiology, School of Medicine Arak University of Medical Sciences Arak Iran
- Molecular and Medicine Research Center Arak University of Medical Sciences Arak Iran
| |
Collapse
|
30
|
Ismatullah H, Jabeen I, Saeed MT. Biological Regulatory Network (BRN) Analysis and Molecular Docking Simulations to Probe the Modulation of IP 3R Mediated Ca 2+ Signaling in Cancer. Genes (Basel) 2020; 12:34. [PMID: 33383780 PMCID: PMC7823498 DOI: 10.3390/genes12010034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 12/21/2022] Open
Abstract
Inositol trisphosphate receptor (IP3R) mediated Ca+2 signaling is essential in determining the cell fate by regulating numerous cellular processes, including cell division and cell death. Despite extensive studies about the characterization of IP3R in cancer, the underlying molecular mechanism initiating the cell proliferation and apoptosis remained enigmatic. Moreover, in cancer, the modulation of IP3R in downstream signaling pathways, which control oncogenesis and cancer progression, is not well characterized. Here, we constructed a biological regulatory network (BRN), and describe the remodeling of IP3R mediated Ca2+ signaling as a central key that controls the cellular processes in cancer. Moreover, we summarize how the inhibition of IP3R affects the deregulated cell proliferation and cell death in cancer cells and results in the initiation of pro-survival responses in resistance of cell death in normal cells. Further, we also investigated the role of stereo-specificity of IP3 molecule and its analogs in binding with the IP3 receptor. Molecular docking simulations showed that the hydroxyl group at R6 position along with the phosphate group at R5 position in 'R' conformation is more favorable for IP3 interactions. Additionally, Arg-266 and Arg-510 showed π-π and hydrogen bond interactions and Ser-278 forms hydrogen bond interactions with the IP3 binding site. Thus, they are identified as crucial for the binding of antagonists.
Collapse
Affiliation(s)
| | - Ishrat Jabeen
- Research Center for Modeling and Simulation (RCMS), National University of Sciences and Technology (NUST), Academic-I Building, H-12 Islamabad 44000, Pakistan; (H.I.); (M.T.S.)
| | | |
Collapse
|
31
|
Jänicke P, Lennicke C, Meister A, Seliger B, Wessjohann LA, Kaluđerović GN. Fluorescent spherical mesoporous silica nanoparticles loaded with emodin: Synthesis, cellular uptake and anticancer activity. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 119:111619. [PMID: 33321661 DOI: 10.1016/j.msec.2020.111619] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 09/24/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022]
Abstract
The natural product emodin (EO) exhibits anti-inflammatory, antiangiogenesis and antineoplastic properties in vitro and in vivo. Due to its biological properties as well as its fluorescence, EO can be useful in pharmacology and pharmacokinetics. To enhance its selectivity to cancer cells, EO was loaded into non-fluorescent and novel fluorescent spherical mesoporous nanoparticles bearing N-methyl isatoic anhydride (SNM~M) or lissamine rhodamine B sulfonyl moieties (SNM~L). The propylamine functionalized mesoporous silica nanomaterial (SNM) were characterized by powder X-ray diffraction (XRD), nitrogen gas sorption, scanning electron microscopy (SEM), transmission electron microscopy (TEM), fluorescence spectroscopy, thermogravimetric analysis (TGA) and UV spectroscopy. The cytotoxicity of EO-loaded nanoparticles was tested against the human colon carcinoma cell line HT-29. Non-loaded SNM did not affect cell proliferation, whereas those loaded with EO were at least as efficient as EO alone. It could be shown by fluorescence microscopy that the uptake of silica nanomaterial by the tumor cells occurred within 2 h and the release of EO occurred within 48 h of treatment. Flow cytometry and Western blot analysis showed that SNM containing EO induced apoptosis in HT-29 cells.
