1
|
Chen G, Zhang Y, Zhou Y, Luo H, Guan H, An B. Targeting the mTOR Pathway in Hepatocellular Carcinoma: The Therapeutic Potential of Natural Products. J Inflamm Res 2024; 17:10421-10440. [PMID: 39659752 PMCID: PMC11630751 DOI: 10.2147/jir.s501270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/24/2024] [Indexed: 12/12/2024] Open
Abstract
Despite advancements in cancer treatment through surgery and drugs, hepatocellular carcinoma (HCC) remains a significant challenge, as reflected by its low survival rates. The mammalian target of rapamycin (mTOR) signaling pathway plays a crucial role in regulating the cell cycle, proliferation, apoptosis, and metabolism. Notably, dysregulation leading to the activation of the mTOR signaling pathway is common in HCC, making it a key focus for in-depth research and a target for current therapeutic strategies. This review focuses on the role of the mTOR signaling pathway and its downstream effectors in regulating HCC cell proliferation, apoptosis, autophagy, cell cycle, and metabolic reprogramming. Moreover, it emphasizes the potential of natural products as modulators of the mTOR signaling pathway. When incorporated into combination therapies, these natural products have been demonstrated to augment therapeutic efficacy and surmount drug resistance. These products target key signaling pathways such as mTOR signaling pathways. Examples include 11-epi-sinulariolide acetate, matrine, and asparagus polysaccharide. Their inhibitory effects on these processes suggest valuable directions for the development of more effective HCC therapeutic strategies. Various natural products have demonstrated the ability to inhibit mTOR signaling pathway and suppress HCC progression. These phytochemicals, functioning as mTOR signaling pathway inhibitors, hold great promise as potential anti-HCC agents, especially in the context of overcoming chemoresistance and enhancing the outcomes of combination therapies.
Collapse
Affiliation(s)
- Guo Chen
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Ya Zhang
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Yaqiao Zhou
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Hao Luo
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Hongzhi Guan
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Baiping An
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| |
Collapse
|
2
|
Cai H, Wen H, Li J, Lu L, Zhao W, Jiang X, Bai R. Small-molecule agents for treating skin diseases. Eur J Med Chem 2024; 268:116269. [PMID: 38422702 DOI: 10.1016/j.ejmech.2024.116269] [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: 09/12/2023] [Revised: 02/16/2024] [Accepted: 02/18/2024] [Indexed: 03/02/2024]
Abstract
Skin diseases are a class of common and frequently occurring diseases that significantly impact daily lives. Currently, the limited effective therapeutic drugs are far from meeting the clinical needs; most drugs typically only provide symptomatic relief rather than a cure. Developing small-molecule drugs with improved efficacy holds paramount importance for treating skin diseases. This review aimed to systematically introduce the pathogenesis of common skin diseases in daily life, list related drugs applied in the clinic, and summarize the clinical research status of candidate drugs and the latest research progress of candidate compounds in the drug discovery stage. Also, it statistically analyzed the number of publications and global attention trends for the involved skin diseases. This review might provide practical information for researchers engaged in dermatological drugs and further increase research attention to this disease area.
Collapse
Affiliation(s)
- Hong Cai
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Hao Wen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Junjie Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Liuxin Lu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Wenxuan Zhao
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Xiaoying Jiang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China.
| | - Renren Bai
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China.
| |
Collapse
|
3
|
Wang D, Chen J, Pu L, Yu L, Xiong F, Sun L, Yu Q, Cao X, Chen Y, Peng F, Peng C. Galangin: A food-derived flavonoid with therapeutic potential against a wide spectrum of diseases. Phytother Res 2023; 37:5700-5723. [PMID: 37748788 DOI: 10.1002/ptr.8013] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/08/2023] [Accepted: 08/30/2023] [Indexed: 09/27/2023]
Abstract
Galangin is an important flavonoid with natural activity, that is abundant in galangal and propolis. Currently, various biological activities of galangin have been disclosed, including anti-inflammation, antibacterial effect, anti-oxidative stress and aging, anti-fibrosis, and antihypertensive effect. Based on the above bioactivities, more and more attention has been paid to the role of galangin in neurodegenerative diseases, rheumatoid arthritis, osteoarthritis, osteoporosis, skin diseases, and cancer. In this paper, the natural sources, pharmacokinetics, bioactivities, and therapeutic potential of galangin against various diseases were systematically reviewed by collecting and summarizing relevant literature. In addition, the molecular mechanism and new preparation of galangin in the treatment of related diseases are also discussed, to broaden the application prospect and provide reference for its clinical application. Furthermore, it should be noted that current toxicity and clinical studies of galangin are insufficient, and more evidence is needed to support its possibility as a functional food.
Collapse
Affiliation(s)
- Daibo Wang
- Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Junren Chen
- Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lin Pu
- Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lei Yu
- Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fang Xiong
- Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Luyao Sun
- Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qian Yu
- Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoyu Cao
- Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan Chen
- Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fu Peng
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Cheng Peng
- Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
4
|
Zhang F, Yan Y, Zhang LM, Li DX, Li L, Lian WW, Xia CY, He J, Xu JK, Zhang WK. Pharmacological activities and therapeutic potential of galangin, a promising natural flavone, in age-related diseases. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 120:155061. [PMID: 37689035 DOI: 10.1016/j.phymed.2023.155061] [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: 05/09/2023] [Revised: 08/24/2023] [Accepted: 08/30/2023] [Indexed: 09/11/2023]
Abstract
BACKGROUND The extension of average life expectancy and the aggravation of population aging have become the inevitable trend of human development. In an aging society, various problems related to medical care for the elderly have become increasingly prominent. However, most of the age-related diseases have the characteristics of multiple diseases at the same time, prone to complications, and atypical clinical manifestations, which bring great difficulties to its treatment. Galangin (3,5,7-trihydroxyflavone) is a natural active compound extracted from the root of Alpinia officinarum Hance (Zingiberaceae). Recently, many studies have shown that galangin has potential advantages in the treatment of neurodegenerative diseases and cardiovascular and cerebrovascular diseases, which are common in the elderly. In addition, it also showed that galangin had prospective activities in the treatment of tumor, diabetes, liver injury, asthma and arthritis. PURPOSE This review aims to systematically summarize and discuss the effects and the underlying mechanism of galangin in the treatment of age-related diseases. METHODS We searched PubMed, SciFinder, Web of Science and CNKI literature database resources, combined with the keywords "galangin", "neurodegenerative disease", "tumor", "diabetes", "pharmacological activity", "drug combination", "pharmacokinetics", "drug delivery system" and "safety", and comprehensively reviewed the pharmacological activities and mechanism of galangin in treating age-related diseases. RESULTS According to the previous studies on galangin, the anti-neurodegenerative activity, cardiovascular and cerebrovascular protective activity, anti-tumor activity, anti-diabetes activity, anti-arthritis activity, hepatoprotective activity and antiasthmatic activity of galangin were discussed, and the related mechanisms were classified and summarized in detail. In addition, the drug combination, pharmacokinetics, drug delivery system and safety of galangin were furtherly discussed. CONCLUSIONS This review will provide reference for galangin in the treatment of age-related diseases. Meanwhile, further experimental research and long-term clinical trials are needed to determine the therapeutic safety and efficacy of galangin.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Pharmacy & Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China; School of Chinese Materia Medica & School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yu Yan
- Department of Pharmacy & Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China
| | - Lin-Mei Zhang
- School of Chinese Materia Medica & School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Dong-Xu Li
- Department of Pharmacy & Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China
| | - Li Li
- Key Laboratory of Cosmetic, China National Light Industry, Beijing Technology and Business University, Beijing 100048, China
| | - Wen-Wen Lian
- Department of Pharmacy & Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China
| | - Cong-Yuan Xia
- Department of Pharmacy & Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China
| | - Jun He
- Department of Pharmacy & Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China.
| | - Jie-Kun Xu
- School of Chinese Materia Medica & School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Wei-Ku Zhang
- Department of Pharmacy & Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China.
| |
Collapse
|
5
|
Raza W, Meena A, Luqman S. 3,5,7-trihydroxyflavone restricts proliferation of androgen-independent human prostate adenocarcinoma cells by inducing ROS-mediated apoptosis and reduces tumour growth. J Biochem Mol Toxicol 2023; 37:e23474. [PMID: 37477197 DOI: 10.1002/jbt.23474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/25/2023] [Accepted: 07/08/2023] [Indexed: 07/22/2023]
Abstract
Flavonoids are among the largest groups of secondary metabolites. Studies suggest that dietary intake of flavonoids reduces the risk of cancer. 3,5,7-trihydroxyflavone (THF) belongs to the flavone class of flavonoids and potentially inhibits the growth of many cancers; however, it is unexplored in prostate cancer. This study reports the antiproliferative potential of THF in prostate cancer cell line via reactive oxygen species (ROS)-mediated cascades and examines the tumour reduction potential in swiss albino mice. The potency of THF was evaluated by employing cytotoxicity assays and wound healing assays. Cell cycle, ROS, mitochondrial membrane potential (MMP), and Annexin-V-FITC assay were performed using a flow cytometer. In vivo, anticancer potential was achieved using the mice Ehrlich Ascites Carcinoma (EAC) model. THF inhibits cell growth with IC50 of 64.30 µM (MTT), 81.22 µM (NRU) and 25.81 µM (SRB), substantiated by cell migration assay. Cell-cycle analysis revealed that THF increases the subdiploid population. Furthermore, the Annexin-V-FITC assay evoked a significant induction of late apoptosis at a higher concentration of THF. THF also disrupts MMP, caused by an increased generation of ROS. In the EAC model, THF significantly inhibits tumour growth and increases the percent survival of mice and ROS levels in EAC cells. Hence, it may be concluded that THF might execute its antiproliferative effect via inducing ROS generation and could be a promising lead for preclinical and clinical validations.
