1
|
Zarezadeh SM, Sharafi AM, Erabi G, Tabashiri A, Teymouri N, Mehrabi H, Golzan SA, Faridzadeh A, Abdollahifar Z, Sami N, Arabpour J, Rahimi Z, Ansari A, Abbasi MR, Azizi N, Tamimi A, Poudineh M, Deravi N. Natural STAT3 Inhibitors for Cancer Treatment: A Comprehensive Literature Review. Recent Pat Anticancer Drug Discov 2024; 19:403-502. [PMID: 37534488 DOI: 10.2174/1574892818666230803100554] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 06/05/2023] [Accepted: 06/09/2023] [Indexed: 08/04/2023]
Abstract
Cancer is one of the leading causes of mortality and morbidity worldwide, affecting millions of people physically and financially every year. Over time, many anticancer treatments have been proposed and studied, including synthetic compound consumption, surgical procedures, or grueling chemotherapy. Although these treatments have improved the daily life quality of patients and increased their survival rate and life expectancy, they have also shown significant drawbacks, including staggering costs, multiple side effects, and difficulty in compliance and adherence to treatment. Therefore, natural compounds have been considered a possible key to overcoming these problems in recent years, and thorough research has been done to assess their effectiveness. In these studies, scientists have discovered a meaningful interaction between several natural materials and signal transducer and activator of transcription 3 molecules. STAT3 is a transcriptional protein that is vital for cell growth and survival. Mechanistic studies have established that activated STAT3 can increase cancer cell proliferation and invasion while reducing anticancer immunity. Thus, inhibiting STAT3 signaling by natural compounds has become one of the favorite research topics and an attractive target for developing novel cancer treatments. In the present article, we intend to comprehensively review the latest knowledge about the effects of various organic compounds on inhibiting the STAT3 signaling pathway to cure different cancer diseases.
Collapse
Affiliation(s)
- Seyed Mahdi Zarezadeh
- Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Mohammad Sharafi
- Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gisou Erabi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Arefeh Tabashiri
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Navid Teymouri
- Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hoda Mehrabi
- Student Research Committee, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Seyyed Amirhossein Golzan
- Student Research Committee, Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arezoo Faridzadeh
- Department of Immunology and Allergy, Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Abdollahifar
- Student Research Committee, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Nafiseh Sami
- Student Research Committee, Tehran Medical Sciences, Islamic Azad University Medical Branch of Tehran, Tehran, Iran
| | - Javad Arabpour
- Department of Microbiology, Faculty of New Sciences, Islamic Azad University Medical Branch of Tehran, Tehran, Iran
| | - Zahra Rahimi
- School of Medicine, Zanjan University of Medical Sciences Zanjan, Iran
| | - Arina Ansari
- Student Research Committee, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | | | - Nima Azizi
- Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Niloofar Deravi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Huong NT, Son NT. Icaritin: A phytomolecule with enormous pharmacological values. PHYTOCHEMISTRY 2023:113772. [PMID: 37356700 DOI: 10.1016/j.phytochem.2023.113772] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 05/24/2023] [Accepted: 06/10/2023] [Indexed: 06/27/2023]
Abstract
Pharmacological studies on flavonoids have always drawn much interest for many years. Icaritin (ICT), a representative flavone containing an 8-prenyl group, is a principal compound detected in medicinal plants of the genus Epimedum, the family Berberidaceae. Experimental results in the phytochemistry and pharmacology of this molecule are abundant now, but a deep overview has not been carried out. The goal of this review is to provide an insight into the natural observation, biosynthesis, biotransformation, synthesis, pharmacology, and pharmacokinetics of prenyl flavone ICT. The relevant data on ICT was collected from bibliographic sources, like Google Scholar, Web of Science, Sci-Finder, and various published journals. "Icaritin" alone or in combination is the main keyword to seek for references, and references have been updated till now. ICT is among the characteristic phytomolecules of Epimedum plants. Bacteria monitored its biosynthesis and biotransformation, while this agent was rapidly synthesized from phloroglucinol by microwave-assistance Claisen rearrangement. ICT is a potential agent in numerous in vitro and in vivo pharmacological records, which demonstrated its role in cancer treatments via apoptotic-related mechanisms. It also brings in various health benefits since it reduced harmful effects on the liver, lung, heart, bone, blood, and skin, and improved immune responses. Pharmacokinetic outcomes indicated that its metabolic pathway involved hydration, hydroxylation, dehydrogenation, glycosylation, and glucuronidation. Molecule mechanisms of action at a cellular level are predominant, but clinical studies are expected to get more. Structure-activity relationship records seem insufficient, and the studies on nano-combined approaches to improve its soluble property in living bodied medium are needed.
Collapse
Affiliation(s)
- Nguyen Thi Huong
- Faculty of Chemical Technology, Hanoi University of Industry, Hanoi, Viet Nam
| | - Ninh The Son
- Institute of Chemistry, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet, Caugiay, Hanoi, Viet Nam.
| |
Collapse
|
3
|
Zhang C, Xu H, Sui X, Chen L, Chen B, Lv H, Wang S, Wang X. Icaritin inhibits PLK1 to activate DNA damage response in NK/T cell lymphoma and increases sensitivity to GELOX regime. Mol Ther Oncolytics 2022; 25:288-304. [PMID: 35663228 PMCID: PMC9127125 DOI: 10.1016/j.omto.2022.04.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 04/29/2022] [Indexed: 02/07/2023] Open
Abstract
Natural killer/T cell lymphoma (NKTCL) is a highly aggressive subtype of non-Hodgkin lymphoma. Gemcitabine, oxaliplatin, and L-asparaginase (GELOX) is one of the first-line chemotherapy regimens of NKTCL. Yet, the prognosis of NKTCL is poor. Icaritin is an herb-derived monomer from icariin with antitumor effects. We found that icaritin induced proliferation inhibition and apoptosis of NKTCL both in vitro and in vivo. Moreover, icaritin inhibited the dissemination of NKTCL in vivo. RNA sequencing revealed the Polo-like kinase 1 (PLK1) gene and DNA damage response (DDR) as the targets of icaritin. Mechanistically, icaritin inhibited PLK1 to promote checkpoint kinase 2 (Chk2) homodimerization and its T387 phosphorylation, which further activated p53, leading to the activation of the DDR pathway. Moreover, inhibiting PLK1 increased Forkhead box O3a nuclear localization, the latter of which activated ataxia telangiectasia mutated (ATM), an early sensor of DNA damage. Then ATM phosphorylated Chk2 T68 and initiated Chk2 activation. Remarkably, the combined treatment of icaritin and GELOX achieved better antitumor efficacy than single treatment in vivo. In summary, our results proved the efficacy of icaritin treating NKTCL, provided insights into its antitumor molecular mechanism, and revealed the application value of icaritin in facilitating clinical NKTCL treatment.
Collapse
Affiliation(s)
- Canjing Zhang
- Key Laboratory of Medical Molecular Virology of Ministry of Education & Ministry of Health, School of Basic Medical Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, People's Republic of China.,Laboratory of Medical Molecular Biology, Experimental Teaching Center, School of Basic Medical Sciences, Fudan University, Shanghai 200032, People's Republic of China
| | - Huiwen Xu
- Key Laboratory of Medical Molecular Virology of Ministry of Education & Ministry of Health, School of Basic Medical Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, People's Republic of China.,Department of Hematology, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China
| | - Xianxian Sui
- Laboratory of Medical Molecular Biology, Experimental Teaching Center, School of Basic Medical Sciences, Fudan University, Shanghai 200032, People's Republic of China
| | - Lina Chen
- Key Laboratory of Medical Molecular Virology of Ministry of Education & Ministry of Health, School of Basic Medical Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, People's Republic of China
| | - Bobin Chen
- Department of Hematology, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China
| | - Haozhen Lv
- Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Songmei Wang
- Laboratory of Medical Molecular Biology, Experimental Teaching Center, School of Basic Medical Sciences, Fudan University, Shanghai 200032, People's Republic of China
| | - Xuanyi Wang
- Key Laboratory of Medical Molecular Virology of Ministry of Education & Ministry of Health, School of Basic Medical Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, People's Republic of China
| |
Collapse
|
4
|
Gao L, Zhang SQ. Antiosteoporosis Effects, Pharmacokinetics, and Drug Delivery Systems of Icaritin: Advances and Prospects. Pharmaceuticals (Basel) 2022; 15:397. [PMID: 35455393 PMCID: PMC9032325 DOI: 10.3390/ph15040397] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/10/2022] [Accepted: 03/22/2022] [Indexed: 12/11/2022] Open
Abstract
Osteoporosis is a systemic skeletal disorder affecting over 200 million people worldwide and contributes dramatically to global healthcare costs. Available anti-osteoporotic drug treatments including hormone replacement therapy, anabolic agents, and bisphosphonates often cause adverse events which limit their long-term use. Therefore, the application of natural products has been proposed as an alternative therapy strategy. Icaritin (ICT) is not only an enzyme-hydrolyzed product of icariin but also an intestinal metabolite of eight major flavonoids of the traditional Chinese medicinal plant Epimedium with extensive pharmacological activities, such as strengthening the kidney and reinforcing the bone. ICT displays several therapeutic effects, including osteoporosis prevention, neuroprotection, antitumor, cardiovascular protection, anti-inflammation, and immune-protective effect. ICT inhibits bone resorption activity of osteoclasts and stimulates osteogenic differentiation and maturation of bone marrow stromal progenitor cells and osteoblasts. As for the mechanisms of effect, ICT regulates relative activities of two transcription factors Runx2 and PPARγ, determines the differentiation of MSCs into osteoblasts, increases mRNA expression of OPG, and inhibits mRNA expression of RANKL. Poor water solubility, high lipophilicity, and unfavorable pharmacokinetic properties of ICT restrict its anti-osteoporotic effects, and novel drug delivery systems are explored to overcome intrinsic limitations of ICT. The paper focuses on osteogenic effects and mechanisms, pharmacokinetics and delivery systems of ICT, and highlights bone-targeting strategies to concentrate ICT on the ideal specific site of bone. ICT is a promising potential novel therapeutic agent for osteoporosis.