Collapse
Affiliation(s)
- Paul Jänicke
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, D 06120 Halle (Saale), Germany
| | - Claudia Lennicke
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, D 06112 Halle (Saale), Germany
| | - Annette Meister
- Institute for Chemistry - Physical and Theoretical Chemistry, Martin Luther University Halle-Wittenberg, D 06099 Halle (Saale), Germany
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, D 06112 Halle (Saale), Germany
| | - Ludger A Wessjohann
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, D 06120 Halle (Saale), Germany
| | - Goran N Kaluđerović
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, D 06120 Halle (Saale), Germany; Department of Engineering and Natural Sciences, University of Applied Sciences Merseburg, Eberhard-Leibnitz-Straße 2, 06217 Merseburg, Germany.
| |
Collapse
|
32
|
Çınar Ayan İ, Çetinkaya S, Dursun HG, Süntar İ. Bioactive Compounds of Rheum ribes L. and its Anticancerogenic Effect via Induction of Apoptosis and miR-200 Family Expression in Human Colorectal Cancer Cells. Nutr Cancer 2020; 73:1228-1243. [DOI: 10.1080/01635581.2020.1792947] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- İlknur Çınar Ayan
- Department of Medical Biology, Medical Faculty, Necmettin Erbakan University, Meram, Konya, Turkey
| | - Sümeyra Çetinkaya
- Biotechnology Research Center of Ministry of Agriculture and Forestry, Yenimahalle, Ankara, Turkey
| | - Hatice Gül Dursun
- Department of Medical Biology, Medical Faculty, Necmettin Erbakan University, Meram, Konya, Turkey
| | - İpek Süntar
- Department of Pharmacognosy Faculty of Pharmacy, Gazi University, Etiler, Ankara, Turkey
| |
Collapse
|
33
|
Isocitrate dehydrogenase 2 contributes to radiation resistance of oesophageal squamous cell carcinoma via regulating mitochondrial function and ROS/pAKT signalling. Br J Cancer 2020; 123:126-136. [PMID: 32367071 PMCID: PMC7340793 DOI: 10.1038/s41416-020-0852-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/06/2020] [Accepted: 04/01/2020] [Indexed: 11/29/2022] Open
Abstract
Background Antioxidase alleviates the accumulation of radiation-induced reactive oxygen species (ROS) and therefore has strong connections with radioresistance. Isocitrate dehydrogenase 2 (IDH2) facilitates the turnover of antioxidase, but its role in radiotherapeutic efficiency in oesophageal squamous cell carcinoma (ESCC) still remains elusive. Methods The involvement of IDH2 in radiotherapeutic efficacy in ESCC was investigated in vitro and vivo by IDH2 knockdown. IDH2 expression in biopsy specimens of 141 patients was identified to evaluate its clinical significance. Results We found that Kyse510 and Kyse140 cells were more radioresistant and had higher IDH2 expression. In these two cell lines, IDH2 knockdown intensified the radiation-induced ROS overload and oxidative damage on lipid, protein, and nucleic acids. In addition, IDH2 silencing aggravated the radiation-induced mitochondrial dysfunction and cell apoptosis and ultimately promoted radiosensitisation via inhibiting AKT phosphorylation in a ROS-dependent manner. Furthermore, IDH2 depletion facilitated the radiation-induced growth inhibition and cell apoptosis in murine xenografts. Finally, IDH2 expression was correlated with definite chemoradiotherapy (dCRT) efficacy and served as an independent prognostic factor for survival of ESCC patients. Conclusions IDH2 plays a key role in the radioresistance of ESCC. Targeting IDH2 could be a promising regimen to improve radiotherapeutic efficiency in ESCC patients.