Collapse
Affiliation(s)
- Waseem Raza
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, Uttar Pradesh, India
- Jawaharlal Nehru University, New Delhi, India
| | - Abha Meena
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Suaib Luqman
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
6
|
Xiong Y, Wu B, Guo X, Shi D, Xia H, Xu H, Liu X. Galangin delivered by retinoic acid-modified nanoparticles targeted hepatic stellate cells for the treatment of hepatic fibrosis. RSC Adv 2023; 13:10987-11001. [PMID: 37033441 PMCID: PMC10077338 DOI: 10.1039/d2ra07561j] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 03/17/2023] [Indexed: 04/11/2023] Open
Abstract
Hepatic fibrosis (HF) is a chronic hepatic pathological process induced by various liver injuries, with few available therapies. Previous research studies revealed that HF is characterized by the accumulation of excess extracellular matrix in the liver, mainly overexpressed by activated hepatic stellate cells (HSC). Therefore, HSC have been targeted in clinical trials for the management of HF. The aim of the present study was to develop an anti-HF drug delivery system with acrylic resin (Eudragit® RS100, Eud RS100) nanoparticles (NPs) through modification by retinoic acid (RA), modified for binding the retinol-binding protein reporter (RBPR) in HSC. Galangin (GA), is a multiple effects flavonoid which has demonstrated an anti-HF effect in our previous studies. In this study, GA was utilized for the treatment of HF. The results revealed that the NPs were well formed (diameter: 70 nm), spherical in shape, and exhibited uniform distribution and a high encapsulation efficiency. Moreover, a prominent controlled release effect and a significant increase in bioavailability was observed following the encapsulation of GA in NPs. These findings indicated that the limitation of low bioavailability due to the hydrophobic feature of GA was overcome. Furthermore, the pharmacodynamics studies demonstrated that NPs could drastically influence the anti-HF effects of GA after modification with retinoic acid. The results of the present study suggested that retinoic acid-modified GA NPs represent a promising candidate in the development of an anti-HF drug delivery system for the treatment of HF.
Collapse
Affiliation(s)
- Yuanguo Xiong
- Department of Pharmacy, Renmin Hospital of Wuhan University Wuhan 430060 China
- School of Pharmaceuticals, Hubei University of Chinese Medicine, No. 1 HuangJiahu Road West Wuhan 430065 China +86 27 68890239
| | - Bing Wu
- Department of Cardiology, Renmin Hospital, Hubei University of Medicine Shiyan 442000 China
- Department of Cardiology, Renmin Hospital of Wuhan University Jiefang Road 238 Wuhan 430060 China +86 27 88041911 +86 27 88041911
- Cardiovascular Research Institute, Wuhan University Jiefang Road 238 Wuhan 430060 China
- Hubei Key Laboratory of Cardiology Wuhan 430060 China
| | - Xianxi Guo
- Department of Pharmacy, Renmin Hospital of Wuhan University Wuhan 430060 China
| | - Dong Shi
- School of Pharmaceuticals, Hubei University of Chinese Medicine, No. 1 HuangJiahu Road West Wuhan 430065 China +86 27 68890239
| | - Hao Xia
- Department of Cardiology, Renmin Hospital of Wuhan University Jiefang Road 238 Wuhan 430060 China +86 27 88041911 +86 27 88041911
- Cardiovascular Research Institute, Wuhan University Jiefang Road 238 Wuhan 430060 China
- Hubei Key Laboratory of Cardiology Wuhan 430060 China
| | - Hanlin Xu
- School of Pharmaceuticals, Hubei University of Chinese Medicine, No. 1 HuangJiahu Road West Wuhan 430065 China +86 27 68890239
| | - Xiaoxiong Liu
- Department of Cardiology, Renmin Hospital of Wuhan University Jiefang Road 238 Wuhan 430060 China +86 27 88041911 +86 27 88041911
- Cardiovascular Research Institute, Wuhan University Jiefang Road 238 Wuhan 430060 China
- Hubei Key Laboratory of Cardiology Wuhan 430060 China
| |
Collapse
|
7
|
Crosstalk between xanthine oxidase (XO) inhibiting and cancer chemotherapeutic properties of comestible flavonoids- a comprehensive update. J Nutr Biochem 2022; 110:109147. [PMID: 36049673 DOI: 10.1016/j.jnutbio.2022.109147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 12/17/2021] [Accepted: 08/10/2022] [Indexed: 01/13/2023]
Abstract
Gout is an inflammatory disease caused by metabolic disorder or genetic inheritance. People throughout the world are strongly dependent on ethnomedicine for the treatment of gout and some receive satisfactory curative treatment. The natural remedies as well as established drugs derived from natural sources or synthetically made exert their action by mechanisms that are closely associated with anticancer treatment mechanisms regarding inhibition of xanthine oxidase, feedback inhibition of de novo purine synthesis, depolymerization and disappearance of microtubule, inhibition of NF-ĸB activation, induction of TRAIL, promotion of apoptosis, and caspase activation and proteasome inhibition. Some anti-gout and anticancer novel compounds interact with same receptors for their action, e.g., colchicine and colchicine analogues. Dietary flavonoids, i.e., chrysin, kaempferol, quercetin, fisetin, pelargonidin, apigenin, luteolin, myricetin, isorhamnetin, phloretinetc etc. have comparable IC50 values with established anti-gout drug and effective against both cancer and gout. Moreover, a noticeable number of newer anticancer compounds have already been isolated from plants that have been using by local traditional healers and herbal practitioners to treat gout. Therefore, the anti-gout plants might have greater potentiality to become selective candidates for screening of newer anticancer leads.
Collapse
|
8
|
Chen QX, Zhou L, Long T, Qin DL, Wang YL, Ye Y, Zhou XG, Wu JM, Wu AG. Galangin Exhibits Neuroprotective Effects in 6-OHDA-Induced Models of Parkinson’s Disease via the Nrf2/Keap1 Pathway. Pharmaceuticals (Basel) 2022; 15:ph15081014. [PMID: 36015161 PMCID: PMC9413091 DOI: 10.3390/ph15081014] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 11/16/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease, and there is still no cure for it. PD is characterized by the degeneration of dopaminergic neurons, and oxidative stress has been considered an important pathological mechanism. Therefore, the discovery of antioxidants to alleviate the oxidative damage of dopaminergic neurons is a promising therapeutic strategy for PD. First, a network pharmacology approach was used, and nine common core targets of galangin and PD were screened, mainly involving cell aging, apoptosis, and cellular responses to hydrogen peroxide and hypoxia. In addition, the Gene Ontology (GO) function and pathway enrichment analysis of the Kyoto Encyclopedia of Genes and Genomes (KEGG) identified apoptosis, PI3K/Akt, and HIF-1 signaling pathways. Furthermore, the molecular docking results revealed a strong affinity between galangin and the NFE2L2/Nrf2 protein. To validate the above predictions, we employed 6-hydroxydopamine (6-OHDA) to induce neuronal death in HT22 cells and Caenorhabditis elegans (C. elegans). MTT, cell morphology observation, and Hoechst 33342-PI staining results showed that galangin significantly increased the viability of 6-OHDA-treated HT22 cells. In addition, galangin inhibited 6-OHDA-induced ROS generation and apoptosis in HT22 cells. Mechanistic studies demonstrated that galangin activates the Nrf2/Keap1 signaling pathway, as evidenced by the decreased protein expression of Keap1 and increased protein expression of Nrf2 and HO-1. In the 6-OHDA-induced PD model of C. elegans, galangin indeed inhibited the degeneration of dopaminergic neurons, improved behavioral ability, and decreased ROS generation. In conclusion, the current study is the first to show that galangin has the capacity to inhibit neuronal degeneration via the Nrf2/Keap1 pathway, suggesting that galangin is a possible PD treatment.
Collapse
Affiliation(s)
- Qiu-Xu Chen
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Materia Medica, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Education Ministry Key Laboratory of Medical Electrophysiology, School of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Ling Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Materia Medica, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Education Ministry Key Laboratory of Medical Electrophysiology, School of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Tao Long
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Materia Medica, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Education Ministry Key Laboratory of Medical Electrophysiology, School of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Da-Lian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Materia Medica, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Education Ministry Key Laboratory of Medical Electrophysiology, School of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Yi-Ling Wang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Materia Medica, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Education Ministry Key Laboratory of Medical Electrophysiology, School of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Yun Ye
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Xiao-Gang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Materia Medica, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Education Ministry Key Laboratory of Medical Electrophysiology, School of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Jian-Ming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Materia Medica, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Education Ministry Key Laboratory of Medical Electrophysiology, School of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
- Correspondence: (J.-M.W.); (A.-G.W.)
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Materia Medica, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Education Ministry Key Laboratory of Medical Electrophysiology, School of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
- Correspondence: (J.-M.W.); (A.-G.W.)
| |
Collapse
|
9
|
Wu B, Xu C, Ding HS, Qiu L, Gao JX, Li M, Xiong Y, Xia H, Liu X. Galangin inhibits neointima formation induced by vascular injury via regulating the PI3K/AKT/mTOR pathway. Food Funct 2022; 13:12077-12092. [DOI: 10.1039/d2fo02441a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Galangin inhibits neointimal hyperplasia after vascular injury by inhibiting vascular smooth muscle cell proliferation, migration, phenotypic switching and promoting autophagy.
Collapse
Affiliation(s)
- Bing Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Changwu Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Hua-Sheng Ding
- Department of Emergency, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Liqiang Qiu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Ji-Xian Gao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Ming Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yuanguo Xiong
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hao Xia
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xiaoxiong Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
10
|
Tuli HS, Sak K, Adhikary S, Kaur G, Aggarwal D, Kaur J, Kumar M, Parashar NC, Parashar G, Sharma U, Jain A. Galangin: A metabolite that suppresses anti-neoplastic activities through modulation of oncogenic targets. Exp Biol Med (Maywood) 2021; 247:345-359. [PMID: 34904901 DOI: 10.1177/15353702211062510] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
With the dramatic increase in cancer incidence all over the world in the last decades, studies on identifying novel efficient anti-cancer agents have been intensified. Historically, natural products have represented one of the most important sources of new lead compounds with a wide range of biological activities. In this article, the multifaceted anti-cancer action of propolis-derived flavonoid, galangin, is presented, discussing its antioxidant, anti-inflammatory, antiproliferative, pro-apoptotic, anti-angiogenic, and anti-metastatic effects in various cancer cells. In addition, co-effects with standard chemotherapeutic drugs as well as other natural compounds are also under discussion, besides highlighting modern nanotechnological advancements for overcoming the low bioavailability issue characteristic of galangin. Although further studies are needed for confirming the anti-cancer potential of galangin in vivo malignant systems, exploring this natural compound might open new perspectives in molecular oncology.