Collapse
Affiliation(s)
- Lifang Gao
- School of Public Health, Capital Medical University, 10 Youanmenwai Xitiao, Beijing 100069, China;
| | - Shuang-Qing Zhang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, 27 Nanwei Road, Beijing 100050, China
| |
Collapse
|
5
|
Zeng Y, Xiong Y, Yang T, Wang Y, Zeng J, Zhou S, Luo Y, Li L. Icariin and its metabolites as potential protective phytochemicals against cardiovascular disease: From effects to molecular mechanisms. Biomed Pharmacother 2022; 147:112642. [DOI: 10.1016/j.biopha.2022.112642] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 02/06/2023] Open
|
6
|
Liu S, Zhang C, Hao J, Liu Y, Zheng S, Yang C, Yang J, Wu H. Icariin Promotes In Vitro Cardiomyocyte Proliferation and Differentiation in Human Bone Marrow-Derived Mesenchymal Stem Cells. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Mesenchymal stem cells (MSCs) are the excellent candidates in myocardial regeneration given their easy accessibility, low immunogenicity and high potential for cardiomyocyte differentiation. This work focused on investigating the role of icariin, a main active component of the Traditional
Chinese herb epimedium, in human bone marrow-derived MSCs (BMSCs) proliferation and differentiation into cardiomyocytes In Vitro. Human BMSCs were cultivated In Vitro, and MTT assay was conducted to measure their proliferation. On this basis, we selected the optimal icariin dose
for promoting the proliferation to induce cardiomyocyte differentiation of MSCs, which were pretreated with or without 5-azacytidine (5-Aza). Cardiac-specific cardiac troponin I (cTnI) and connexin 43 (Cx43)-positive cells were detected by immunofluorescent staining. The differentiation ratio
of MSCs was examined by flow cytometry. This study measured early cardiac transcription factors (TFs) Nkx2.5 and GATA4 levels through RT-PCR and Western blotting (WB). As a result, icariin increased MSC proliferation dependent on its dose, and the optimal dose was determined to be 80 μg/l.
Furthermore, MSCs showed minimal cardiomyogenic differentiation when induced by icariin alone as confirmed by the expression of cardiac-related markers. Moreover, a synergic interaction was observed when icariin and 5-Aza cooperated to induce cardiomyocyte differentiation of MSCs. In conclusion,
Icariin stimulates proliferation and facilitates cardiomyocyte differentiation of MSCs In Vitro and may be potentially used as a new method for enhancing the MSCs efficacy in cardiovascular disease.
Collapse
Affiliation(s)
- Shaoying Liu
- Department of Cardiology, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, Dongjie 3, Yongding Road, Haidian District, Beijing 100039, China
| | - Chengying Zhang
- Department of Cardiology, Traditional Chinese Medical Hospital of Beijing Huairou, 1 Houheng Jie, Huairou District, Beijing 101400, China
| | - Jing Hao
- Jimenli Community Health Service Center, Jimenli Community, Beisanhuan West Road, Haidian District, Beijing 100191, China
| | - Yuna Liu
- Department of Laboratory, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, Dongjie 3, Yongding Road, Haidian District, Beijing 100039, China
| | - Sidao Zheng
- Department of Cardiology, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, Dongjie 3, Yongding Road, Haidian District, Beijing 100039, China
| | - Cui Yang
- Department of Cardiology, Traditional Chinese Medical Hospital of Beijing Huairou, 1 Houheng Jie, Huairou District, Beijing 101400, China
| | - Jiyuan Yang
- Department of Cardiology, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, Dongjie 3, Yongding Road, Haidian District, Beijing 100039, China
| | - Hongjin Wu
- Beijing Haidian Hospital, Haidian Section of Peking University Third Hospital, 29 Zhongguancun Dajie, Haidian District, Beijing 100080, China
| |
Collapse
|
7
|
Lu Y, Ding Y, Wei J, He S, Liu X, Pan H, Yuan B, Liu Q, Zhang J. Anticancer effects of Traditional Chinese Medicine on epithelial-mesenchymal transition(EMT) in breast cancer: Cellular and molecular targets. Eur J Pharmacol 2021; 907:174275. [PMID: 34214582 DOI: 10.1016/j.ejphar.2021.174275] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 06/15/2021] [Accepted: 06/18/2021] [Indexed: 12/31/2022]
Abstract
Breast cancer is a malignant epithelial tumor of ductal or lobular origin. Breast cancer remains the most frequently diagnosed invasive cancer in women and is the leading cause of cancer-associated mortality worldwide. Epithelial-mesenchymal transition (EMT), a phenotypic process of conversion from epithelial to mesenchymal cells, allows tumor cells to acquire infiltration and metastasization properties. Therapies directed at pathways, which are primarily involved in malignant transformation, can lead to clinical implications. In recent years, EMT has gained increasing attention as a potential therapeutic target in cancer therapy. Moreover, for the past few decades, increasing numbers of studies have suggested that Traditional Chinese Medicine(TCM) compounds can significantly inhibit the growth and development of breast cancer cells through the inhibition of EMT in breast cancer cells. This review discusses some essential signaling pathways associated with EMT and summarizes the effects and mechanism of TCM components on that inhibit EMT in breast cancer therapy.
Collapse
Affiliation(s)
- Yiran Lu
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin, 130062, PR China
| | - Yu Ding
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin, 130062, PR China
| | - Jiahui Wei
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin, 130062, PR China
| | - Song He
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin, 130062, PR China
| | - Xinmiao Liu
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin, 130062, PR China
| | - Huihao Pan
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin, 130062, PR China
| | - Bao Yuan
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin, 130062, PR China
| | - Qing Liu
- The Second Clinical School of Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China.
| | - Jiabao Zhang
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin, 130062, PR China.
| |
Collapse
|
8
|
Icaritin inhibits lung cancer-induced osteoclastogenesis by suppressing the expression of IL-6 and TNF-a and through AMPK/mTOR signaling pathway. Anticancer Drugs 2021; 31:1004-1011. [PMID: 32701561 DOI: 10.1097/cad.0000000000000976] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Bone metastasis is one of the common phenomena in the late stage of lung cancer. Inhibition of bone metastasis can improve the survival of lung cancer patients. However, the current drugs for the treatment of bone metastasis have shown little effect on overall survival. Therefore, there is an urgent necessity to identify novel drugs capable of preventing and treating bone metastasis of lung cancer. Our study determined that icaritin (ICT) can inhibit lung cancer-mediated osteoclastogenesis and induce the apoptosis of osteoclasts. Exposure to ICT increased the activation of adenosine 5'-monophosphate-activated protein kinase (AMPK), reduced the activation of mammalian target of rapamycin (mTOR) and decreased the expression of bcl-2. The bioactivity of ICT on osteoclastogenesis was associated with the regulation of the AMPK/mTOR signaling pathway. Blocking AMPK significantly increased osteoclast differentiation, decreased osteoclast apoptosis and canceled the effects of ICT on the phosphorylation of AMPK as well as the inhibition of mTOR and bcl-2. Furthermore, ICT decreased the levels of IL-6 and TNF-α in osteoclasts, while the AMPK inhibitor compound C significantly abolished the inhibitory effects of ICT on IL-6 and TNF-α. Thus, the present study demonstrated that ICT may be a potential natural agent for the treatment of bone metastasis in patients with lung cancer.
Collapse
|
9
|
Li H, Li Y, Ao H, Fu J, Guo Y, Han M, Yan X, Chen X, Wang X. A comparative study on the in vitro and in vivo antitumor efficacy of icaritin and hydrous icaritin nanorods. Drug Deliv 2021; 27:1176-1187. [PMID: 32762483 PMCID: PMC7470086 DOI: 10.1080/10717544.2020.1801892] [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] [Indexed: 02/04/2023] Open
Abstract
Icaritin (ICT) and hydrous icaritin (HICT) are two similar flavonoids compounds isolated from Epimedium Genus. This is the first comparative study on their in vitro and in vivo antitumor effects. Nanorods (NRs) were prepared for ICT and HICT by anti-solvent precipitation method using D-alpha tocopherol acid polyethylene glycol succinate (TPGS) as a stabilizer. The prepared ICT-NRs and HICT-NRs had similar diameter (155.5 nm and 201.7 nm), high drug loading content (43.30 ± 0.22% and 41.08 ± 0.19%), excellent stability and a similar sustaining drug release manner. Nanorods improved the in vitro toxicity against 4 different cancer cells in contrast to free ICT or free HICT; however, no significant difference was observed in this regard between ICT-NRs and HICT NRs. In the in vivo study on the anticancer efficacy on MCF-7 and PLC/PRE/5 tumor-bearing mice model, HICR-NRs displayed certain advantage over ICT NRs with higher tumor inhibition rate.
Collapse
Affiliation(s)
- Haowen Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Yijing Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Hui Ao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Jingxin Fu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Yifei Guo
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Meihua Han
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Xueying Yan
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Xi Chen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Xiangtao Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| |
Collapse
|
10
|
Shan HJ, Zhu LQ, Yao C, Zhang ZQ, Liu YY, Jiang Q, Zhou XZ, Wang XD, Cao C. MAFG-driven osteosarcoma cell progression is inhibited by a novel miRNA miR-4660. MOLECULAR THERAPY-NUCLEIC ACIDS 2021; 24:385-402. [PMID: 33868783 PMCID: PMC8039776 DOI: 10.1016/j.omtn.2021.03.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 03/10/2021] [Indexed: 12/23/2022]
Abstract
Osteosarcoma (OS) is the most common primary bone malignancy in the adolescent population. MAFG (v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog G) forms a heterodimer with Nrf2 (NF-E2-related factor 2), binding to antioxidant response element (ARE), which is required for Nrf2 signaling activation. We found that MAFG mRNA and protein expression is significantly elevated in human OS tissues as well as in established and primary human OS cells. In human OS cells, MAGF silencing or knockout (KO) largely inhibited OS cell growth, proliferation, and migration, simultaneously inducing oxidative injury and apoptosis activation. Conversely, ectopic overexpression of MAFG augmented OS cell progression in vitro. MicroRNA-4660 (miR-4660) directly binds the 3′ untranslated region (UTR) of MAFG mRNA in the cytoplasm of OS cells. MAFG 3′ UTR luciferase activity and expression as well as OS cell growth were largely inhibited with forced miR-4660 overexpression but augmented with miR-4660 inhibition. In vivo, MAGF short hairpin RNA (shRNA) or forced overexpression of miR-4660 inhibited subcutaneous OS xenograft growth in severe combined immunodeficient mice. Furthermore, MAFG silencing or miR-4660 overexpression inhibited OS xenograft in situ growth in proximal tibia of the nude mice. In summary, MAFG overexpression-driven OS cell progression is inhibited by miR-4660. The miR-4660-MAFG axis could be novel therapeutic target for human OS.