Collapse
|
34
|
Ashrafizadeh M, Javanmardi S, Moradi-Ozarlou M, Mohammadinejad R, Farkhondeh T, Samarghandian S, Garg M. Natural products and phytochemical nanoformulations targeting mitochondria in oncotherapy: an updated review on resveratrol. Biosci Rep 2020; 40:BSR20200257. [PMID: 32163546 PMCID: PMC7133519 DOI: 10.1042/bsr20200257] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 12/11/2022] Open
Abstract
Mitochondria are intracellular organelles with two distinct membranes, known as an outer mitochondrial membrane and inner cell membrane. Originally, mitochondria have been derived from bacteria. The main function of mitochondria is the production of ATP. However, this important organelle indirectly protects cells by consuming oxygen in the route of energy generation. It has been found that mitochondria are actively involved in the induction of the intrinsic pathways of apoptosis. So, there have been efforts to sustain mitochondrial homeostasis and inhibit its dysfunction. Notably, due to the potential role of mitochondria in the stimulation of apoptosis, this organelle is a promising target in cancer therapy. Resveratrol is a non-flavonoid polyphenol that exhibits significant pharmacological effects such as antioxidant, anti-diabetic, anti-inflammatory and anti-tumor. The anti-tumor activity of resveratrol may be a consequence of its effect on mitochondria. Multiple studies have investigated the relationship between resveratrol and mitochondria, and it has been demonstrated that resveratrol is able to significantly enhance the concentration of reactive oxygen species, leading to the mitochondrial dysfunction and consequently, apoptosis induction. A number of signaling pathways such as sirtuin and NF-κB may contribute to the mitochondrial-mediated apoptosis by resveratrol. Besides, resveratrol shifts cellular metabolism from glycolysis into mitochondrial respiration to induce cellular death in cancer cells. In the present review, we discuss the possible interactions between resveratrol and mitochondria, and its potential application in cancer therapy.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Sara Javanmardi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Masoumeh Moradi-Ozarlou
- Department of Pathobiology, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Reza Mohammadinejad
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Healthy Ageing Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh 201313, India
| |
Collapse
|
35
|
Ashrafizadeh M, Rafiei H, Mohammadinejad R, Afshar EG, Farkhondeh T, Samarghandian S. Potential therapeutic effects of curcumin mediated by JAK/STAT signaling pathway: A review. Phytother Res 2020; 34:1745-1760. [PMID: 32157749 DOI: 10.1002/ptr.6642] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 01/11/2020] [Accepted: 01/31/2020] [Indexed: 12/11/2022]
Abstract
Curcumin is a naturally occurring nutraceutical compound with a number of therapeutic and biological activities such as antioxidant, anti-inflammatory, anti-diabetic, antitumor, and cardioprotective. This plant-derived chemical has demonstrated great potential in targeting various signaling pathways to exert its protective effects. Signal transducers and activator of transcription (STAT) is one of the molecular pathways involved in a variety of biological processes such as cell proliferation and cell apoptosis. Accumulating data demonstrates that the STAT pathway is an important target in treatment of a number of disorders, particularly cancer. Curcumin is capable of affecting STAT signaling pathway in induction of its therapeutic impacts. Curcumin is able to enhance the level of anti-inflammatory cytokines and improve inflammatory disorders such as colitis by targeting STAT signaling pathway. Furthermore, studies show that inhibition of JAK/STAT pathway by curcumin is involved in reduced migration and invasion of cancer cells. Curcumin normalizes the expression of JAK/STAT signaling pathway to exert anti-diabetic, renoprotective, and neuroprotective impacts. At the present review, we provide a comprehensive discussion about the effect of curcumin on JAK/STAT signaling pathway to direct further studies in this field.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Hossein Rafiei
- Department of Biology, Faculty of Sciences, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Elham G Afshar
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Healthy Ageing Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.,Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
36
|