Collapse
Affiliation(s)
- Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala 133207, Haryana, India
| | | | - Shubham Adhikary
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's, NMIMS, Mumbai 400056, India
| | - Ginpreet Kaur
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's, NMIMS, Mumbai 400056, India
| | - Diwakar Aggarwal
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala 133207, Haryana, India
| | - Jagjit Kaur
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics (CNBP), Faculty of Engineering, The University of New South Wales, Sydney 2052, Australia
| | - Manoj Kumar
- Department of Chemistry, Maharishi Markandeshwar University, Sadopur 134007, India
| | | | - Gaurav Parashar
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala 133207, Haryana, India
| | - Uttam Sharma
- Department of Zoology, Central University of Punjab, Village-Ghudda 151401, Punjab, India
| | - Aklank Jain
- Department of Zoology, Central University of Punjab, Village-Ghudda 151401, Punjab, India
| |
Collapse
|
11
|
The antitumour effect of galangin and luteolin with doxorubicin on chemically induced hepatocellular carcinoma in rats. Contemp Oncol (Pozn) 2021; 25:174-184. [PMID: 34729037 PMCID: PMC8547180 DOI: 10.5114/wo.2021.110048] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/02/2021] [Indexed: 11/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC), a highly malignant tumour with very high morbidity and mortality, remains the second cause of cancer-related deaths worldwide. Galangin is a naturally occurring flavonoid extracted from the propolis and root of Alpinia officinarum, which possesses antitumour efficacy, which has resulted in an increase in interest in related research. Additionally, galangin inhibits cell proliferation and induces apoptosis in several human malignancies. On the other hand, luteolin, a naturally occurring flavonoid found in a variety of edible plants, augments cytotoxicity in different cancer cells through the inhibition of cell-survival pathways and activation of apoptosis. Moreover, luteolin blocks the activity of anti-apoptotic Bcl-2 family members. The present study aimed to assess the antitumour effect of galangin and luteolin in combination and the antitumour effect of a combination of galangin and luteolin together with doxorubicin (DOX) in a chemically induced HCC rat model. Our analyses demonstrated that the combination treatment with galangin, luteolin, and DOX showed the greatest antineoplastic activity against HCC, which was observed by significant decreases in the levels of HCC markers, including serum α-fetoprotein-L3, and hepatic tissue expression of both glypican 3 and heat shock proteins. On the other hand, the hepatic tissue expression of caspase-3 was significantly increased. These results suggest that combination treatment with galangin and luteolin is a promising candidate for clinical use in HCC chemotherapy, especially when used in combination with DOX.
Collapse
|
12
|
Baby J, Devan AR, Kumar AR, Gorantla JN, Nair B, Aishwarya TS, Nath LR. Cogent role of flavonoids as key orchestrators of chemoprevention of hepatocellular carcinoma: A review. J Food Biochem 2021; 45:e13761. [PMID: 34028054 DOI: 10.1111/jfbc.13761] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/23/2021] [Accepted: 04/24/2021] [Indexed: 02/05/2023]
Abstract
Chemopreventive approaches with food-derived phytochemicals are progressively rising as a significant aspect of tumor management and control. Herein, we have showcased the major phytoconstituents belonging to the group of flavanoid, as anti-cancer agents used for the treatment and prevention of hepatocellular carcinoma (HCC). Sorafenib is the sole drug used for the treatment of advanced HCC, but its clinical application is limited because of its severe adverse effects and drug resistance. Diet-based chemoprevention seems to be the way forward for this disease of malignant nature. As HCC is derived from a chronic inflammatory milieu, the regular incorporation of bioactive phytochemicals in the diet will confer protection and prevent progression to hepatocarcinogenesis. Many preclinical studies proved that the health benefits of flavonoids confer cytotoxic potential against various types of cancers including hepatocellular carcinoma. As flavonoids with excellent safety profile are abundantly present in common vegetables and fruits, they can be better utilized for chemoprevention and chemosensitization in such chronic condition. This review highlights the plausible role of the eight most promising flavonoids (Curcumin, Kaempferol, Resveratrol, Quercetin, Silibinin, Baicalein, Galangin and Luteolin) as key orchestrators of chemoprevention in hepatocellular carcinoma with preclinical and clinical evidence. An attempt to address the challenges in its clinical translation is also included. This review also provides an insight into the close association of HCC and metabolic disorders which may further decipher the chemopreventive effect of dietary bioactive from a proof of concept to extensive clinical translation. PRACTICAL APPLICATIONS: According to GLOBOCAN 2020 database, it is estimated that 905,677 new cases of liver cancer and approximately 830,180 deaths related to that. The cancer incidence and mortality are almost similar as it is diagnosed at an advanced stage in patients where systemic drug therapy is the sole approach. Due to the emergence of multidrug resistance and drug-related toxicities, most of the patient can not adhere to the therapy regimen. Flavonoids are known to be a potential anticancer agent with an excellent safety profile. These are found to be effective preclinically against hepatocellular carcinoma through modulation of numerous pathways in hepatocarcinogenesis. But, the bioavailability issue, lack of well designed-validated clinical evidence, the possibility of food-drug interaction etc limit its clinical utility. The research inputs mainly to overcome pharmacokinetic issues along with suitable validation of efficacy and toxicity will be a critical point for establishing flavonoids as an effective, safe, affordable therapeutics.
Collapse
Affiliation(s)
- Jasmine Baby
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi, Kerala, India
| | - Aswathy R Devan
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi, Kerala, India
| | - Ayana R Kumar
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi, Kerala, India
| | | | - Bhagyalakshmi Nair
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi, Kerala, India
| | - Thanatharayil Sathian Aishwarya
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi, Kerala, India
| | - Lekshmi R Nath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi, Kerala, India
| |
Collapse
|
13
|
A comprehensive review on chemotherapeutic potential of galangin. Biomed Pharmacother 2021; 141:111808. [PMID: 34175820 DOI: 10.1016/j.biopha.2021.111808] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/01/2021] [Accepted: 06/07/2021] [Indexed: 12/12/2022] Open
Abstract
Galangin, a non-toxic phytochemical is known to possess several therapeutic applications. Mounting evidences have demonstrated that galangin a naturally available flavonoid exerts anticancer effects via several mechanisms. The phytocompound induces apoptosis and renders antiangiogenic property. Additionally, galangin has demonstrated significate results in combating various cancer types when administered in combination with other phytocompounds or with gold nanoparticles (GNPs). The present article is a critical review of galangin for its treatment on different types of cancer and its usability as an alternative cancer therapeutics.
Collapse
|
14
|
Liu R, Li H, Wei N, Tan Y. Simultaneous determination of two galangin metabolites from Alpinia Officinarum Hance in rat plasma by UF LC-MS/MS and its application in pharmacokinetics study. PeerJ 2021; 9:e11041. [PMID: 33777530 PMCID: PMC7977375 DOI: 10.7717/peerj.11041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/09/2021] [Indexed: 12/12/2022] Open
Abstract
Galangin has multiple pharmacological efficacies, such as anti-cancer, anti-inflammation and anti-oxidation. Galangin can be rapidly converted into glucuronidated metabolites in vivo. This study aimed to establish an UFLC-MS/MS analytical method to simultaneously determine the concentrations of two glucuronidated metabolites of galangin, galangin-3-O-β-D-glucuronic acid (GG-1) and galangin-7-O-β-D-glucuronic acid (GG-2) in rat plasma. After oral administration of galangal extract (0.3 g/kg), blood samples were collected from the orbital sinus, then treated by methanol precipitation and further gradient-eluted with Phenomenex Kinetex 2.6 µm XB-C18 column. The mass spectrometer was manipulated in the negative electrospray ionization (ESI) and selected multiple reaction monitoring (MRM) mode for the analytes. The precursor-to-product ion pairs applied for GG-1, GG-2 and chrysin (as the internal standard, IS) were m/z 445.2→269.0, 445.2→268.9 and 253.0→142.9, respectively. The results showed that the linear ranges for both GG-1 and GG-2 were 2.0–2000.0 ng/mL (r2 > 0.995). The inter- and intra-day precision were 89.3%–109.2%, RSD was less than 15%, and the repeatability was good. The recoveries of both metabolites and IS were over 89%, and matrix effect was within 15%. The validated analytical method was further applied to study the pharmacokinetic profiles of GG-1 and GG-2 in vivo. The pharmacokinetic parameters suggested that Tmax of GG-1 was equivalent to that of GG-2, and MRT0-t, t1/2 of GG-2 were a little higher than those of GG-1. Importantly, AUC0-t and Cmax of GG-2 were almost twice as those of GG-1. In short, the validated UFLCMS/MS analytical method was feasible to simultaneously determine two galangin metabolites GG-1 and GG-2 in rat plasma and further analyze in vivo metabolism of galangin.
Collapse
Affiliation(s)
- Rangru Liu
- Hainan Provincial Key Laboratory of R&D of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, China.,Haikou Key Laboratory of Li Nationality Medicine, School of Pharmacy, Hainan Medical University, Haikou, China.,Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education, Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Hailong Li
- Hainan Provincial Key Laboratory of R&D of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, China.,Haikou Key Laboratory of Li Nationality Medicine, School of Pharmacy, Hainan Medical University, Haikou, China
| | - Na Wei
- Hainan Provincial Key Laboratory of R&D of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, China.,Haikou Key Laboratory of Li Nationality Medicine, School of Pharmacy, Hainan Medical University, Haikou, China
| | - Yinfeng Tan
- Hainan Provincial Key Laboratory of R&D of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, China.,Haikou Key Laboratory of Li Nationality Medicine, School of Pharmacy, Hainan Medical University, Haikou, China
| |
Collapse
|
15
|
Autophagy: Mechanisms and Therapeutic Potential of Flavonoids in Cancer. Biomolecules 2021; 11:biom11020135. [PMID: 33494431 PMCID: PMC7911475 DOI: 10.3390/biom11020135] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/11/2021] [Accepted: 01/18/2021] [Indexed: 12/13/2022] Open
Abstract
Autophagy, which is a conserved biological process and essential mechanism in maintaining homeostasis and metabolic balance, enables cells to degrade cytoplasmic constituents through lysosomes, recycle nutrients, and survive during starvation. Autophagy exerts an anticarcinogenic role in normal cells and inhibits the malignant transformation of cells. On the other hand, aberrations in autophagy are involved in gene derangements, cell metabolism, the process of tumor immune surveillance, invasion and metastasis, and tumor drug-resistance. Therefore, autophagy-targeted drugs may function as anti-tumor agents. Accumulating evidence suggests that flavonoids have anticarcinogenic properties, including those relating to cellular proliferation inhibition, the induction of apoptosis, autophagy, necrosis, cell cycle arrest, senescence, the impairment of cell migration, invasion, tumor angiogenesis, and the reduction of multidrug resistance in tumor cells. Flavonoids, which are a group of natural polyphenolic compounds characterized by multiple targets that participate in multiple pathways, have been widely studied in different models for autophagy modulation. However, flavonoid-induced autophagy commonly interacts with other mechanisms, comprehensively influencing the anticancer effect. Accordingly, targeted autophagy may become the core mechanism of flavonoids in the treatment of tumors. This paper reviews the flavonoid-induced autophagy of tumor cells and their interaction with other mechanisms, so as to provide a comprehensive and in-depth account on how flavonoids exert tumor-suppressive effects through autophagy.