Collapse
Affiliation(s)
- Hua-Jian Shan
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215003, China
| | - Lun-Qing Zhu
- Department of Pediatric Orthopedics, The Children's Hospital of Soochow University, Suzhou 215100, China
| | - Chen Yao
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of TCM, Nanjing, China
| | - Zhi-Qing Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Yuan-Yuan Liu
- Department of Radiotherapy and Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Qin Jiang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing 210029, China
| | - Xiao-Zhong Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215003, China
| | - Xiao-Dong Wang
- Department of Pediatric Orthopedics, The Children's Hospital of Soochow University, Suzhou 215100, China
| | - Cong Cao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China.,The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| |
Collapse
|
11
|
Carresi C, Scicchitano M, Scarano F, Macrì R, Bosco F, Nucera S, Ruga S, Zito MC, Mollace R, Guarnieri L, Coppoletta AR, Gliozzi M, Musolino V, Maiuolo J, Palma E, Mollace V. The Potential Properties of Natural Compounds in Cardiac Stem Cell Activation: Their Role in Myocardial Regeneration. Nutrients 2021; 13:275. [PMID: 33477916 PMCID: PMC7833367 DOI: 10.3390/nu13010275] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs), which include congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, and many other cardiac disorders, cause about 30% of deaths globally; representing one of the main health problems worldwide. Among CVDs, ischemic heart diseases (IHDs) are one of the major causes of morbidity and mortality in the world. The onset of IHDs is essentially due to an unbalance between the metabolic demands of the myocardium and its supply of oxygen and nutrients, coupled with a low regenerative capacity of the heart, which leads to great cardiomyocyte (CM) loss; promoting heart failure (HF) and myocardial infarction (MI). To date, the first strategy recommended to avoid IHDs is prevention in order to reduce the underlying risk factors. In the management of IHDs, traditional therapeutic options are widely used to improve symptoms, attenuate adverse cardiac remodeling, and reduce early mortality rate. However, there are no available treatments that aim to improve cardiac performance by replacing the irreversible damaged cardiomyocytes (CMs). Currently, heart transplantation is the only treatment being carried out for irreversibly damaged CMs. Hence, the discovery of new therapeutic options seems to be necessary. Interestingly, recent experimental evidence suggests that regenerative stem cell medicine could be a useful therapeutic approach to counteract cardiac damage and promote tissue regeneration. To this end, researchers are tasked with answering one main question: how can myocardial regeneration be stimulated? In this regard, natural compounds from plant extracts seem to play a particularly promising role. The present review will summarize the recent advances in our knowledge of stem cell therapy in the management of CVDs; focusing on the main properties and potential mechanisms of natural compounds in stimulating and activating stem cells for myocardial regeneration.
Collapse
Affiliation(s)
- Cristina Carresi
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Miriam Scicchitano
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Federica Scarano
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Roberta Macrì
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Francesca Bosco
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Saverio Nucera
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Stefano Ruga
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Maria Caterina Zito
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Rocco Mollace
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Lorenza Guarnieri
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Anna Rita Coppoletta
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Micaela Gliozzi
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Vincenzo Musolino
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Jessica Maiuolo
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Ernesto Palma
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88100 Catanzaro, Italy
| | - Vincenzo Mollace
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88100 Catanzaro, Italy
| |
Collapse
|
12
|
Roles of Reactive Oxygen Species in Cardiac Differentiation, Reprogramming, and Regenerative Therapies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2102841. [PMID: 32908625 PMCID: PMC7475763 DOI: 10.1155/2020/2102841] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022]
Abstract
Reactive oxygen species (ROS) have been implicated in mechanisms of heart development and regenerative therapies such as the use of pluripotent stem cells. The roles of ROS mediating cell fate are dependent on the intensity of stimuli, cellular context, and metabolic status. ROS mainly act through several targets (such as kinases and transcription factors) and have diverse roles in different stages of cardiac differentiation, proliferation, and maturation. Therefore, further detailed investigation and characterization of redox signaling will help the understanding of the molecular mechanisms of ROS during different cellular processes and enable the design of targeted strategies to foster cardiac regeneration and functional recovery. In this review, we focus on the roles of ROS in cardiac differentiation as well as transdifferentiation (direct reprogramming). The potential mechanisms are discussed in regard to ROS generation pathways and regulation of downstream targets. Further methodological optimization is required for translational research in order to robustly enhance the generation efficiency of cardiac myocytes through metabolic modulations. Additionally, we highlight the deleterious effect of the host's ROS on graft (donor) cells in a paracrine manner during stem cell-based implantation. This knowledge is important for the development of antioxidant strategies to enhance cell survival and engraftment of tissue engineering-based technologies. Thus, proper timing and level of ROS generation after a myocardial injury need to be tailored to ensure the maximal efficacy of regenerative therapies and avoid undesired damage.
Collapse
|
13
|
Bozorgi A, Khazaei S, Khademi A, Khazaei M. Natural and herbal compounds targeting breast cancer, a review based on cancer stem cells. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:970-983. [PMID: 32952942 PMCID: PMC7478260 DOI: 10.22038/ijbms.2020.43745.10270] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 03/09/2020] [Indexed: 12/13/2022]
Abstract
Cancer stem cells (CSCs) are known as the major reason for therapy resistance. Recently, natural herbal compounds are suggested to have a significant role in inhibiting the breast cancer stem cells (BCSCs). The aim of this study was to explore the effective natural herbal compounds against BCSCs.This review article was designed based on the BCSCs, mechanisms of therapy resistance and natural herbal compounds effective to inhibit their activity. Therefore, Science direct, PubMed and Scopus databases were explored and related original articles were investigated from 2010 to 2019. BCSCs use different mechanisms including special membrane transporters, anti-apoptotic, pro-survival, and self-renewal- related signaling pathways. Natural herbal compounds could disturb these mechanisms, therefore may inhibit or eradicate the BCSCs. Studies show that a broad range of plants, either as a food or medicine, contain anti-cancer agents that phenolic components and their different derivatives share a large quantity. Natural herbal compounds play a pivotal role in the eradication of BCSCs, through the inhibition of biological activities and induction of apoptosis. Although it is necessary to conduct more clinical investigation.
Collapse
Affiliation(s)
- Azam Bozorgi
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Saber Khazaei
- Dental Research Center, Dental Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abbasali Khademi
- Dental Research Center, Dental Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mozafar Khazaei
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
14
|
Godoy-Parejo C, Deng C, Zhang Y, Liu W, Chen G. Roles of vitamins in stem cells. Cell Mol Life Sci 2020; 77:1771-1791. [PMID: 31676963 PMCID: PMC11104807 DOI: 10.1007/s00018-019-03352-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/12/2019] [Accepted: 10/21/2019] [Indexed: 12/13/2022]
Abstract
Stem cells can differentiate to diverse cell types in our body, and they hold great promises in both basic research and clinical therapies. For specific stem cell types, distinctive nutritional and signaling components are required to maintain the proliferation capacity and differentiation potential in cell culture. Various vitamins play essential roles in stem cell culture to modulate cell survival, proliferation and differentiation. Besides their common nutritional functions, specific vitamins are recently shown to modulate signal transduction and epigenetics. In this article, we will first review classical vitamin functions in both somatic and stem cell cultures. We will then focus on how stem cells could be modulated by vitamins beyond their nutritional roles. We believe that a better understanding of vitamin functions will significantly benefit stem cell research, and help realize their potentials in regenerative medicine.
Collapse
Affiliation(s)
- Carlos Godoy-Parejo
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Chunhao Deng
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Yumeng Zhang
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Weiwei Liu
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
- Bioimaging and Stem Cell Core Facility, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Guokai Chen
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China.
- Bioimaging and Stem Cell Core Facility, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China.
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China.
| |
Collapse
|
15
|
Hu J, Wu X, Yang C, Rashid K, Ma C, Hu M, Ding Q, Jiang H. Anticancer effect of icaritin on prostate cancer via regulating miR-381-3p and its target gene UBE2C. Cancer Med 2019; 8:7833-7845. [PMID: 31646760 PMCID: PMC6912031 DOI: 10.1002/cam4.2630] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 10/07/2019] [Accepted: 10/08/2019] [Indexed: 12/14/2022] Open
Abstract
Prostate cancer (PCa) is one of the most common health-related issues in the male individuals of western countries. Icaritin (ICT) is a traditional Chinese herbal medicine that exhibits antitumor efficacy in variety of cancers including PCa. However, the precise function and detailed molecular mechanism of ICT in the regression of PCa remain unclear. Ubiquitin-conjugating enzyme E2C (UBE2C) is an anaphase-promoting complex/cyclosome (APC/C)-specific ubiquitin conjugating enzyme, which acts as an oncogene in PCa progression. The function of ICT in PCa was investigated in transgenic adenocarcinoma mouse prostate (TRAMP) mice using survival analysis, hematoxylin and eosin (HE) staining, TUNEL assay, and immunohistochemistry and in human PCa cell lines using various molecular techniques and functional assays including plasmid construction and transfection. Bioinformatic analyses were performed to identify the interaction between miRNA and UBE2C via the TargetScan algorithm. We demonstrated that ICT inhibited the development and progression of PCa in TRAMP mice by improving the survival rate and tumor differentiation. Furthermore, we found that ICT could significantly inhibit cell proliferation and invasion and induce apoptosis in PCa cells. Consistently, downregulation of UBE2C suppressed the proliferation and invasion of PCa cells. Moreover, a luciferase reporter assay confirmed that UBE2C was a direct target of miR-381-3p. Meanwhile, ICT simultaneously downregulated UBE2C expression and upregulated miR-381-3p levels in human PCa cells. Altogether, our findings provide a strong rationale for the clinical application of ICT as a potential oncotherapeutic agent against PCa via a novel molecular mechanism of regulating the miR-381-3p/UBE2C pathway.