Zhang N, Ren Y, Wang Y, Zhao L, Wang B, Ma N, Gao Z, Cao B. LRG1 Suppresses Migration and Invasion of Esophageal Squamous Cell Carcinoma by Modulating Epithelial to Mesenchymal Transition. J Cancer 2020; 11:1486-1494. [PMID: 32047555 PMCID: PMC6995366 DOI: 10.7150/jca.36189] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 12/08/2019] [Indexed: 01/01/2023] Open
Abstract
Background: Esophageal squamous cell carcinoma (ESCC) is a common cancer with poor prognosis. The molecular pathogenesis underlying ESCC remains to be explored. Leucine-rich ɑ-2-glycoprotein 1 (LRG1) has been implicated in the pathogenesis of various cancer types, however its role in ESCC is unknown. Materials and Methods: Data from the public database was analyzed to address the expression of LRG1 in ESCC. Gain-of-function studies were performed in select ESCC cell lines by over-expression or addition of recombinant LRG1, while loss-of-function studies achieved by small interfering RNA mediated knockdown. Wound healing and transwell assays were conducted to investigate ESCC cell migration and invasion upon manipulating LRG1 levels. Western blot and Immunofluorescence staining were used to examine the changes in epithelial to mesenchymal transition (EMT) and TGFβ signaling pathway. Results: LRG1 mRNA levels were found to be significantly down-regulated in patients with ESCC as well as in several ESCC cell lines. Silencing of LRG1 promoted, while overexpression of LRG1 inhibited ESCC cell migration and invasion. In line with this, Silencing of LRG1 enhanced, while overexpression of LRG1 reduced TGFβ signaling and EMT of ESCC cells. Conclusion/Significance: LRG1 suppresses ESCC cell migration and invasion via negative modulation of TGFβ signaling and EMT. Down-regulation of LRG1 in ESCC patients may favor tumor metastasis and disease progression.
Collapse
Affiliation(s)
- Ninggang Zhang
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China.,Shanxi Cancer Hospital Affiliated to Shanxi Medical University, No. 3 of Zhigong Xincun Street, Xinghualing District, Taiyuan, Shanxi 030013, China
| | - Yaqiong Ren
- Shanxi Cancer Hospital Affiliated to Shanxi Medical University, No. 3 of Zhigong Xincun Street, Xinghualing District, Taiyuan, Shanxi 030013, China
| | - Yusheng Wang
- Shanxi Cancer Hospital Affiliated to Shanxi Medical University, No. 3 of Zhigong Xincun Street, Xinghualing District, Taiyuan, Shanxi 030013, China
| | - Lei Zhao
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Bin Wang
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Nina Ma
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Zhengxing Gao
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Bangwei Cao
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| |
Collapse
|
37
|
Liu L, Chen J, Cao M, Wang J, Wang S. NO donor inhibits proliferation and induces apoptosis by targeting PI3K/AKT/mTOR and MEK/ERK pathways in hepatocellular carcinoma cells. Cancer Chemother Pharmacol 2019; 84:1303-1314. [PMID: 31555866 DOI: 10.1007/s00280-019-03965-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 09/16/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND PABA/NO, O2-{2,4-dinitro-5-[4-(N-methylamino) benzoyloxy] phenyl} 1-(N, N-dimethylamino) diazen-1-ium-1,2-diolate, is a diazeniumdiolate-based NO-donor prodrug that releases exogenous nitric oxide at high concentrations to induce apoptosis in many tumor cell lines. PURPOSE This study aimed to determine the effects of PABA/NO on hepatocellular carcinoma proliferation and apoptosis induction both in vitro and in vivo experiments. RESULTS PABA/NO dramatically inhibited the growth of Bel-7402 hepatocellular carcinoma cells and significantly induced apoptosis in a concentration-dependent manner, accompanied by down-regulation of Bcl-2 and Bcl-xL, up-regulation of Bax and Bad, release of Cyt c and activation of cleaved-caspase-9/3 and cleaved-PARP, which were related to suppressing PI3K/AKT/mTOR and MEK/ERK signaling pathways. LY294002 (a PI3K inhibitor) and U0126 (an ERK inhibitor) prior to PABA/NO were found to synergistically enhance PABA/NO-induced apoptosis. Carboxy-PTIO as a NO scavenger obviously attenuated PABA/NO-induced apoptosis. Additionally, H22 tumor-bearing mice experiments demonstrated that PABA/NO exerted good anti-tumor effects via reducing tumor volume, tumor weight and decreasing the expression of CD34. Furthermore, PABA/NO treatment strongly inhibited the phosphorylation of PI3K/AKT/mTOR and MEK/ERK signaling pathways in H22 hepatocellular carcinoma tissues. CONCLUSIONS PABA/NO induced apoptosis through inhibition of PI3K/Akt/mTOR and MEK/ERK pathway in hepatocellular carcinoma cells.