Collapse
|
16
|
Zhao Q, Peng C, Zheng C, He XH, Huang W, Han B. Recent Advances in Characterizing Natural Products that Regulate Autophagy. Anticancer Agents Med Chem 2020; 19:2177-2196. [PMID: 31749434 DOI: 10.2174/1871520619666191015104458] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/16/2018] [Accepted: 08/26/2019] [Indexed: 02/07/2023]
Abstract
Autophagy, an intricate response to nutrient deprivation, pathogen infection, Endoplasmic Reticulum (ER)-stress and drugs, is crucial for the homeostatic maintenance in living cells. This highly regulated, multistep process has been involved in several diseases including cardiovascular and neurodegenerative diseases, especially in cancer. It can function as either a promoter or a suppressor in cancer, which underlines the potential utility as a therapeutic target. In recent years, increasing evidence has suggested that many natural products could modulate autophagy through diverse signaling pathways, either inducing or inhibiting. In this review, we briefly introduce autophagy and systematically describe several classes of natural products that implicated autophagy modulation. These compounds are of great interest for their potential activity against many types of cancer, such as ovarian, breast, cervical, pancreatic, and so on, hoping to provide valuable information for the development of cancer treatments based on autophagy.
Collapse
Affiliation(s)
- Qian Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Chuan Zheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Xiang-Hong He
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China.,The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, United States
| |
Collapse
|
17
|
Xiong Y, Lu H, Xu H. Galangin Reverses Hepatic Fibrosis by Inducing HSCs Apoptosis via the PI3K/Akt, Bax/Bcl-2, and Wnt/β-Catenin Pathway in LX-2 Cells. Biol Pharm Bull 2020; 43:1634-1642. [PMID: 32893252 DOI: 10.1248/bpb.b20-00258] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hepatic fibrosis (HF) is a common disease, with currently no available treatment. Galangin, a natural flavonoid extracted from Alpinia officinaruim Hance, has multiple effects demonstrated in previous studies. The aim of the present study was to explore the anti-fibrogenic effect of galangin in vitro, and research its potential molecular mechanisms. LX-2 cells were chosen as an in vitro HF model, and were treated with galangin in different concentrations. Cell viability was analyzed using Cell Counting Kit-8 (CCK-8) and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, cell apoptosis was measured using flow cytometry, and the anti-fibrogenic effect of galangin was determined using RT-quantitative (q)PCR, immunofluorescence, and Western blotting. The results show that the proliferation of LX-2 cells was efficiently inhibited by galangin, and apoptosis was induced in a dose-dependent manner. Both the mRNA and protein expression of alpha-smooth muscle actin (α-SMA) and collagen I were markedly downregulated. Galangin also inhibited the phosphatidylinositol 3-kinase (PI3K)/Akt and Wnt/β-catenin signaling pathways and increased the Bax/Bcl-2 ratio. The results of this study suggest that galangin has an anti-fibrogenic effect and may represent a promising agent in the treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Yuanguo Xiong
- School of Pharmaceuticals, Hubei University of Chinese Medicine
| | - Hao Lu
- School of Pharmaceuticals, Hubei University of Chinese Medicine
| | - Hanlin Xu
- School of Pharmaceuticals, Hubei University of Chinese Medicine
| |
Collapse
|
18
|
Kong Y, Feng Z, Chen A, Qi Q, Han M, Wang S, Zhang Y, Zhang X, Yang N, Wang J, Huang B, Zhang Q, Xiang G, Li W, Zhang D, Wang J, Li X. The Natural Flavonoid Galangin Elicits Apoptosis, Pyroptosis, and Autophagy in Glioblastoma. Front Oncol 2019; 9:942. [PMID: 31612107 PMCID: PMC6776614 DOI: 10.3389/fonc.2019.00942] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 09/06/2019] [Indexed: 12/31/2022] Open
Abstract
Galangin (GG), a flavonoid, elicits a potent antitumor activity in diverse cancers. Here, we evaluated the efficacy of GG in the treatment of human glioblastoma multiforme (GBM) and investigated the molecular basis for its inhibitory effects in the disease. GG inhibited viability and proliferation of GBM cells (U251, U87MG, and A172) in a dose-dependent manner (IC50 = 221.8, 262.5, 273.9 μM, respectively; P < 0.001; EdU, ~40% decrease at 150 μM, P < 0.001), and the number of colonies formed was significantly reduced (at 50 μM, P < 0.001). However, normal human astrocytes were more resistant to its cytotoxic effects (IC50 >450 μM). Annexin-V/PI staining was increased indicating that GG induced apoptosis in GBM cells (26.67 and 30.42%, U87MG and U251, respectively) and associated proteins including BAX and cleaved PARP-1 were increased (~3×). Cells also underwent pyroptosis as determined under phase-contrast microscopy. Knockdown of gasdermin E (GSDME), a protein involved in pyroptosis, alleviated pyroptosis induced by GG through aggravating nuclear DNA damage in GBM cells. Meanwhile, fluorescent GFP-RFP-MAP1LC3B puncta associated with autophagy increased under GG treatment, and transmission electron microscopy confirmed the formation of autophagic vesicles. Inhibition of autophagy enhanced GG-induced apoptosis and pyroptosis in GBM cells. Finally, in an orthotopic xenograft model in nude mice derived from U87MG cells, treatment with GG in combination with an inhibitor of autophagy, chloroquine, suppressed tumor growth, and enhanced survival compared to GG monotherapy (P < 0.05). Our results demonstrated that GG simultaneously induces apoptosis, pytoptosis, and protective autophagy in GBM cells, indicating that combination treatment of GG with autophagy inhibitors may be an effective therapeutic strategy for GBM.
Collapse
Affiliation(s)
- Yang Kong
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Zichao Feng
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China.,Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Anjing Chen
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Qichao Qi
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Mingzhi Han
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China.,Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Shuai Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Yulin Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Xin Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Ning Yang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Jiwei Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Qing Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Guo Xiang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Wenjie Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Di Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China.,Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| |
Collapse
|
19
|
Zhang Z, Yu HJ, Wu S, Huang H, Si LP, Liu HY, Shi L, Zhang HT. Synthesis, characterization, and photodynamic therapy activity of 5,10,15,20-Tetrakis(carboxyl)porphyrin. Bioorg Med Chem 2019; 27:2598-2608. [PMID: 30992204 DOI: 10.1016/j.bmc.2019.03.051] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 01/22/2023]
Abstract
Water-soluble porphyrins are considered promising drug candidates for photodynamic therapy (PDT). This study investigated the PDT activity of a new water-soluble, anionic porphyrin (1-Zn), which possesses four negative charges. The photodynamic anticancer activity of 1-Zn was investigated by the MTT assay, with mTHPC as a positive control. The cellular distribution was determined by fluorescence microscopy. Holographic and phase contrast images were recorded after 1-Zn treatment with a HoloMonitor™ M3 instrument. The inhibition of A549 cell growth achieved by inducing apoptosis was investigated by flow cytometry and fluorescence microscopy. DNA damage was investigated by the comet assay. The expression of apoptosis-related proteins was also measured by western blot assays. 1-Zn had better phototoxicity against A549 cells than HeLa and HepG2 cancer cells. Interestingly, 1-Zn was clearly located almost entirely in the cell cytoplasmic region/organelles. The late apoptotic population was less than 1.0% at baseline in the untreated and only light-treated cells and increased to 40.5% after 1-Zn treatment and irradiation (P < 0.05). 1-Zn triggered significant ROS generation after irradiation, causing ΔΨm disruption (P < 0.01) and DNA damage. 1-Zn induced A549 cell apoptosis via the mitochondrial apoptosis pathway. In addition, 1-Zn bound in the groove of DNA via an outside binding mode by pi-pi stacking and hydrogen bonding. 1-Zn exhibits good photonuclease activity and might serve as a potential photosensitizer (PS) for lung cancer cells.
Collapse
Affiliation(s)
- Zhao Zhang
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang 524023, PR China
| | - Hua-Jun Yu
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang 524023, PR China
| | - Shang Wu
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang 524023, PR China
| | - Hui Huang
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang 524023, PR China
| | - Li-Ping Si
- School of Materials Science and Energy Engineering, Foshan University, Foshan 528000, PR China
| | - Hai-Yang Liu
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, South China University of Technology, Guangzhou 510640, PR China.
| | - Lei Shi
- Department of Chemistry, Guangdong University of Education, Guangzhou 510303, PR China.
| | - Hai-Tao Zhang
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang 524023, PR China.
| |
Collapse
|
20
|
Zhang Z, Yu HJ, Huang H, Wan B, Wu S, Liu HY, Zhang HT. The photocytotoxicity effect of cationic sulfonated corrole towards lung cancer cells: in vitro and in vivo study. Lasers Med Sci 2019; 34:1353-1363. [DOI: 10.1007/s10103-019-02725-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/15/2019] [Indexed: 11/24/2022]
|
21
|
The photodynamic activity and toxicity evaluation of 5,10,15-tris(ethoxylcarbonyl)corrole phosphorus(V) in vivo and in vitro. Eur J Med Chem 2019; 163:779-786. [DOI: 10.1016/j.ejmech.2018.12.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/06/2018] [Accepted: 12/13/2018] [Indexed: 12/17/2022]
|
22
|
Fang D, Xiong Z, Xu J, Yin J, Luo R. Chemopreventive mechanisms of galangin against hepatocellular carcinoma: A review. Biomed Pharmacother 2019; 109:2054-2061. [DOI: 10.1016/j.biopha.2018.09.154] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 09/26/2018] [Accepted: 09/26/2018] [Indexed: 02/07/2023] Open
|
23
|
Galangin Inhibits Thrombin-Induced MMP-9 Expression in SK-N-SH Cells via Protein Kinase-Dependent NF-κB Phosphorylation. Int J Mol Sci 2018; 19:ijms19124084. [PMID: 30562971 PMCID: PMC6321481 DOI: 10.3390/ijms19124084] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/07/2018] [Accepted: 12/14/2018] [Indexed: 12/20/2022] Open
Abstract
Galangin, a member of the flavonol compounds of the flavonoids, could exert anti-inflammatory effects in various cell types. It has been used for the treatment of arthritis, airway inflammation, stroke, and cognitive impairment. Thrombin, one of the regulators of matrix metalloproteinase (MMPs), has been known as a vital factor of physiological and pathological processes, including cell migration, the blood–brain barrier breakdown, brain edema formation, neuroinflammation, and neuronal death. MMP-9 especially may contribute to neurodegenerative diseases. However, the effect of galangin in combating thrombin-induced MMP-9 expression is not well understood in neurons. Therefore, we attempted to explore the molecular mechanisms by which galangin inhibited MMP-9 expression and cell migration induced by thrombin in SK-N-SH cells (a human neuroblastoma cell line). Gelatin zymography, western blot, real-time PCR, and cell migration assay were used to elucidate the inhibitory effects of galangin on the thrmbin-mediated responses. The results showed that galangin markedly attenuated the thrombin-stimulated phosphorylation of proto-oncogene tyrosine-protein kinase (c-Src), proline-rich tyrosine kinase 2 (Pyk2), protein kinase C (PKC)α/β/δ, protein kinase B (Akt), mammalian target of rapamycin (mTOR), p42/p44 mitogen-activated protein kinase (MAPK), Jun amino-terminal kinases (JNK)1/2, p38 MAPK, forkhead box protein O1 (FoxO1), p65, and c-Jun and suppressed MMP-9 expression and cell migration in SK-N-SH cells. Our results concluded that galangin blocked the thrombin-induced MMP-9 expression in SK-N-SH cells via inhibiting c-Src, Pyk2, PKCα/βII/δ, Akt, mTOR, p42/p44 MAPK, JNK1/2, p38 MAPK, FoxO1, c-Jun, and p65 phosphorylation and ultimately attenuated cell migration. Therefore, galangin may be a potential candidate for the management of brain inflammatory diseases.