Collapse
Affiliation(s)
- Jimeng Hu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaobo Wu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Chen Yang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Khalid Rashid
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chenkai Ma
- Department of Surgery, Royal Melbourne Hospital, University of Melbourne, Melbourne, Vic., Australia
| | - Mengbo Hu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiang Ding
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Haowen Jiang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Chen X, Song L, Hou Y, Li F. Reactive oxygen species induced by icaritin promote DNA strand breaks and apoptosis in human cervical cancer cells. Oncol Rep 2018; 41:765-778. [PMID: 30431140 PMCID: PMC6312933 DOI: 10.3892/or.2018.6864] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 10/08/2018] [Indexed: 12/25/2022] Open
Abstract
Increased production of reactive oxygen species (ROS) is a distinct feature of various types of cancer. ROS drive tumor progression and render cancer cells vulnerable to additional oxidative insult. The various natural herb compounds have been shown to induce additional production of ROS in cancer cells, although the physiological implications of ROS under these conditions are not fully determined. In the present study, icaritin, a natural compound derived from the medicinal plants Epimedium, was demonstrated to potently suppresses the proliferation of human HeLa and SiHa cervical cancer cells, without similar affects on non-cancerous CCD-1095Sk fibroblasts and 293 cells, as measured by MTT and colony formation assays. Icaritin treatment caused a rapid increase in ROS in HeLa and SiHa cells, which was followed by a prominent increase in the number of DNA strand breaks. Consequently, the levels of the pro-apoptotic protein Bax and activated caspase 3 and 9 enzymes were increased, while the levels of the anti-apoptotic proteins Bcl-2 and XIAP were downregulated. These protein expression changes were accompanied by marked induction of apoptosis in icaritin-treated cancer cells. The results suggested that the icaritin-induced ROS overload promoted cancer cell death via induction of extensive oxidative DNA damage, which subsequently resulted in large numbers of DNA strand breaks and the activation of the intrinsic apoptotic pathway.
Collapse
Affiliation(s)
- Xin Chen
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Liyan Song
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Yuefang Hou
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Fan Li
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
17
|
Xiao J, Ke ZP, Shi Y, Zeng Q, Cao Z. The cardioprotective effect of thymoquinone on ischemia-reperfusion injury in isolated rat heart via regulation of apoptosis and autophagy. J Cell Biochem 2018; 119:7212-7217. [PMID: 29932232 DOI: 10.1002/jcb.26878] [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: 09/10/2017] [Accepted: 03/21/2018] [Indexed: 12/14/2022]
Abstract
Thymoquinone (TQ), as the active constituents of black cumin (Nigella sativa) seed oil, has been reported to have potential protective effects on the cardiovascular system. This study aimed to investigate the effects and the underlying mechanisms of TQ on myocardial ischemia-reperfusion (I/R) injury in Langendorff-perfused rat hearts. Wister rat hearts were subjected to I/R and the experimental group were pretreated with TQ prior to I/R. Hemodynamic parameters, myocardial infarct size, cardiac marker enzymes, superoxide dismutase (SOD), malondialdehyde (MDA) content, and cardiomyocyte apoptosis were assayed. Compared with the untreated group, TQ preconditioning significantly improved cardiac function, reduced infarct size, decreased cardiac lactate dehydrogenase (LDH) and creatine kinase-MB (CK-MB) levels, suppressed enedoxidative stress, and apoptosis. In addition, TQ treatment promoted autophagy, which was partially reversed by chloroquine (CQ), a kind of autophagy blocker. Our study suggests that TQ can protect heart against I/R injury, which is associated with anti-oxidative and anti-apoptotic effects through activation of autophagy.
Collapse
Affiliation(s)
- Junhui Xiao
- Department of Cardiology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China
| | - Zun-Ping Ke
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Yan Shi
- The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Qiutang Zeng
- Department of Cardiology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China
| | - Zhe Cao
- Department of Cardiology,The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
18
|
Hu J, Zhu W, Wei B, Wen H, Mao S, Xu H, Hu M, Yang T, Jiang H. Antitumoral action of icaritin in LNCaP prostate cancer cells by regulating PEA3/HER2/AR signaling. Anticancer Drugs 2017; 27:944-52. [PMID: 27537398 DOI: 10.1097/cad.0000000000000420] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human epidermal growth factor receptor type 2 (HER2) and androgen receptor (AR) are critical factors for prostate cancer (PCa) progression. These factors regulate tumor cell survival and proliferation, and remain as crucial drivers of castration-resistant PCa progression. Icaritin (ICT) is a prenyl flavonoid derived from the Epimedium genus, which has many biological and pharmacological effects. Using androgen-sensitive human prostate carcinoma LNCaP cell lines, we found that 35 μg/ml of ICT could inhibit more than 50% of cell proliferation, induce cell apoptosis, and lead to a strong G1 phase arrest by targeting cyclin-related proteins and suppressing the ability of cell invasion. Moreover, ICT exerts its potent anticancer efficacy by inducing polyomavirus enhancer activator 3 (PEA3) to inhibit the aberrantly activated HER2/AR signaling. In addition, after PEA3 expression was silenced by specific small-interference RNA, we found that both the ICT-inhibited effect on LNCaP cell proliferation and the ICT-induced cell apoptosis rate decreased. These results provide alternative mechanisms for the antitumor actions of ICT, indicating that ICT might be a promising therapeutic agent, as well as a preventive agent, for hormone therapy-resistant PCa.
Collapse
Affiliation(s)
- Jimeng Hu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Zheng ZG, Zhang X, Zhou YP, Lu C, Thu PM, Qian C, Zhang M, Li P, Li HJ, Xu X. Anhydroicaritin, a SREBPs inhibitor, inhibits RANKL-induced osteoclastic differentiation and improves diabetic osteoporosis in STZ-induced mice. Eur J Pharmacol 2017; 809:156-162. [DOI: 10.1016/j.ejphar.2017.05.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 05/04/2017] [Accepted: 05/10/2017] [Indexed: 12/22/2022]
|
20
|
Wu Z, Ou L, Wang C, Yang L, Wang P, Liu H, Xiong Y, Sun K, Zhang R, Zhu X. Icaritin induces MC3T3-E1 subclone14 cell differentiation through estrogen receptor-mediated ERK1/2 and p38 signaling activation. Biomed Pharmacother 2017; 94:1-9. [PMID: 28742995 DOI: 10.1016/j.biopha.2017.07.071] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 06/27/2017] [Accepted: 07/17/2017] [Indexed: 10/19/2022] Open
Abstract
Icaritin (ICT), a hydrolytic product of icariin from the genus Epimedium, has many indicated pharmacological and biological activities. Several studies have shown that ICT has potential osteoprotective effects, including stimulation of osteoblast differentiation and inhibition of osteoclast differentiation. However, the molecular mechanism for this anabolic action of ICT remains largely unknown. Here, we found that ICT could enhance MC3T3-E1 subclone 14 preosteoblastic cell differentiation associated with increased mRNA levels and protein expression of the differentiation markers alkaline phosphatase (ALP), type 1 collagen (COL1), osteocalcin (OC), osteoponin (OPN) and runt-related transcription factor 2 (RUNX2), and improved mineralization, confirmed by bone nodule formation and collagen synthesis. To characterize the underlying mechanisms, we examined the effect of ICT on estrogen receptor (ER) and mitogen-activated protein kinase (MAPK) signaling. ICT treatment induced p38 kinase and extracellular signal-regulated kinase 1/2 (ERK1/2) activation, but it demonstrated at the same time point no effect on activation of c-Jun N-terminal kinase (JNK). ER antagonist ICI182780, p38 antagonist SB203580 and ERK1/2 antagonist PD98059 markedly inhibited the ICT-induced the mRNA expression of ALP, COL1, OC and OPN. ICI182780 attenuated the ICT-induced phosphorylation of p38 and ERK1/2. These observations indicate a potential mechanism of osteogenic effects of ICT involving the ERK1/2 and p38 pathway activation through the ER.
Collapse
Affiliation(s)
- Zhidi Wu
- Department of Chinese Medicine, College of Pharmacy of Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Ling Ou
- Department of Chinese Medicine, College of Pharmacy of Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Chaopeng Wang
- Department of Chinese Medicine, College of Pharmacy of Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Li Yang
- Department of Chinese Medicine, College of Pharmacy of Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Panpan Wang
- Department of Chinese Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Hengrui Liu
- Department of Chinese Medicine, College of Pharmacy of Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Yingquan Xiong
- Department of Chinese Medicine, College of Pharmacy of Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Kehuan Sun
- Department of Chinese Medicine, College of Pharmacy of Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Ronghua Zhang
- Department of Chinese Medicine, College of Pharmacy of Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Xiaofeng Zhu
- Department of Chinese Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, PR China.
| |
Collapse
|
21
|
Tang LL, Wang JD, Xu TT, Zhao Z, Zheng JJ, Ge RS, Zhu DY. Mitochondrial toxicity of perfluorooctane sulfonate in mouse embryonic stem cell-derived cardiomyocytes. Toxicology 2017; 382:108-116. [PMID: 28288859 DOI: 10.1016/j.tox.2017.03.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 03/09/2017] [Accepted: 03/09/2017] [Indexed: 11/30/2022]
Abstract
Perfluorooctane sulfonate (PFOS) is a persistent organic contaminant that may cause cardiotoxicity in animals and humans. However, little is known about the underlying mechanism by which it affects the organelle toxicity in cardiomyocytes during the cardiogenesis. Our previous proteomic study showed that differences of protein expression mainly existed in mitochondria of cardiomyocytes differentiated from embryonic stem (ES) cells after exposure to PFOS. Here, we focused on mitochondrial toxicity of PFOS in ES cell-derived cardiomyocytes. The cardiomyogenesis from ES cells in vitro was inhibited, and the expression of L-type Ca2+ channel (LTCC) was decreased to interrupt [Ca2+]c transient amplitude in cardiomyocytes after PFOS treatment. Transmission electron microscope revealed that swollen mitochondrion with vacuole in PFOS-treated cells. Meanwhile, mitochondrial transmembrane potential (ΔΨm) was declined and ATP production was lowered. These changes were related to the increased EGFR phosphorylation, activated Rictor signaling, then mediated HK2 binding to mitochondrial membrane. Furthermore, PFOS reduced the interaction of IP3R-Grp75-VDAC and accumulated intracellular fatty acids by activating Rictor, thereby attenuating PGC-1α and Mfn2 expressions, then destroying mitochondria-associated endoplasmic reticulum membrane (MAM), which resulted in the decrease of [Ca2+]mito transient amplitude triggered by ATP. In conclusion, mitochondrial structure damages and abnormal Ca2+ shuttle were the important aspects in PFOS-induced cardiomyocytes toxicity from ES cells by activating Rictor signaling pathway.