Collapse
Affiliation(s)
- Ling Liu
- Department of Pharmacy, Medical College, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, China.
| | - Jingjing Chen
- Department of Pharmacy, Medical College, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, China
| | - Mengyao Cao
- Department of Pharmacy, Medical College, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, China
| | - Jiangang Wang
- Department of Pharmacy, Medical College, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, China
| | - Shuying Wang
- Department of Pharmacy, Medical College, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, China
| |
Collapse
|
38
|
Luo H, Vong CT, Chen H, Gao Y, Lyu P, Qiu L, Zhao M, Liu Q, Cheng Z, Zou J, Yao P, Gao C, Wei J, Ung COL, Wang S, Zhong Z, Wang Y. Naturally occurring anti-cancer compounds: shining from Chinese herbal medicine. Chin Med 2019; 14:48. [PMID: 31719837 PMCID: PMC6836491 DOI: 10.1186/s13020-019-0270-9] [Citation(s) in RCA: 315] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/23/2019] [Indexed: 12/24/2022] Open
Abstract
Numerous natural products originated from Chinese herbal medicine exhibit anti-cancer activities, including anti-proliferative, pro-apoptotic, anti-metastatic, anti-angiogenic effects, as well as regulate autophagy, reverse multidrug resistance, balance immunity, and enhance chemotherapy in vitro and in vivo. To provide new insights into the critical path ahead, we systemically reviewed the most recent advances (reported since 2011) on the key compounds with anti-cancer effects derived from Chinese herbal medicine (curcumin, epigallocatechin gallate, berberine, artemisinin, ginsenoside Rg3, ursolic acid, silibinin, emodin, triptolide, cucurbitacin B, tanshinone I, oridonin, shikonin, gambogic acid, artesunate, wogonin, β-elemene, and cepharanthine) in scientific databases (PubMed, Web of Science, Medline, Scopus, and Clinical Trials). With a broader perspective, we focused on their recently discovered and/or investigated pharmacological effects, novel mechanism of action, relevant clinical studies, and their innovative applications in combined therapy and immunomodulation. In addition, the present review has extended to describe other promising compounds including dihydroartemisinin, ginsenoside Rh2, compound K, cucurbitacins D, E, I, tanshinone IIA and cryptotanshinone in view of their potentials in cancer therapy. Up to now, the evidence about the immunomodulatory effects and clinical trials of natural anti-cancer compounds from Chinese herbal medicine is very limited, and further research is needed to monitor their immunoregulatory effects and explore their mechanisms of action as modulators of immune checkpoints.