Collapse
|
24
|
Whang YM, Kim MJ, Cho MJ, Yoon H, Choi YW, Kim TH, Chang IH. Rapamycin enhances growth inhibition on urothelial carcinoma cells through LKB1 deficiency-mediated mitochondrial dysregulation. J Cell Physiol 2018; 234:13083-13096. [PMID: 30549029 DOI: 10.1002/jcp.27979] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 11/21/2018] [Indexed: 12/20/2022]
Abstract
Rapamycin, a mammalian target of rapamycin (mTOR) inhibitor, has significant potential for application in the treatment of urothelial carcinoma (URCa) of the bladder. Previous studies have shown that regulation of the AMP-activated serine/threonine protein kinase (AMPK)-mTOR signaling pathway enhances apoptosis by inducing autophagy or mitophagy in bladder cancer. Alteration of liver kinase B1 (LKB1)-AMPK signaling leads to mitochondrial dysfunction and the accumulation of autophagy-related proteins as a result of mitophagy, resulting in enhanced cell sensitivity to drug treatments. Therefore, we hypothesized that LKB1 deficiency in URCa cells could lead to increased sensitivity to rapamycin by inducing mitochondrial defect-mediated mitophagy. To test this, we established stable LKBI-knockdown URCa cells and analyzed the effects of rapamycin on their growth. Rapamycin enhanced growth inhibition and apoptosis in stable LKB1-knockdown URCa cells and in a xenograft mouse model. In spite of the stable downregulation of LKB1 expression, rapamycin induced AMPK activation in URCa cells, causing loss of the mitochondrial membrane potential, ATP depletion, and ROS accumulation, indicating an alteration of mitochondrial biogenesis. Our findings suggest that the absence of LKB1 can be targeted to induce dysregulated mitochondrial biogenesis by rapamycin treatment in the design of novel therapeutic strategies for bladder cancer.
Collapse
Affiliation(s)
- Young Mi Whang
- Department of Urology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Myeong Joo Kim
- Department of Urology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Min Ji Cho
- Department of Urology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Hoyub Yoon
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Young Wook Choi
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Tae-Hyoung Kim
- Department of Urology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - In Ho Chang
- Department of Urology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
25
|
Luo S, Li Z, Mao L, Chen S, Sun S. Sodium butyrate induces autophagy in colorectal cancer cells through LKB1/AMPK signaling. J Physiol Biochem 2018; 75:53-63. [PMID: 30362049 DOI: 10.1007/s13105-018-0651-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 10/08/2018] [Indexed: 02/06/2023]
Abstract
Butyrate is produced by the fermentation of undigested dietary fibers and acts as the promising candidate for cancer treatment. However, the mechanism underlying sodium butyrate (NaB)-induced autophagy in colorectal cancer is not yet completely understood. The expressions of LC3-II protein and mRNA were detected by western blot and quantitative RT-PCR in colorectal cancer (CRC) cell lines HCT-116 and HT-29, respectively. Autolysosome formation was observed by transmission electron microscope. AMPK and LKB1 were inhibited by chemical inhibitor or siRNAs and confirmed by western blot. NaB elevated the protein and mRNA expressions of LC3 in a dose-dependent manner. NaB treatment increased the formation of autolysosome and expression of phosphorylated liver kinase B1 (LKB1), AMP-activated protein kinase (AMPK), and acetyl-CoA carboxylase (ACC). Treatment with compound C (an inhibitor of AMPK) and siRNA-mediated knockdown of AMPK and LKB1 significantly attenuated NaB-induced autophagy in CRC cells. Collectively, these findings indicated that LKB1 and AMPK are critical for NaB-mediated autophagy and may act as the novel targets for colorectal cancer therapy in the future.
Collapse
Affiliation(s)
- Shunli Luo
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, No.1023 South Sha-Tai Rd, Guangzhou, 510515, People's Republic of China.,School of Laboratory Medicine, Hunan University of Medicine, Huaihua, 418000, People's Republic of China
| | - Ziyin Li
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, No.1023 South Sha-Tai Rd, Guangzhou, 510515, People's Republic of China
| | - Lianzhi Mao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, No.1023 South Sha-Tai Rd, Guangzhou, 510515, People's Republic of China
| | - Siqiang Chen
- Guangzhou Customs District, No.66, Huacheng Avenue, Zhujiang Xincheng, Guangzhou, 510623, People's Republic of China
| | - Suxia Sun
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, No.1023 South Sha-Tai Rd, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
26
|
Abubakar IB, Malami I, Yahaya Y, Sule SM. A review on the ethnomedicinal uses, phytochemistry and pharmacology of Alpinia officinarum Hance. JOURNAL OF ETHNOPHARMACOLOGY 2018; 224:45-62. [PMID: 29803568 DOI: 10.1016/j.jep.2018.05.027] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 05/18/2018] [Accepted: 05/18/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alpinia officinarum Hance is a perennial plant that has been traditionally used for many decades to treat several ailments including inflammation, pain, stomach-ache, cold, amongst others. Pharmacological studies over the years have demonstrated remarkable bioactivities that could be further explored for development of new therapeutic agents against various ailments. AIM OF THE STUDY The paper critically reviewed the ethno-medicinal uses, pharmacology, and phytochemistry of A. officinarum. METHODS Keywords including A. officinarum and its synonyms were searched using electronic databases including ISI web of knowledge, Science direct, Scopus, PubMed, Google scholar and relevant database for Masters and Doctoral theses. RESULTS A. officinarum is prepared in Asia, Turkey, Morocco and Iran as a decoction, infusion or juice as a single preparation or in combination with other herbs, food or drinks for the treatment of general health problems including cold, inflammation, digestive disorders, etc. Pharmacological studies revealed the potent in vitro and in vivo bioactivities of various parts of A. officinarum that include anti-inflammatory, cytotoxicity, homeostasis, lipid regulation, antioxidant, antiviral, antimicrobial, antiosteoporosis, etc. Over 90 phytochemical constituents have been identified and isolated from A. officinarum comprising vastly of phenolic compounds especially diarylheptanoids isolated from the rhizome and considered the most active bioactive components. CONCLUSION In vitro and in vivo studies have confirmed the potency of A. officinarum. However, further studies are required to establish the mechanisms mediating its bioactivities in relation to the medicinal uses as well as investigating any potential toxicity for future clinical studies.
Collapse
Affiliation(s)
- Ibrahim Babangida Abubakar
- Department of Biochemistry, Faculty of Science, Kebbi State University of Science and Technology, PMB 1144 Aliero, Nigeria.
| | - Ibrahim Malami
- Department of Pharmacognosy and Ethnopharmacy, Faculty of Pharmaceutical Sciences, Usmanu Danfodiyo University, PMB 2346 Sokoto, Nigeria.
| | - Yakubu Yahaya
- Department of Pure and Applied Chemistry, Faculty of Science, Kebbi State University of Science and Technology, PMB 1144 Aliero, Nigeria.
| | - Sahabi Manga Sule
- Department of Biological Sciences, Faculty of Science, Kebbi State University of Science and Technology, PMB 1144 Aliero, Nigeria.
| |
Collapse
|
27
|
Mak KK, Tan JJ, Marappan P, Balijepalli MK, Choudhury H, Ramamurthy S, Pichika MR. Galangin’s potential as a functional food ingredient. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.04.054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
28
|
Liu D, You P, Luo Y, Yang M, Liu Y. Galangin Induces Apoptosis in MCF-7 Human Breast Cancer Cells Through Mitochondrial Pathway and Phosphatidylinositol 3-Kinase/Akt Inhibition. Pharmacology 2018; 102:58-66. [PMID: 29879712 DOI: 10.1159/000489564] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 04/24/2018] [Indexed: 12/31/2022]
Abstract
AIMS The study aimed to investigate the molecular mechanism of inhibition of proliferation and apoptosis induction by galangin against MCF-7 human breast cancer cells. METHODS Cell Counting Kit-8 assay was used to assess cell viability and flow cytometry was used to detect cell apoptosis. The expression level of apoptosis-related proteins (cleaved-caspase-9, cleaved-caspase-8, cleaved-caspase-3, Bad, cleaved-Bid, Bcl-2, Bax, p-phosphatidylinositol 3-kinase [PI3K], and p-Akt) and cell cycle-related proteins (cyclin D3, cyclin B1, cyclin-dependent kinases CDK1, CDK2, CDK4, p21, p27, p53) were evaluated by Western blotting. RESULTS Galangin increased the expression of Bax and decreased the expression of Bcl-2 in a concentration-dependent manner, inhibited cell viability, and induced apoptosis. Meanwhile, the expression of cleavage of caspase-9, caspase-8, caspase-3, Bid, and Bad increased significantly while the expression of p-PI3K and p-Akt proteins decreased. In addition, the protein levels of cyclin D3, cyclin B1, CDK1, CDK2, and CDK4 were downregulated while the expression levels of p21, p27, and p53 were upregulated significantly. CONCLUSION Galangin could suppress the viability of MCF-7 cells and induce cell apoptosis via the mitochondrial pathway and PI3K/Akt inhibition as well as cell cycle arrest.