Collapse
Affiliation(s)
- Lei-Lei Tang
- Institute of Pharmacology and Toxicology, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China; Department of Pharmacy, Xiaoshan Hospital, Hangzhou 311200, China
| | - Jia-Dan Wang
- Institute of Pharmacology and Toxicology, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Ting-Ting Xu
- Institute of Pharmacology and Toxicology, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Zhe Zhao
- Undergraduate Students in Research Training Project at Zhejiang University, Hangzhou 310058, China
| | - Jia-Jie Zheng
- Undergraduate Students in Research Training Project at Zhejiang University, Hangzhou 310058, China
| | - Ren-Shan Ge
- The Population Council at the Rockefeller University, New York, NY 10021, USA; Institute of Reproductive Biomedicine, The 2nd Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Dan-Yan Zhu
- Institute of Pharmacology and Toxicology, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.
| |
Collapse
|
22
|
Abstract
Although As2O3 (ATO) has been recommended as the front-line agent for treatment of acute promyelocytic leukemia (APL), particularly for relapsed or refractory APL, it has been associated with profound toxicity. Icariin is a natural compound with activity against a variety of cancers. This study was designed to investigate the effect of Icariin on APL cells and to determine whether Icariin can potentiate the antitumor activity of ATO in APL cells. Cell proliferation and apoptosis were measured using MTT assay and flow cytometry, respectively. The expression of apoptosis and proliferation-related molecules was detected by Western blotting. Reactive oxygen species (ROS) and mitochondrial membrane potential were determined with florescence staining. Icariin inhibited proliferation in a dose-dependent manner and induced apoptosis in both of the tested APL cell lines. Icariin enhanced the in vitro antitumor activity of ATO against APL. The antitumor activity of Icariin and its enhancement of the antitumor activity of ATO correlated with the increase in accumulation of intracellular ROS. Our results showed that Icariin, by increasing intracellular ROS, exhibited antitumor activity and potentiated the antitumor activity of ATO against APL. Therefore, combination treatment with Icariin and ATO might offer a novel therapeutic option for patients with APL, although further studies are needed.
Collapse
|
23
|
Icaritin enhances mESC self-renewal through upregulating core pluripotency transcription factors mediated by ERα. Sci Rep 2017; 7:40894. [PMID: 28091581 PMCID: PMC5238509 DOI: 10.1038/srep40894] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 12/12/2016] [Indexed: 12/28/2022] Open
Abstract
Utilization of small molecules in modulation of stem cell self-renewal is a promising approach to expand stem cells for regenerative therapy. Here, we identify Icaritin, a phytoestrogen molecule enhances self-renewal of mouse embryonic stem cells (mESCs). Icaritin increases mESCs proliferation while maintains their self-renewal capacity in vitro and pluripotency in vivo. This coincides with upregulation of key pluripotency transcription factors OCT4, NANOG, KLF4 and SOX2. The enhancement of mESCs self-renewal is characterized by increased population in S-phase of cell cycle, elevation of Cylin E and Cyclin-dependent kinase 2 (CDK2) and downregulation of p21, p27 and p57. PCR array screening reveals that caudal-related homeobox 2 (Cdx2) and Rbl2/p130 are remarkably suppressed in mESCs treated with Icaritin. siRNA knockdown of Cdx2 or Rbl2/p130 upregulates the expression of Cyclin E, OCT4 and SOX2, and subsequently increases cell proliferation and colony forming efficiency of mESCs. We then demonstrate that Icaritin co-localizes with estrogen receptor alpha (ERα) and activates its nuclear translocation in mESCs. The promotive effect of Icaritin on cell cycle and pluripotency regulators are eliminated by siRNA knockdown of ERα in mESCs. The results suggest that Icaritin enhances mESCs self-renewal by regulating cell cycle machinery and core pluripotency transcription factors mediated by ERα.
Collapse
|
24
|
Zheng B, Wang J, Tang L, Tan C, Zhao Z, Xiao Y, Ge R, Zhu D. Involvement of Rictor/mTORC2 in cardiomyocyte differentiation of mouse embryonic stem cells in vitro. Int J Biol Sci 2017; 13:110-121. [PMID: 28123351 PMCID: PMC5264266 DOI: 10.7150/ijbs.16312] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 10/21/2016] [Indexed: 11/05/2022] Open
Abstract
Rictor is a key regulatory/structural subunit of the mammalian target of rapamycin complex 2 (mTORC2) and is required for phosphorylation of Akt at serine 473. It plays an important role in cell survival, actin cytoskeleton organization and other processes in embryogenesis. However, the role of Rictor/mTORC2 in the embryonic cardiac differentiation has been uncovered. In the present study, we examined a possible link between Rictor expression and cardiomyocyte differentiation of the mouse embryonic stem (mES) cells. Knockdown of Rictor by shRNA significantly reduced the phosphorylation of Akt at serine 473 followed by a decrease in cardiomyocyte differentiation detected by beating embryoid bodies. The protein levels of brachyury (mesoderm protein), Nkx2.5 (cardiac progenitor cell protein) and α-Actinin (cardiomyocyte biomarker) decreased in Rictor knockdown group during cardiogenesis. Furthermore, knockdown of Rictor specifically inhibited the ventricular-like cells differentiation of mES cells with reduced level of ventricular-specific protein, MLC-2v. Meanwhile, patch-clamp analysis revealed that shRNA-Rictor significantly increased the number of cardiomyocytes with abnormal electrophysiology. In addition, the expressions and distribution patterns of cell-cell junction proteins (Cx43/Desmoplakin/N-cadherin) were also affected in shRNA-Rictor cardiomyocytes. Taken together, the results demonstrated that Rictor/mTORC2 might play an important role in the cardiomyocyte differentiation of mES cells. Knockdown of Rictor resulted in inhibiting ventricular-like myocytes differentiation and induced arrhythmias symptom, which was accompanied by interfering the expression and distribution patterns of cell-cell junction proteins. Rictor/mTORC2 might become a new target for regulating cardiomyocyte differentiation and a useful reference for application of the induced pluripotent stem cells.
Collapse
Affiliation(s)
- Bei Zheng
- Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou 310058, CHINA
| | - Jiadan Wang
- Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou 310058, CHINA
| | - Leilei Tang
- Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou 310058, CHINA
| | - Chao Tan
- Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou 310058, CHINA
| | - Zhe Zhao
- Undergraduate students in Research Training Project at Zhejiang University
| | - Yi Xiao
- Undergraduate students in Research Training Project at Zhejiang University
| | - Renshan Ge
- The Population Council at the Rockefeller University, New York, NY 10021, USA.; Institute of Reproductive Biomedicine, the 2nd Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, CHINA
| | - Danyan Zhu
- Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou 310058, CHINA
| |
Collapse
|
25
|
Icaritin Reduces Oral Squamous Cell Carcinoma Progression via the Inhibition of STAT3 Signaling. Int J Mol Sci 2017; 18:ijms18010132. [PMID: 28085115 PMCID: PMC5297765 DOI: 10.3390/ijms18010132] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 12/20/2016] [Accepted: 01/06/2017] [Indexed: 12/15/2022] Open
Abstract
Icaritin, a traditional Chinese medicine, possesses antitumor activity. The current study aimed to investigate icaritin effect and potential mechanism on oral squamous cell carcinoma (OSCC) development. OSCC cells proliferation, apoptosis, and autophagy were analyzed after incubation with icaritin at different concentrations and incubation times. The expressions of proteins related to proliferation, apoptosis, and autophagy, as well as signal transducer and activator of transcription 3 (STAT3) signal network, were also evaluated by western blot. Furthermore, STAT3 was knocked down by siRNA transfection to determine STAT3 role in OSCC cell proliferation and apoptosis. An oral specific carcinogenesis mouse model was used to explore icaritin effect on OSCC in vivo. Icaritin significantly inhibited OSCC proliferation in vitro and reduced the expression of both the cell-cycle progression proteins cyclin A2 and cyclin D1. Besides, icaritin increased cleaved caspase 3 and cleaved poly-(ADP-ribose) polymerase expression leading to apoptosis, and it activated autophagy. Icaritin significantly inhibited the expression of phospho-STAT3 (p-STAT3) in a dose- and time-dependent manner. In the in vivo experiment, the number of malignant tumors in the icaritin-treated group was significantly lower than the control. Overall, icaritin suppressed proliferation, promoted apoptosis and autophagy, and inhibited STAT3 signaling in OSCC in vitro and in vivo. In conclusion, icaritin might be a potential therapeutic agent against OSCC development.