Collapse
Affiliation(s)
- Hua Luo
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Chi Teng Vong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Hanbin Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yan Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peng Lyu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Ling Qiu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Mingming Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Qiao Liu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zehua Cheng
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jian Zou
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peifen Yao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Caifang Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jinchao Wei
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Carolina Oi Lam Ung
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Shengpeng Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zhangfeng Zhong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yitao Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| |
Collapse
|
39
|
Wang Z, Chen H, Chen J, Hong Z, Liao Y, Zhang Q, Tong H. Emodin sensitizes human pancreatic cancer cells to EGFR inhibitor through suppressing Stat3 signaling pathway. Cancer Manag Res 2019; 11:8463-8473. [PMID: 31572001 PMCID: PMC6756157 DOI: 10.2147/cmar.s221877] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 08/28/2019] [Indexed: 12/17/2022] Open
Abstract
Background Excessive expression of EGFR is closely related to tumor formation, transfer and deterioration, which has attracted much attention. EGFR overexpression may be detected in up to 90% of pancreatic tumors. However, drug resistance of EGFR inhibitors targeting treatment severely limits its clinical application. Methods In this study, Western blotting was used to detect the expression of p-Stat3, EGFR, Bcl-2, cleaved-caspase3 and Bax. Cell apoptosis was evaluated via flow cytometry. The colon assay and MTT assay were applied for detecting the cell proliferation in vitro. The xenograft mouse model was used to examine the cell proliferation in vivo. Results Emodin remarkably enhanced the anti-cancer effect of EGFR inhibitor on pancreatic cancer cells. In addition, emodin promoted afatinib-induced apoptosis by inhibiting the Stat3 signaling pathway. Meanwhile, siRNAs against Stat3 significantly increased the apoptosis of pancreatic cancer cells. EGFR inhibitor promoted phosphorylation of Stat3 in pancreatic cancer cells. Interestingly, emodin combined with EGFR inhibitor inhibited the proliferation of pancreatic cancer cells in vitro. The tumor xenograft mice model was further confirmed that emodin possessed a synergy anticancer effect with afatinib on pancreatic cancer cells by regulating the Stat3 expression. Conclusion These results indicate that the combination of emodin with EGFR inhibitor is an effective therapeutic strategy to sensitize human pancreatic cancer.
Collapse
Affiliation(s)
- Zhaohong Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, People's Republic of China.,Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, People's Republic of China
| | - Hui Chen
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, People's Republic of China
| | - Jingjing Chen
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, People's Republic of China
| | - Zhong Hong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, People's Republic of China
| | - Yi Liao
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, People's Republic of China
| | - Qiyu Zhang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, People's Republic of China.,Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, People's Republic of China
| | - Hongfei Tong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, People's Republic of China
| |
Collapse
|
40
|
Reza Nazifi SM, Asgharshamsi MH, Dehkordi MM, Zborowski KK. Antioxidant properties of Aloe vera components: a DFT theoretical evaluation. Free Radic Res 2019; 53:922-931. [PMID: 31357895 DOI: 10.1080/10715762.2019.1648798] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Prediction of the antioxidant activity of three Aloe vera components (aloesone, aloe-emodin, and isoeleutheol) was performed based on density functional theory calculations using the B3LYP hybrid functional and the 6-311++ G** basis set. Calculation of highest occupied molecular orbital (HOMO), lowest occupied molecular orbital (LUMO), and Egap revealed that aloe-emodin has the lowest Egap value, indicating good antioxidant activity. Also in terms of electron affinity, softness, electrophilicity, and chemical potential, aloe-emodin is a potent structure with potential high radical scavenging activity. Calculation of the ionisation potential revealed that isoeleutherol likely also possesses a high degree of antiradical scavenging. To study the conjugating system of the radicals, density plots of HOMO, natural bond orbital analyses, and spin density plots were used. According to calculations, the isoeleutherol radical is more delocalised and the most stable radical. Calculated proton affinity values revealed that the most probable antioxidant mechanism is sequential proton loss-electron transfer. Our results were compared with available experimental data. Published experimental data were found to correlate well with our theoretical predictions. These results support the usefulness of theoretical calculations not only for identifying potentially useful structures of studied compounds but also for predicting their relative activity.
Collapse
Affiliation(s)
- S Mohamad Reza Nazifi
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences , Isfahan , Iran
| | - Mohammad H Asgharshamsi
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences , Isfahan , Iran
| | - Mehrdad M Dehkordi
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences , Isfahan , Iran
| | | |
Collapse
|