Collapse
Affiliation(s)
- Dan Liu
- School of Pharmacy, Hubei Key Laboratory of Resource Science and Chemistry in Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Pengtao You
- School of Pharmacy, Hubei Key Laboratory of Resource Science and Chemistry in Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Yan Luo
- Affiliated Renhe Hospital of China Three Gorges University, Yichang, China
| | - Min Yang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Yanwen Liu
- School of Pharmacy, Hubei Key Laboratory of Resource Science and Chemistry in Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
29
|
Yildiz G. Integrated multi-omics data analysis identifying novel drug sensitivity-associated molecular targets of hepatocellular carcinoma cells. Oncol Lett 2018; 16:113-122. [PMID: 29930714 PMCID: PMC6006500 DOI: 10.3892/ol.2018.8634] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 02/01/2018] [Indexed: 12/22/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of liver cancer and the third-leading cause of malignancy-associated mortality worldwide. HCC cells are highly resistant to chemotherapeutic agents. Therefore, there are currently only two US Food and Drug Administration-approved drugs available for the treatment of HCC. The objective of the present study was to analyze the results of previously published high-throughput drug screening, and in vitro genomic and transcriptomic data from HCC cell lines, and to integrate the obtained results to define the underlying molecular mechanisms of drug sensitivity and resistance in HCC cells. The results of treatment with 225 different small molecules on 14 different HCC cell lines were retrieved from the Genomics of Drug Sensitivity in Cancer database and analyzed. Cluster analysis using the treatment results determined that HCC cell lines consist of two groups, according to their drug response profiles. Continued analyses of these two groups with Gene Set Enrichment Analysis method revealed 6 treatment-sensitive molecular targets (epidermal growth factor receptor, mechanistic target of rapamycin, deoxyribonucleic acid-dependent protein kinase, the Aurora kinases, Bruton's tyrosine kinase and phosphoinositide 3-kinase; all P<0.05) and partially effective drugs. Genetic and genome-wide gene expression data analyses of the determined targets and their known biological partners revealed 2 somatically mutated and 13 differentially expressed genes, which differed between drug-resistant and drug-sensitive HCC cells. Integration of the obtained data into a short molecular pathway revealed a drug treatment-sensitive signaling axis in HCC cells. In conclusion, the results of the present study provide novel drug sensitivity-associated molecular targets for the development of novel personalized and targeted molecular therapies against HCC.
Collapse
Affiliation(s)
- Gokhan Yildiz
- Department of Medical Biology, Faculty of Medicine, Karadeniz Technical University, Trabzon 61080, Turkey
| |
Collapse
|
30
|
Wang Y, Lin B, Li H, Lan L, Yu H, Wu S, Wu J, Zhang H. Galangin suppresses hepatocellular carcinoma cell proliferation by reversing the Warburg effect. Biomed Pharmacother 2017; 95:1295-1300. [DOI: 10.1016/j.biopha.2017.09.056] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/09/2017] [Accepted: 09/12/2017] [Indexed: 12/20/2022] Open
|
31
|
Deng Y, Zhu L, Cai H, Wang G, Liu B. Autophagic compound database: A resource connecting autophagy-modulating compounds, their potential targets and relevant diseases. Cell Prolif 2017; 51:e12403. [PMID: 29094410 DOI: 10.1111/cpr.12403] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 10/08/2017] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES Autophagy, a highly conserved lysosomal degradation process in eukaryotic cells, can digest long-lived proteins and damaged organelles through vesicular trafficking pathways. Nowadays, mechanisms of autophagy have been gradually elucidated and thus the discovery of small-molecule drugs targeting autophagy has always been drawing much attention. So far, some autophagy-related web servers have been available online to facilitate scientists to obtain the information relevant to autophagy conveniently, such as HADb, CTLPScanner, iLIR server and ncRDeathDB. However, to the best of our knowledge, there is not any web server available about the autophagy-modulating compounds. METHODS According to published articles, all the compounds and their relations with autophagy were anatomized. Subsequently, an online Autophagic Compound Database (ACDB) (http://www.acdbliulab.com/) was constructed, which contained information of 357 compounds with 164 corresponding signalling pathways and potential targets in different diseases. RESULTS We achieved a great deal of information of autophagy-modulating compounds, including compounds, targets/pathways and diseases. ACDB is a valuable resource for users to access to more than 300 curated small-molecule compounds correlated with autophagy. CONCLUSIONS Autophagic compound database will facilitate to the discovery of more novel therapeutic drugs in the near future.
Collapse
Affiliation(s)
- Yiqi Deng
- Department of Laboratory Medicine, Precision Medicine Center, State Key Laboratory of Biotherapy and Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Collaborative Innovation Center, Sichuan University, Chengdu, China
| | - Lingjuan Zhu
- Department of Laboratory Medicine, Precision Medicine Center, State Key Laboratory of Biotherapy and Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Collaborative Innovation Center, Sichuan University, Chengdu, China.,School of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Haoyang Cai
- Center of Growth, Metabolism, and Aging, Key Laboratory of Bio-Resources and Eco-Environment, College of Life Sciences, Sichuan University, Chengdu, China
| | - Guan Wang
- Department of Laboratory Medicine, Precision Medicine Center, State Key Laboratory of Biotherapy and Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Collaborative Innovation Center, Sichuan University, Chengdu, China
| | - Bo Liu
- Department of Laboratory Medicine, Precision Medicine Center, State Key Laboratory of Biotherapy and Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Collaborative Innovation Center, Sichuan University, Chengdu, China
| |
Collapse
|
32
|
Lei D, Zhang F, Yao D, Xiong N, Jiang X, Zhao H. Galangin increases ERK1/2 phosphorylation to decrease ADAM9 expression and prevents invasion in A172 glioma cells. Mol Med Rep 2017; 17:667-673. [PMID: 29115634 DOI: 10.3892/mmr.2017.7920] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Accepted: 06/29/2017] [Indexed: 11/06/2022] Open
Abstract
Galangin (3,5,7‑trihydroxyflavone), is a natural flavonoid present in plants. Galangin is reported to exhibit anti‑cancer properties against various cancer types. The aim of the present study was to display the effects of galangin on glioma and its mechanism of action in A172 human glioma cancer cells. The results clearly indicated that treatment of galangin inhibited A172 cell migration and invasion under non‑toxic doses. A human proteinase array assay was conducted to elucidate the potential effects of galangin, and the obtained results demonstrated that treatment of galangin inhibited ADAM9 protein expression and mRNA expression, that are known to contribute to cancer progression. Sustained extracellular signal‑regulated kinase (Erk)1/2 activation was also monitored, which contributed to ADAM9 protein expression and mRNA inhibition as investigated using western blotting analysis and reverse transcription‑quantitative polymerase chain reaction experiment. Erk1/2 inhibition by inhibitor or small interfering (si)Erk transfection markedly terminated galangin‑inhibited A172 migration and invasion via an Erk1/2 activation mechanism. Collective results suggested that galangin may act as an effective chemotherapeutic agent for glioma cancer depending on its ability to bring about ADAM9 and Erk1/2 activation.
Collapse
Affiliation(s)
- Deqiang Lei
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Fangcheng Zhang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Dongxiao Yao
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Nanxiang Xiong
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Hongyang Zhao
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
33
|
Bacanlı M, Başaran AA, Başaran N. The antioxidant, cytotoxic, and antigenotoxic effects of galangin, puerarin, and ursolic acid in mammalian cells. Drug Chem Toxicol 2017; 40:256-262. [PMID: 27461151 DOI: 10.1080/01480545.2016.1209680] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Phenolic compounds not only contribute to the sensory qualities of fruits and vegetables but also exhibit several health protective properties. Galangin, puerarin, and ursolic acid are commonly used plant phenolics in folk medicine. In this study, the antioxidant capacities of galangin, puerarin, and ursolic acid by the trolox equivalent antioxidant capacity (TEAC) assay and the cytotoxic effects by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay in V79 cells were investigated. The genotoxic potentials of galangin, puerarin, and ursolic acid were evaluated by micronucleus (MN) and alkaline COMET assays in human lymphocytes and in V79 cells. Galangin, puerarin, and ursolic acid (10, 100, 500, 1000, 2000, 5000, 10 000, and 20 000 μM) were found to have antioxidant activities at the studied concentrations. IC50 values of galangin, puerarin, and ursolic acid in V79 cells were found to be 275.48 μM, 2503.712 μM, and 224.85 μM, respectively. Galangin, puerarin, and ursolic acid, at the all concentrations, have not exerted genotoxic effects and galangin, puerarin, and ursolic acid revealed a reduction in the frequency of MN and DNA damage induced by H2O2.
Collapse
Affiliation(s)
- Merve Bacanlı
- a Department of Pharmaceutical Toxicology , Faculty of Pharmacy, Hacettepe University , Ankara , Turkey and
| | - A Ahmet Başaran
- b Department of Pharmacognosy , Faculty of Pharmacy, Hacettepe University , Ankara , Turkey
| | - Nurşen Başaran
- a Department of Pharmaceutical Toxicology , Faculty of Pharmacy, Hacettepe University , Ankara , Turkey and
| |
Collapse
|
34
|
Zhang Z, Wang HH, Yu HJ, Xiong YZ, Zhang HT, Ji LN, Liu HY. Synthesis, characterization and in vitro and in vivo photodynamic activities of a gallium(iii) tris(ethoxycarbonyl)corrole. Dalton Trans 2017; 46:9481-9490. [DOI: 10.1039/c7dt00992e] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A gallium(iii) tris(ethoxycarbonyl)corrole is a highly effective photosensitizer against A549 cancer cells via p38 MAPK signaling cascade pathways.