Collapse
|
26
|
Zheng ZG, Zhou YP, Zhang X, Thu PM, Xie ZS, Lu C, Pang T, Xue B, Xu DQ, Chen Y, Chen XW, Li HJ, Xu X. Anhydroicaritin improves diet-induced obesity and hyperlipidemia and alleviates insulin resistance by suppressing SREBPs activation. Biochem Pharmacol 2016; 122:42-61. [PMID: 27816546 DOI: 10.1016/j.bcp.2016.10.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 10/31/2016] [Indexed: 11/24/2022]
Abstract
SREBPs play important roles in the regulation of lipid metabolism, and are closely related to the occurrence and development of many metabolic diseases. Small molecular inhibitors of SERBPs are important tools in developing efficient treatment of metabolic diseases. However, there are no listing drug targeting SREBPs. Therefore, there is an urgent need to develop highly specific small molecules that inhibit SREBPs. In this study, using a hepatocyte-based high-throughput screening, we identified anhydroicaritin (AHI) as a novel inhibitor of SREBPs. HepG2, HL-7702, and human primary hepatocytes were used to verify the effects of AHI. We explored the mechanism by which AHI blocks the binding of SCAP/SREBPs complex with Sec23α/24D via regulating LKB1/AMPK/mTOR pathway. AHI reduced liver cell lipid level by preventing de novo lipogenesis. In diet induced obese mice, AHI ameliorated obesity, insulin resistance, fatty accumulation in liver and hyperlipemia. In conclusion, AHI improves diet-induced obesity and alleviates insulin resistance by suppressing SREBPs maturation which is dependent on LKB1/AMPK/mTOR pathway. Thus, AHI can serve as a leading compound for pharmacological control of metabolic diseases.
Collapse
Affiliation(s)
- Zu-Guo Zheng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009 Nanjing, Jiangsu, China
| | - Ya-Ping Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009 Nanjing, Jiangsu, China
| | - Xin Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009 Nanjing, Jiangsu, China
| | - Pyone Myat Thu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009 Nanjing, Jiangsu, China
| | - Zhi-Shen Xie
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009 Nanjing, Jiangsu, China
| | - Chong Lu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009 Nanjing, Jiangsu, China
| | - Tao Pang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009 Nanjing, Jiangsu, China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, 210009 Nanjing, Jiangsu, China
| | - Bin Xue
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing University, 210093 Nanjing, Jiangsu, China
| | - Da-Qian Xu
- Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences, 200031 Shanghai, China
| | - Yan Chen
- Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences, 200031 Shanghai, China
| | - Xiao-Wei Chen
- Institute of Molecular Medicine, Peking University, 100871 Beijing, China
| | - Hui-Jun Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009 Nanjing, Jiangsu, China.
| | - Xiaojun Xu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009 Nanjing, Jiangsu, China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, 210009 Nanjing, Jiangsu, China.
| |
Collapse
|
27
|
A novel anti-cancer agent Icaritin suppresses hepatocellular carcinoma initiation and malignant growth through the IL-6/Jak2/Stat3 pathway. Oncotarget 2016; 6:31927-43. [PMID: 26376676 PMCID: PMC4741651 DOI: 10.18632/oncotarget.5578] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Accepted: 08/20/2015] [Indexed: 01/05/2023] Open
Abstract
Tumor-initiating cell (TIC) is a subpopulation of cells in tumors that are responsible for tumor initiation and progression. Recent studies indicate that hepatocellular carcinoma-initiating cells (HCICs) confer the high malignancy, recurrence and multi-drug resistance in hepatocellular carcinoma (HCC). In this study, we found that Icaritin, a prenylflavonoid derivative from Epimedium Genus, inhibited malignant growth of HCICs. Icaritin decreased the proportion of EpCAM-positive (a HCICs marker) cells, suppressed tumorsphere formation in vitro and tumor formation in vivo. We also found that Icaritin reduced expression of Interleukin-6 Receptors (IL-6Rs), attenuated both constitutive and IL-6-induced phosphorylation of Janus-activated kinases 2 (Jak2) and Signal transducer and activator of transcription 3 (Stat3), and inhibited Stat3 downstream genes, such as Bmi-1 and Oct4. The inhibitory activity of Icaritin in HCICs was augmented by siRNA-mediated silencing of Stat3 but attenuated by constitutive activation of Stat3. Taken together, our results indicate that Icaritin is able to inhibit malignant growth of HCICs and suggest that Icaritin may be developed into a novel therapeutic agent for effective treatment of HCC.
Collapse
|
28
|
A novel anticancer agent icaritin inhibited proinflammatory cytokines in TRAMP mice. Int Urol Nephrol 2016; 48:1649-55. [PMID: 27282153 DOI: 10.1007/s11255-016-1341-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 06/03/2016] [Indexed: 01/07/2023]
Abstract
PURPOSE We aimed to investigate whether icaritin (ICT) would inhibit serum proinflammatory cytokines and postpone prostate cancer (PCa) development and progression in both normal diet and high-fat diet (HFD) transgenic adenocarcinoma mouse prostate (TRAMP) mice. METHODS TRAMP mice were randomly divided into four groups: normal diet with/without ICT group and HFD with/without ICT group. Each TRAMP mouse received intraperitoneal injection of ICT solution at the dose of 30 mg/kg 5 times per week. RESULTS ICT treatment could significantly increase the survival when compared with those in normal diet group (P = 0.015, log-rank test) and HFD group (P = 0.009, log-rank test). Proinflammatory cytokine levels, including IL-1α, IL-1β, IL-6, and TNF-α, were decreased more or less in ICT-treated TRAMP mice. Moreover, significant higher inflammation scores were detected in normal diet group and HFD group compared with their relevant ICT treatment groups (P = 0.026 and P = 0.006, respectively). Meanwhile, the incidences of well-differentiated tumor tissue in two ICT treatment groups (39.13 and 31.82 %) were moderately higher than control groups (29.41 and 20.00 %, respectively), though no significant difference was observed. CONCLUSIONS Taken together, our findings indicate that ICT could inhibit the development and progression of PCa in TRAMP mice via inhibiting proinflammatory cytokines.
Collapse
|
29
|
Zhang YY, Tang LL, Zheng B, Ge RS, Zhu DY. Protein profiles of cardiomyocyte differentiation in murine embryonic stem cells exposed to perfluorooctane sulfonate. J Appl Toxicol 2016; 36:726-40. [PMID: 26178269 DOI: 10.1002/jat.3207] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 06/03/2015] [Accepted: 06/03/2015] [Indexed: 12/30/2022]
Abstract
Perfluorooctane sulfonate (PFOS) is a persistent organic contaminant that may affect diverse systems in animals and humans, including the cardiovascular system. However, little is known about the mechanism by which it affects the biological systems. Herein, we used embryonic stem cell test procedure as a tool to assess the developmental cardiotoxicity of PFOS. The differentially expressed proteins were identified by quantitative proteomics that combines the stable isotope labeling of amino acids with high-performance liquid chromatography-electrospray ionization tandem mass spectrometry. Results of the embryonic stem cell test procedure suggested that PFOS was a weak embryotoxic chemical. Nevertheless, a few marker proteins related to cardiovascular development (Brachyury, GATA4, MEF2C, α-actinin) were significantly reduced by exposure to PFOS. In total, 176 differential proteins were identified by proteomics analysis, of which 67 were upregulated and 109 were downregulated. Gene ontology annotation classified these proteins into 13 groups by molecular functions, 12 groups by cellular locations and 10 groups by biological processes. Most proteins were mainly relevant to either catalytic activity (25.6%), nucleus localization (28.9%) or to cellular component organization (19.8%). Pathway analysis revealed that 32 signaling pathways were affected, particularly these involved in metabolism. Changes in five proteins, including L-threonine dehydrogenase, X-ray repair cross-complementing 5, superoxide dismutase 2, and DNA methyltransferase 3b and 3a were confirmed by Western blotting, suggesting the reliability of the technique. These results revealed potential new targets of PFOS on the developmental cardiovascular system.
Collapse
Affiliation(s)
- Ying-Ying Zhang
- Institute of Pharmacology, Toxicology, and Biochemical Pharmaceutics, Zhejiang University, Hangzhou, China
| | - Lei-Lei Tang
- Institute of Pharmacology, Toxicology, and Biochemical Pharmaceutics, Zhejiang University, Hangzhou, China
| | - Bei Zheng
- Institute of Pharmacology, Toxicology, and Biochemical Pharmaceutics, Zhejiang University, Hangzhou, China
| | - Ren-Shan Ge
- Institute of Reproductive Biomedicine and the 2nd Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dan-Yan Zhu
- Institute of Pharmacology, Toxicology, and Biochemical Pharmaceutics, Zhejiang University, Hangzhou, China
| |
Collapse
|
30
|
Icaritin suppresses multiple myeloma, by inhibiting IL-6/JAK2/STAT3. Oncotarget 2016; 6:10460-72. [PMID: 25865044 PMCID: PMC4496367 DOI: 10.18632/oncotarget.3399] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 02/15/2015] [Indexed: 01/05/2023] Open
Abstract
Icaritin is an active prenylflavonoid derived from Epimedium genus, a traditional Chinese medicine. Icaritin has a wide range of pharmacological and biological activities, including cardiovascular function improvement, hormone regulation and antitumor activity. Here, we investigated the effect of icaritin on multiple myeloma (MM) in vitro and in vivo. Icaritin inhibited cell growth of MM cell line and primary MM cells. In contrast, icaritin had low or no cytotoxic effect on normal hematopoiesis. We also demonstrated that in MM xenograft mouse models, icaritin suppressed tumor growth and decreased serum IL-6 and IgE levels, but did not show adverse reactions such as body weight loss. The anti-MM activity of icaritin was mainly mediated by inhibiting IL-6/JAK2/STAT3 signaling. We suggest that icaritin can be further tested in clinical trials in MM.
Collapse
|
31
|
Ke Z, Liu J, Xu P, Gao A, Wang L, Ji L. The Cardioprotective Effect of Icariin on Ischemia-Reperfusion Injury in Isolated Rat Heart: Potential Involvement of the PI3K-Akt Signaling Pathway. Cardiovasc Ther 2016; 33:134-40. [PMID: 25847837 DOI: 10.1111/1755-5922.12121] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
AIMS Icariin (ICA), a flavonoid isolated from epimedii, has been reported to have potential protective effects on the cardiovascular system. This study is to investigate the effect and the underlying mechanisms of ICA on ischemia/reperfusion (I/R) injury. METHODS Wister rat hearts were subjected to I/R using Langendorff perfusion system. Cardiac function, myocardial infarct size, lactate dehydrogenase (LDH), and creatine kinase-MB (CK-MB) activities in coronary effluent, and superoxide dismutase (SOD) and malondialdehyde (MDA) content in heart tissue and cardiomyocyte apoptosis were assayed. RESULTS Compared with the I/R group, ICA treatment significantly improved cardiac function, decreased myocardial infarct size, enzyme activity, oxidative stress, and apoptosis. In addition, ICA treatment lead to an increased p-Akt level, which was partially reversed by LY294002, a PI3K pathway inhibitor. CONCLUSION Our study suggests that ICA has a cardioprotective effect against I/R injury, which is associated with its antioxidative and anti-apoptotic effect, at least partially, through the activation of PI3K-Akt signaling pathway.
Collapse
Affiliation(s)
- Zunping Ke
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Jingjing Liu
- Department of Nephrology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Peng Xu
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Aimei Gao
- Department of Pharmacy, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Lei Wang
- Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Lijuan Ji
- Department of Rehabilitation, The Second People's Hospital of Huai'an, Huai'an, Jiangsu, China
| |
Collapse
|
32
|
Ye LH, Xiao BX, Cao FR, Zheng Y, Pan RL, Chang Q. Identification of Icaritin Metabolites in Rats by LC-MS/MS. CHINESE HERBAL MEDICINES 2015. [DOI: 10.1016/s1674-6384(15)60055-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
33
|
Pan XW, Li L, Huang Y, Huang H, Xu DF, Gao Y, Chen L, Ren JZ, Cao JW, Hong Y, Cui XG. Icaritin acts synergistically with epirubicin to suppress bladder cancer growth through inhibition of autophagy. Oncol Rep 2015; 35:334-42. [PMID: 26496799 DOI: 10.3892/or.2015.4335] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 08/19/2015] [Indexed: 11/06/2022] Open
Abstract
Bladder cancer is one of the most commonly diagnosed urological malignancies. Acquired resistance to chemotherapy is a great barrier for achieving successful treatment of bladder cancer. In the present study, we investigated the effect and mechanisms of icaritin, a flavonol glycoside derived from genus Epimedium, against human bladder cancer cells. It was found that despite the low cytotoxicity in normal human HEK293 cells, icaritin significantly inhibited the proliferation and colony formation of BT5637 and T24 bladder cancer cells time- and dose-dependently compared to the DMSO vehicle control. Moreover, cell viability monitored through mitochondrial membrane potential was inhibited markedly after icaritin treatment. Further investigation indicated that icaritin may inhibit epirubicin (EPI)-induced autophagy, and acted synergistically with EPI to suppress the proliferation of BT5637 and T24 cells. These findings suggest that icaritin may prove to be a novel potent therapeutic agent for the treatment of bladder cancer.
Collapse
Affiliation(s)
- Xiu-Wu Pan
- Department of Urinary Surgery of Changzheng Hospital, Second Military Medical University, Huangpu, Sanghai 200003, P.R. China
| | - Lin Li
- Department of Urinary Surgery of Changzheng Hospital, Second Military Medical University, Huangpu, Sanghai 200003, P.R. China
| | - Yi Huang
- Department of Urinary Surgery of Changzheng Hospital, Second Military Medical University, Huangpu, Sanghai 200003, P.R. China
| | - Hai Huang
- Department of Urinary Surgery of Changzheng Hospital, Second Military Medical University, Huangpu, Sanghai 200003, P.R. China
| | - Dan-Feng Xu
- The Urology Research Center of the Chinese People's Liberation Army, Changzheng Hospital, Second Military Medical University, Huangpu, Shanghai 200003, P.R. China
| | - Yi Gao
- Department of Urinary Surgery of Changzheng Hospital, Second Military Medical University, Huangpu, Sanghai 200003, P.R. China
| | - Lu Chen
- Department of Urinary Surgery of Changzheng Hospital, Second Military Medical University, Huangpu, Sanghai 200003, P.R. China
| | - Ji-Zhong Ren
- Department of Urinary Surgery of Changzheng Hospital, Second Military Medical University, Huangpu, Sanghai 200003, P.R. China
| | - Jian-Wei Cao
- Navy No. 411 Hospital of PLA, Hongkou, Shanghai 200003, P.R. China
| | - Yi Hong
- Department of Urinary Surgery of Changzheng Hospital, Second Military Medical University, Huangpu, Sanghai 200003, P.R. China
| | - Xin-Gang Cui
- Department of Urinary Surgery of Changzheng Hospital, Second Military Medical University, Huangpu, Sanghai 200003, P.R. China
| |
Collapse
|
34
|
Zhang W, Xing B, Yang L, Shi J, Zhou X. Icaritin Attenuates Myocardial Ischemia and Reperfusion Injury Via Anti-Inflammatory and Anti-Oxidative Stress Effects in Rats. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2015; 43:1083-97. [PMID: 26364662 DOI: 10.1142/s0192415x15500627] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Icaritin (ICT) is a traditional Chinese medicinal herb proved to be neuroprotective and exerts promoting effects on cardiac differentiation. However, its role in cardioprotection against myocardial ischemia/reperfusion (MI/R) injury remains largely unknown. This study aimed to investigate the effects of ICT treatment on MI/R injury and the underlying mechanisms. Rats were subjected to 30 min of myocardial ischemic insult followed by 3 h of reperfusion. ICT (3, 10, and 30 mg/kg) was administered intraperitoneally 10 min before reperfusion. ICT treatment at the dose of 10 and 30 mg/kg improved cardiac function and limited infarct size following MI/R. Meanwhile, ICT reduced plasma creatine kinase (CK), lactate dehydrogenase (LDH) activities and cardiomyocyte apoptosis in I/R heart tissue. Moreover, ICT treatment not only inhibited the pro-inflammatory cytokine TNF-α production and increased the anti-inflammatory cytokine IL-10 level in myocardium but also reduced the increase in the generation of superoxide content and malondialdehyde (MDA) formation and simultaneously increased the anti-oxidant capability in I/R hearts. Furthermore, ICT treatment increased Akt phosphorylation and inhibited PTEN expression in I/R hearts. PI3K inhibitor wortmannin inhibited ICT-enhanced Akt phosphorylation, and blunted ICT-mediated anti-oxidative and anti-inflammatory effects and cardioprotection. Our study indicated for the first time that ICT reduces inflammation and oxidative stress and protects against MI/R injury in rats, which might be via a PI3K–Akt-dependent mechanism.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Cardiothoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
- Department of Cardiothoracic Surgery, Peace Hospital of Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Baichun Xing
- Department of Anesthesiology, Peace Hospital of Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Linlin Yang
- Department of Anesthesiology, Peace Hospital of Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Jialun Shi
- Department of Cardiothoracic Surgery, Peace Hospital of Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Xinmin Zhou
- Department of Cardiothoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
35
|
Lei SW, Cui G, Leung GPH, Luk SCW, Hoi MPM, Wang L, Mahady GB, Lee SMY. Icaritin protects against oxidative stress-induced injury in cardiac H9c2 cells via Akt/Nrf2/HO-1 and calcium signalling pathways. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.06.054] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
|
36
|
Affiliation(s)
- Dennis Schade
- Department
of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse
6, 44227 Dortmund, Germany
| | - Alleyn T. Plowright
- Department
of Medicinal Chemistry, Cardiovascular and Metabolic Diseases Innovative
Medicines, AstraZeneca, Pepparedsleden 1, Mölndal, 43183, Sweden
| |
Collapse
|
37
|
Li C, Li Q, Mei Q, Lu T. Pharmacological effects and pharmacokinetic properties of icariin, the major bioactive component in Herba Epimedii. Life Sci 2015; 126:57-68. [PMID: 25634110 DOI: 10.1016/j.lfs.2015.01.006] [Citation(s) in RCA: 183] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 12/22/2014] [Accepted: 01/10/2015] [Indexed: 12/22/2022]
Abstract
Herba Epimedii is an important medicinal plant which has been used in various traditional Chinese formulations for thousands of years as well as in modern proprietary traditional Chinese medicine products. It has extensive clinical indications, especially for the treatment of sexual dysfunction and osteoporosis. There have been more than 260 chemical moieties identified in the genus Epimedium most of which belong to flavonoids. Icariin is the most abundant constituent in Herba Epimedii. Icariin is pharmacologically bioactive and demonstrates extensive therapeutic capacities such as osteoprotective effect, neuroprotective effect, cardiovascular protective effect, anti-cancer effect, anti-inflammation effect, immunoprotective effect and reproductive function. Particularly, the significant osteogenic effect of icariin made it a promising drug candidate in bone tissue engineering. The current review paper aims to summarize the literatures reporting the pharmacological effects of icariin. The pharmacokinetic properties of bioactive ingredients in Herba Epimedii have also been discussed.
Collapse
Affiliation(s)
- Chenrui Li
- Key Laboratory for Space Biosciences & Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Qiang Li
- Department of Radiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Qibing Mei
- Key Laboratory for Space Biosciences & Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Tingli Lu
- Key Laboratory for Space Biosciences & Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.
| |
Collapse
|
38
|
Sun L, Peng Q, Qu L, Gong L, Si J. Anticancer agent icaritin induces apoptosis through caspase-dependent pathways in human hepatocellular carcinoma cells. Mol Med Rep 2014; 11:3094-100. [PMID: 25434584 DOI: 10.3892/mmr.2014.3007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 10/31/2014] [Indexed: 11/06/2022] Open
Abstract
Icaritin is an active ingredient derived from the plant Herba epimedium, which exhibits various pharmacological and biological activities. However, the function, and the underlying mechanisms of icaritin on the growth of SMMC‑7721 human hepatoma cells have yet to be elucidated. The present study aimed to investigate the function and underlying mechanisms of icaritin in the growth of SMMC‑7721 cells. The cells were treated with varying concentrations of icaritin for 12, 24 and 48 h, respectively, prior to cytotoxic analysis. Apoptosis of SMMC‑7721 cells following treatment with icaritin was measured using flow cytometry. The gene expression of mitochondria‑ and Fas‑mediated caspase‑dependent pathways was detected by reverse transcription‑quantitative polymerase chain reaction and western blotting. Statistical analysis was performed by Student's t‑test and one‑way analysis or variance. The present study demonstrated that treatment with icaritin significantly inhibited growth, and induced apoptosis of SMMC‑7721 cells, in a time‑ and dose‑dependent manner. In addition, icaritin triggered the mitochondrial/caspase apoptotic pathway, by decreasing the Bcl‑2/Bax protein ratio and increasing activation of caspase‑3. Icaritin also activated the Fas‑mediated apoptosis pathway, as was evident by the increased expression levels of Fas and activation of caspase‑8. These data suggest that icaritin may be a potent growth inhibitor and induce apoptosis of SMMC‑7721 cells through the mitochondria‑ and Fas‑mediated caspase‑dependent pathways. The present study may provide experimental evidence for preclinical and clinical evaluations of icaritin for HCC therapy.