Collapse
Affiliation(s)
- Zhao Zhang
- Department of Chemistry
- Key Laboratory of Functional Molecular Engineering of Guangdong Province
- South China University of Technology
- Guangzhou
- P. R. China
| | - Hua-Hua Wang
- Department of Chemistry
- Key Laboratory of Functional Molecular Engineering of Guangdong Province
- South China University of Technology
- Guangzhou
- P. R. China
| | - Hua-Jun Yu
- Guangdong Medical University Laboratory Animal Center
- Guang Dong Medical University
- Zhanjiang
- P. R. China
| | - Yu-Zhen Xiong
- Department of Biochemistry and Molecular Biology
- Guang Dong Medical University
- Zhanjiang
- P. R. China
| | - Hai-Tao Zhang
- Department of Biochemistry and Molecular Biology
- Guang Dong Medical University
- Zhanjiang
- P. R. China
| | - Liang-Nian Ji
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry and Chemical Engineering
- Sun Yat-Sen University
- Guangzhou 510275
- China
| | - Hai-Yang Liu
- Department of Chemistry
- Key Laboratory of Functional Molecular Engineering of Guangdong Province
- South China University of Technology
- Guangzhou
- P. R. China
| |
Collapse
|
35
|
Galangin Induces Autophagy via Deacetylation of LC3 by SIRT1 in HepG2 Cells. Sci Rep 2016; 6:30496. [PMID: 27460655 PMCID: PMC4962058 DOI: 10.1038/srep30496] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 07/06/2016] [Indexed: 12/31/2022] Open
Abstract
Galangin suppresses proliferation and induces apoptosis and autophagy in hepatocellular carcinoma (HCC) cells, but the precise mechanism is not clear. In this study, we demonstrated that galangin induced autophagy, enhanced the binding of SIRT1-LC3 and reduced the acetylation of endogenous LC3 in HepG2 cells. But this autophagy was inhibited by inactivation of SIRT1 meanwhile, galangin failed to reduce the acetylation of endogenous LC3 after SIRT1 was knocked-down. Collectively, these findings demonstrate a new mechanism by which galangin induces autophagy via the deacetylation of endogenous LC3 by SIRT1.
Collapse
|
36
|
Zhou Y, Wu Y, Qin Y, Liu L, Wan J, Zou L, Zhang Q, Zhu J, Mi M. Ampelopsin Improves Insulin Resistance by Activating PPARγ and Subsequently Up-Regulating FGF21-AMPK Signaling Pathway. PLoS One 2016; 11:e0159191. [PMID: 27391974 PMCID: PMC4938387 DOI: 10.1371/journal.pone.0159191] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 06/28/2016] [Indexed: 12/24/2022] Open
Abstract
Ampelopsin (APL), a major bioactive constituent of Ampelopsis grossedentata, exerts a number of biological effects. Here, we explored the anti-diabetic activity of APL and elucidate the underlying mechanism of this action. In palmitate-induced insulin resistance of L6 myotubes, APL treatment markedly up- regulated phosphorylated insulin receptor substrate-1 and protein kinase B, along with a corresponding increase of glucose uptake capacity. APL treatment also increased expressions of fibroblast growth factor (FGF21) and phosphorylated adenosine 5’-monophosphate -activated protein kinase (p-AMPK), however inhibiting AMPK by Compound C or AMPK siRNA, or blockage of FGF21 by FGF21 siRNA, obviously weakened APL -induced increases of FGF21 and p-AMPK as well as glucose uptake capacity in palmitate -pretreated L6 myotubes. Furthermore, APL could activate PPAR γ resulting in increases of glucose uptake capacity and expressions of FGF21 and p-AMPK in palmitate -pretreated L6 myotubes, whereas all those effects were obviously abolished by addition of GW9662, a specific inhibitor of peroxisome proliferator- activated receptor –γ (PPARγ), and PPARγsiRNA. Using molecular modeling and the luciferase reporter assays, we observed that APL could dock with the catalytic domain of PPARγ and dose-dependently up-regulate PPARγ activity. In summary, APL maybe a potential agonist of PPARγ and promotes insulin sensitization by activating PPARγ and subsequently regulating FGF21- AMPK signaling pathway. These results provide new insights into the protective health effects of APL, especially for the treatment of Type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Yong Zhou
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Research Center for Medical Nutrition, Chongqing, 400038, China
| | - Ying Wu
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Research Center for Medical Nutrition, Chongqing, 400038, China
| | - Yu Qin
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Research Center for Medical Nutrition, Chongqing, 400038, China
| | - Lei Liu
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Research Center for Medical Nutrition, Chongqing, 400038, China
| | - Jing Wan
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Research Center for Medical Nutrition, Chongqing, 400038, China
| | - Lingyun Zou
- College of Basic Medical Sciences, Third Military Medical University, Chongqing, 400038, China
| | - Qianyong Zhang
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Research Center for Medical Nutrition, Chongqing, 400038, China
| | - Jundong Zhu
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Research Center for Medical Nutrition, Chongqing, 400038, China
| | - Mantian Mi
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Research Center for Medical Nutrition, Chongqing, 400038, China
- * E-mail:
| |
Collapse
|
37
|
Han MA, Lee DH, Woo SM, Seo BR, Min KJ, Kim S, Park JW, Kim SH, Choi YH, Kwon TK. Galangin sensitizes TRAIL-induced apoptosis through down-regulation of anti-apoptotic proteins in renal carcinoma Caki cells. Sci Rep 2016; 6:18642. [PMID: 26725939 PMCID: PMC4698673 DOI: 10.1038/srep18642] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 11/23/2015] [Indexed: 12/17/2022] Open
Abstract
Galangin, bioflavonoids, has been shown anti-cancer properties in various cancer cells. In this study, we investigated whether galangin could enhance TRAIL-mediated apoptosis in TRAIL resistant renal carcinoma Caki cells. Galangin alone and TRAIL alone had no effect on apoptosis, while combined treatment with galangin and TRAIL significantly induced apoptosis in renal carcinoma (Caki, ACHN and A498) but not normal cells (normal mouse kidney cells and human normal mesangial cells). Galangin induced down-regulation of Bcl-2 protein at the transcriptional level via inhibition of NF-κB activation but not p53 pathway. Furthermore, galangin induced down-regulation of cFLIP, Mcl-1 and survivin expression at the post-translational levels, and the over-expression of Bcl-2, cFLIP, Mcl-1 and survivin markedly reduced galangin-induced TRAIL sensitization. In addition, galangin increased proteasome activity, but galangin had no effect on expression of proteasome subunits (PSMA5 and PSMD4). In conclusion, our investigation suggests that galangin is a potent candidate for sensitizer of TRAIL resistant cancer cell therapy.
Collapse
Affiliation(s)
- Min Ae Han
- Department of Immunology, School of Medicine, Keimyung University, 2800 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, South Korea
| | - Dong Hee Lee
- Department of Immunology, School of Medicine, Keimyung University, 2800 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, South Korea
| | - Seon Min Woo
- Department of Immunology, School of Medicine, Keimyung University, 2800 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, South Korea
| | - Bo Ram Seo
- Department of Immunology, School of Medicine, Keimyung University, 2800 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, South Korea
| | - Kyoung-Jin Min
- Department of Immunology, School of Medicine, Keimyung University, 2800 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, South Korea
| | - Shin Kim
- Department of Immunology, School of Medicine, Keimyung University, 2800 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, South Korea
| | - Jong-Wook Park
- Department of Immunology, School of Medicine, Keimyung University, 2800 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, South Korea
| | - Sang Hyun Kim
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Yung Hyun Choi
- Department of Biochemistry, College of Oriental Medicine, Dong-Eui University, Busan, South Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, 2800 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, South Korea
| |
Collapse
|
38
|
Feng WH, Zhang HH, Zhang Y, Sun M, Niu JL. Determination of galangin in rat plasma by UPLC and pharmacokinetic study. J Chromatogr B Analyt Technol Biomed Life Sci 2015; 998-999:26-30. [DOI: 10.1016/j.jchromb.2015.06.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 06/18/2015] [Accepted: 06/21/2015] [Indexed: 12/26/2022]
|
39
|
Shi L, Zhang T, Liang X, Hu Q, Huang J, Zhou Y, Chen M, Zhang Q, Zhu J, Mi M. Dihydromyricetin improves skeletal muscle insulin resistance by inducing autophagy via the AMPK signaling pathway. Mol Cell Endocrinol 2015; 409:92-102. [PMID: 25797177 DOI: 10.1016/j.mce.2015.03.009] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 03/12/2015] [Accepted: 03/16/2015] [Indexed: 01/10/2023]
Abstract
Skeletal muscle insulin resistance (SMIR) plays an important role in the pathogenesis of type 2 diabetes. Dihydromyricetin (DHM), a natural flavonoid, exerts various bioactivities including anti-oxidative and hepatoprotective effects. Herein, we intended to determine the effect of DHM on SMIR and the underlying mechanisms. We found that DHM increased the expression of phosphorylated insulin receptor substrate-1, phosphorylated Akt and glucose uptake capacity in palmitate-treated L6 myotubes under insulin-stimulated conditions. The expression of light chain 3, Beclin 1, autophagy-related gene 5 (Atg5), the degradation of sequestosome 1 and the formation of autophagosomes were also upregulated by DHM. Suppression of autophagy by 3-methyladenine and bafilomycin A1 or Atg5 and Beclin1 siRNA abolished the favorable effects of DHM on SMIR. Furthermore, DHM increased the levels of phosphorylated AMP-activated protein kinase (AMPK) and Ulk1, and decreased phosphorylated mTOR levels. AMPK inhibitor compound C (CC) and AMPK siRNA abrogated DHM-induced autophagy, subsequently suppressed DHM-induced SMIR improvement. Additionally, DHM inhibited the activity of F1F0-ATPase thereby activating AMPK. Finally, the results of in vivo study conducted in high fat diet-fed rats were consistent with the findings of in vitro study. In conclusion, DHM improved SMIR by inducing autophagy via the activation of AMPK signaling pathway.
Collapse
Affiliation(s)
- Linying Shi
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing 400038, China
| | - Ting Zhang
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing 400038, China
| | - Xinyu Liang
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing 400038, China
| | - Qin Hu
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing 400038, China
| | - Juan Huang
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing 400038, China
| | - Yong Zhou
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing 400038, China
| | - Mingliang Chen
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing 400038, China
| | - Qianyong Zhang
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing 400038, China
| | - Jundong Zhu
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing 400038, China.
| | - Mantian Mi
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing 400038, China.
| |
Collapse
|
40
|
Huang H, Chen AY, Rojanasakul Y, Ye X, Rankin GO, Chen YC. Dietary compounds galangin and myricetin suppress ovarian cancer cell angiogenesis. J Funct Foods 2015; 15:464-475. [PMID: 26113875 DOI: 10.1016/j.jff.2015.03.051] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Galangin and myricetin are flavonoids isolated from vegetables and fruits which exhibit anti-proliferative activity in human cancer cells. In this study, their anti-angiogenic effects were investigated with in vitro (HUVEC) and in vivo (CAM) models, which showed that galangin and myricetin inhibited angiogenesis induced by OVCAR-3 cells. The molecular mechanisms through which galangin and myricetin suppress angiogenesis were also studied. It was observed that galangin and myricetin inhibited secretion of the key angiogenesis mediator vascular endothelial growth factor (VEGF) and decreased levels of p-Akt, p-70S6K and hypoxia-inducible factor-1α (HIF-1α) proteins in A2780/CP70 and OVCAR-3 cells. Transient transfection experiments showed that galangin and myricetin inhibited secretion of VEGF by the Akt/p70S6K/ HIF-1α pathway. Moreover, a novel pathway, p21/HIF-1α/VEGF, was found to be involved in the inhibitory effect of myricetin on angiogenesis in OVCAR-3 cells. These data suggest that galangin and myricetin might serve as potential anti-angiogenic agents in the prevention of ovarian cancers dependent on new blood vessel networks.
Collapse
Affiliation(s)
- Haizhi Huang
- College of Biosystems Engineering and Food Science, Fuli Institute of Food Science, Zhejiang University, China ; College of Science, Technology & Mathematics, Alderson Broaddus University, Philippi, WV 26416, USA
| | - Allen Y Chen
- Department of Pharmaceutical Science, West Virginia University, Morgantown, WV 26506, USA
| | - Yon Rojanasakul
- Department of Pharmaceutical Science, West Virginia University, Morgantown, WV 26506, USA
| | - Xingqian Ye
- College of Biosystems Engineering and Food Science, Fuli Institute of Food Science, Zhejiang University, China
| | - Gary O Rankin
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Yi Charlie Chen
- College of Science, Technology & Mathematics, Alderson Broaddus University, Philippi, WV 26416, USA
| |
Collapse
|
41
|
Chaurasiya ND, Ibrahim MA, Muhammad I, Walker LA, Tekwani BL. Monoamine oxidase inhibitory constituents of propolis: kinetics and mechanism of inhibition of recombinant human MAO-A and MAO-B. Molecules 2014; 19:18936-52. [PMID: 25412041 PMCID: PMC6271006 DOI: 10.3390/molecules191118936] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 10/07/2014] [Accepted: 10/11/2014] [Indexed: 11/29/2022] Open
Abstract
Propolis is the resinous material that bees gather from leaf buds, flowers and vegetables. Propolis extracts contain constituents with a broad spectra of pharmacological properties and are important ingredients of popular dietary supplements. Propolis extracts were evaluated in vitro for inhibition of recombinant human monoamine oxidase (MAO)-A and MAO-B. The dichloromethane extract of propolis showed potent inhibition of human MAO-A and MAO-B. Further fractionation identified the most active fractions as rich in flavonoids. Galangin and apigenin were identified as the principal MAO-inhibitory constituents. Inhibition of MAO-A by galangin was about 36 times more selective than MAO-B, while apigenin selectivity for MAO-A vs. MAO-B was about 1.7 fold. Apigenin inhibited MAO-B significantly more potently than galangin. Galangin and apigenin were further evaluated for kinetic characteristics and the mechanism for the enzymes’ inhibition. Binding of galangin and apigenin with MAO-A and -B was not time-dependent and was reversible, as suggested by enzyme-inhibitor binding and dissociation-dialysis assay. The inhibition kinetics studies suggested that galangin and apigenin inhibited MAO-A and -B by a competitive mechanism. Presence of prominent MAO inhibitory constituents in propolis products suggests their potential for eliciting pharmacological effects that might be useful in depression or other neurological disorders. The results may also have important implications in drug-dietary supplement interactions.
Collapse
Affiliation(s)
- Narayan D Chaurasiya
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA.
| | - Mohamed A Ibrahim
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA.
| | - Ilias Muhammad
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA.
| | - Larry A Walker
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA.
| | - Babu L Tekwani
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA.
| |
Collapse
|
42
|
Wang Y, Wu J, Lin B, Li X, Zhang H, Ding H, Chen X, Lan L, Luo H. Galangin suppresses HepG2 cell proliferation by activating the TGF-β receptor/Smad pathway. Toxicology 2014; 326:9-17. [PMID: 25268046 DOI: 10.1016/j.tox.2014.09.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 09/21/2014] [Accepted: 09/22/2014] [Indexed: 12/19/2022]
Abstract
Galangin can suppress hepatocellular carcinoma (HCC) cell proliferation. In this study, we demonstrated that galangin induced autophagy by activating the transforming growth factor (TGF)-β receptor/Smad pathway and increased TGF-β receptor I (RI), TGF-βRII, Smad1, Smad2, Smad3 and Smad4 levels but decreased Smad6 and Smad7 levels. Autophagy induced by galangin appears to depend on the TGF-β receptor/Smad signalling pathway because the down-regulation of Smad4 by siRNA or inhibition of TGF-β receptor activation by LY2109761 blocked galangin-induced autophagy. The down-regulation of Beclin1, autophagy-related gene (ATG) 16L, ATG12 and ATG3 restored HepG2 cell proliferation and prevented galangin-induced apoptosis. Our findings indicate a novel mechanism for galangin-induced autophagy via activation of the TGF-β receptor/Smad pathway. The induction of autophagy thus reflects the anti-proliferation effect of galangin on HCC cells.
Collapse
Affiliation(s)
- Yajun Wang
- Department of Biochemistry and Molecular Biology, Guangdong Medical College, Dongguan, Guangdong 523808, China; Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, China
| | - Jun Wu
- Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, China
| | - Biyun Lin
- Department of Biochemistry and Molecular Biology, Guangdong Medical College, Dongguan, Guangdong 523808, China
| | - Xv Li
- Department of Biochemistry and Molecular Biology, Guangdong Medical College, Dongguan, Guangdong 523808, China
| | - Haitao Zhang
- Department of Biochemistry and Molecular Biology, Guangdong Medical College, Dongguan, Guangdong 523808, China.
| | - Hang Ding
- Department of Biochemistry and Molecular Biology, Guangdong Medical College, Dongguan, Guangdong 523808, China
| | - Xiaoyi Chen
- Department of Biochemistry and Molecular Biology, Guangdong Medical College, Dongguan, Guangdong 523808, China
| | - Liubo Lan
- Department of Biochemistry and Molecular Biology, Guangdong Medical College, Dongguan, Guangdong 523808, China
| | - Hui Luo
- Department of Chemistry, Guangdong Medical College, Zhanjiang, Guangdong 524023, China.
| |
Collapse
|
43
|
Huo SX, Liu XM, Ge CH, Gao L, Peng XM, Zhao PP, Yan M. The Effects of Galangin on a Mouse Model of Vitiligo Induced by Hydroquinone. Phytother Res 2014; 28:1533-8. [DOI: 10.1002/ptr.5161] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 04/10/2014] [Indexed: 11/11/2022]
Affiliation(s)
- Shi-Xia Huo
- Department of Cell and Molecular Laboratory Xinjiang Institute of Traditional Uighur Medicine; Xinjiang Laboratory of Uighur Medical Prescription; Urumqi Xinjiang 830049 China
| | - Xin-Ming Liu
- Department of Cell and Molecular Laboratory Xinjiang Institute of Traditional Uighur Medicine; Xinjiang Laboratory of Uighur Medical Prescription; Urumqi Xinjiang 830049 China
- Institute of Medicinal Plant Development; Chinese Academy of Medical Sciences; Beijing 100193 China
| | - Chun-Hui Ge
- Department of Cell and Molecular Laboratory Xinjiang Institute of Traditional Uighur Medicine; Xinjiang Laboratory of Uighur Medical Prescription; Urumqi Xinjiang 830049 China
| | - Li Gao
- Department of Cell and Molecular Laboratory Xinjiang Institute of Traditional Uighur Medicine; Xinjiang Laboratory of Uighur Medical Prescription; Urumqi Xinjiang 830049 China
| | - Xiao-Ming Peng
- Department of Cell and Molecular Laboratory Xinjiang Institute of Traditional Uighur Medicine; Xinjiang Laboratory of Uighur Medical Prescription; Urumqi Xinjiang 830049 China
| | - Ping-Ping Zhao
- Department of Cell and Molecular Laboratory Xinjiang Institute of Traditional Uighur Medicine; Xinjiang Laboratory of Uighur Medical Prescription; Urumqi Xinjiang 830049 China
| | - Ming Yan
- Department of Cell and Molecular Laboratory Xinjiang Institute of Traditional Uighur Medicine; Xinjiang Laboratory of Uighur Medical Prescription; Urumqi Xinjiang 830049 China
| |
Collapse
|
44
|
Xiao J, Niu G, Yin S, Xie S, Li Y, Nie D, Ma L, Wang X, Wu Y. The role of AMP-activated protein kinase in quercetin-induced apoptosis of HL-60 cells. Acta Biochim Biophys Sin (Shanghai) 2014; 46:394-400. [PMID: 24705122 DOI: 10.1093/abbs/gmu014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Our previous studies have shown that quercetin inhibits Cox-2 and Bcl-2 expressions, and induces human leukemia HL-60 cell apoptosis. In order to investigate the role of AMP-activated protein kinase (AMPK) on quercetin-induced apoptosis of HL-60 cells, we used flow cytometry to detect cell apoptosis. The expressions of LKB1, phosphorylated AMPK (p-AMPK), and Cox-2 protein were detected in HL-60 cells and normal peripheral blood mononuclear cells (PBMCs) by western blot. The expressions of LKB1, p-AMPK, and Cox-2 were detected in HL-60 cells after culture with quercetin. The expressions of p-AMPK were detected in HL-60 cells after culture with AMPK inhibitor Compound C. Then, the expressions of LKB1, p-AMPK, and Cox-2 were detected in HL-60 cells after culture with quercetin alone or quercetin + Compound C. It was found that there was no significant difference in LKB1 between PBMCs and HL-60. p-AMPK in PBMCs was higher than that in HL-60, while Cox-2 was lower. After culture of HL-60 with quercetin, p-AMPK was increased, Cox-2 was decreased, but LKB1 remained unchanged. After culture of HL-60 with Compound C, p-AMPK was decreased. There was no significant difference in LKB1 between the quercetin-alone and the quercetin + Compound C groups. p-AMPK decreased more significantly, while Cox-2 increased more significantly in the quercetin + Compound C groups than those in the quercetin-alone groups. Taken together, these findings suggested that quercetin activates AMPK expression in HL-60 cells independent of LKB1 activation, inhibits Cox-2 expression by activating AMPK, and further regulates the Bcl-2-dependent pathways of apoptosis to exert its anti-leukemia effect.
Collapse
Affiliation(s)
- Jie Xiao
- Department of Hematology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | | | | | | | | | | | | | | | | |
Collapse
|