Collapse
Affiliation(s)
- Li Sun
- Department of Laboratory Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Qisong Peng
- Department of Laboratory Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu 211100, P.R. China
| | - Lili Qu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Lailing Gong
- Department of Laboratory Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Jin Si
- Department of Laboratory Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| |
Collapse
|
39
|
Zhang SQ. Biodistribution evaluation of icaritin in rats by ultra-performance liquid chromatography-tandem mass spectrometry. JOURNAL OF ETHNOPHARMACOLOGY 2014; 155:1382-1387. [PMID: 25086407 DOI: 10.1016/j.jep.2014.07.045] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 07/10/2014] [Accepted: 07/19/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Icaritin (ICT) is a major bioactive prenylflavonoid derivative contained in the Epimedium which is a widely used herbal medicine for the treatment of infertility, impotence, cardiovascular and skeletal diseases listed in the Chinese Pharmacopoeia. The aim of this study is to investigate the tissue distribution of ICT in rats by ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) MATERIALS AND METHODS: ICT was intraperitoneally administrated to rats for 7 consecutive days at dose levels of 20, 40 and 60 mg/kg/day, respectively. Various tissue homogenates were pretreated by protein precipitation with acetonitrile. ICT and internal standard coumestrol were separated on a BEH C18 column with a gradient mobile phase and detected using precursor-product ion transitions of m/z 367.1→297.1 for ICT and 267.0→211.1 for coumestrol at the negative ionization mode, respectively. RESULTS ICT was widely distributed in rat's various tissues and its concentrations in tissues increased with elevated doses. A sensitive and reliable UPLC-MS/MS method was firstly established to quantify ICT in rat tissues. The lower limit of quantification was 0.5 ng/mL based on 100 μL of tissue homogenates. The intra- and inter-day accuracy at all levels fell in the ranges of 90.8-103.4% and 91.6-100.3%, and the intra- and inter-day precision (RSD) were in the ranges of 2.9-10.5% and 2.6-9.1%, respectively. CONCLUSIONS The UPLC-MS/MS showed good accuracy, precision and recovery and was suitable for the quantification of ICT in rat tissues. Wide distribution of ICT could helpfully elucidate systemic effects and various functions of ICT.
Collapse
Affiliation(s)
- Shuang-Qing Zhang
- Department of Nutrition and Metabolism, National Institute for Nutrition and Food Safety, China Center for Disease Control and Prevention, Beijing 100050, China.
| |
Collapse
|
40
|
Liu P, Jin X, Lv H, Li J, Xu W, Qian HH, Yin Z. Icaritin ameliorates carbon tetrachloride-induced acute liver injury mainly because of the antioxidative function through estrogen-like effects. In Vitro Cell Dev Biol Anim 2014; 50:899-908. [DOI: 10.1007/s11626-014-9792-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Accepted: 06/19/2014] [Indexed: 12/16/2022]
|
41
|
Wang XF, Wang J. Icaritin suppresses the proliferation of human osteosarcoma cells in vitro by increasing apoptosis and decreasing MMP expression. Acta Pharmacol Sin 2014; 35:531-9. [PMID: 24608674 DOI: 10.1038/aps.2013.178] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Accepted: 11/15/2013] [Indexed: 01/13/2023] Open
Abstract
AIM To explore whether icaritin, a prenylflavonoid derivative of the Chinese tonic herb Epimedium, could suppress the proliferation of human osteosarcoma cells in vitro, and to elucidate the mechanisms of the action. METHODS Human osteosarcoma SaOS2 cell line was used in the present study. The proliferation of the cells was examined using MTT assay and immunofluorescence DAPI staining. Cell motility was studied with the scratch assay. Cell apoptosis was determined by Annexin V-FITC and PI double staining using flow cytometry. Western blotting and RT-PCR were used to measure the expression of mRNAs and proteins in the cells. RESULTS Icaritin (5-15 μmol/L) suppressed the proliferation of SaOS2 cells in vitro in a dose-dependent manner. Furthermore, the cell motility was significantly decreased after exposure to icaritin. Moreover, icaritin (5 μmol/L) time-dependently induced the apoptosis of SaOS2 cells, markedly suppressed MMP-2 and MMP-9 expression, upregulated caspase-3 and caspase-9 expression, and increased the level of cleaved caspase-3 in the cells. Co-exposure to the caspase-3 inhibitor zVAD-fmk (10 μmol/L) compromised the icaritin-induced caspase-3 expression and apoptosis in SaOS2 cells. CONCLUSION Icaritin suppresses the proliferation of SaOS2 human osteosarcoma cells by increasing apoptosis and downregulating MMP expression.
Collapse
|
42
|
Icaritin attenuates cigarette smoke-mediated oxidative stress in human lung epithelial cells via activation of PI3K-AKT and Nrf2 signaling. Food Chem Toxicol 2014; 64:307-13. [DOI: 10.1016/j.fct.2013.12.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 11/26/2013] [Accepted: 12/03/2013] [Indexed: 11/23/2022]
|
43
|
Icaritin inhibits JAK/STAT3 signaling and growth of renal cell carcinoma. PLoS One 2013; 8:e81657. [PMID: 24324713 PMCID: PMC3855768 DOI: 10.1371/journal.pone.0081657] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 10/15/2013] [Indexed: 12/20/2022] Open
Abstract
Signal transducer and activator of transcription-3 (STAT3) is critical for cancer progression by regulating tumor cell survival, proliferation, and angiogenesis. Herein, we investigated the regulation of STAT3 activation and the therapeutic effects of Icaritin, a prenyl flavonoid derivative from Epimedium Genus, in renal cell carcinoma (RCC). Icaritin showed significant anti-tumor activity in the human and mouse RCC cell lines, 786-O and Renca, respectively. Icaritin inhibited both constitutive and IL-6-induced phospho-STAT3 (STAT3(Y705)) and reduced the level of STAT3-regulated proteins Bcl-xL, Mcl-1, Survivin, and CyclinD1 in a dose-dependent manner. Icaritin also inhibited activation of Janus-activated kinase-2 (JAK2), while it showed minimal effects on the activation of other key signaling pathways, including AKT and MAPK. Expression of the constitutively active form of STAT3 blocked Icaritin-induced apoptosis, while siRNA directed against STAT3 potentiated apoptosis. Finally, Icaritin significantly blunted RCC tumor growth in vivo, reduced STAT3 activation, and inhibited Bcl-xL and Cyclin E, as well as VEGF expression in tumors, which was associated with reduced tumor angiogenesis. Overall, these results suggest that Icaritin strongly inhibits STAT3 activation and is a potentially effective therapeutic option for the treatment of renal cell carcinoma.
Collapse
|
44
|
Sun L, Chen W, Qu L, Wu J, Si J. Icaritin reverses multidrug resistance of HepG2/ADR human hepatoma cells via downregulation of MDR1 and P‑glycoprotein expression. Mol Med Rep 2013; 8:1883-7. [PMID: 24145579 DOI: 10.3892/mmr.2013.1742] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Accepted: 10/11/2013] [Indexed: 11/06/2022] Open
Abstract
Multidrug resistance (MDR) of tumor cells is a serious obstacle encountered in cancer treatment. In the current study a multiple drug‑resistant HepG2/adriamycin (HepG2/ADR) cell line was established and its MDR was characterized. Icaritin, an active ingredient isolated from the medical plant Herba Epimedium, was observed to reverse MDR in the present model. Icaritin significantly increased the intracellular accumulation of ADR and decreased the expression of the MDR1 gene in HepG2/ADR cells compared with drug‑sensitive HepG2 cells. In addition, the present results showed that icaritin may significantly downregulate the expression of P‑glycoprotein. These results indicate that icaritin is a novel and potent MDR reversal agent and may be a promising drug for tumor chemotherapy.
Collapse
Affiliation(s)
- Li Sun
- Department of Laboratory Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | | | | | | | | |
Collapse
|
45
|
Hong J, Zhang Z, Lv W, Zhang M, Chen C, Yang S, Li S, Zhang L, Han D, Zhang W. Icaritin synergistically enhances the radiosensitivity of 4T1 breast cancer cells. PLoS One 2013; 8:e71347. [PMID: 23977023 PMCID: PMC3744569 DOI: 10.1371/journal.pone.0071347] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 07/01/2013] [Indexed: 01/15/2023] Open
Abstract
Icaritin (ICT) is a hydrolytic form of icariin isolated from plants of the genus Epimedium. This study was to investigate the radiosensitization effect of icaritin and its possible underlying mechanism using murine 4T1 breast cancer cells. The combination of Icaritin at 3 µM or 6 µM with 6 or 8 Gy of ionizing radiation (IR) in the clonogenic assay yielded an ER (enhancement ratio) of 1.18 or 1.28, CI (combination index) of 0.38 or 0.19 and DRI (dose reducing index) of 2.51 or 5.07, respectively. These strongly suggest that Icaritin exerted a synergistic killing (?) effect with radiation on the tumor cells. This effect might relate with bioactivities of ICT: 1) exert an anti-proliferative effect in a dose- and time-dependent manner, which is different from IR killing effect but likely work together with the IR effect; 2) suppress the IR-induced activation of two survival paths, ERK1/2 and AKT; 3) induce the G2/M blockage, enhancing IR killing effect; and 4) synergize with IR to enhance cell apoptosis. In addition, ICT suppressed angiogenesis in chick embryo chorioallantoic membrane (CAM) assay. Taken together, ICT is a new radiosensitizer and can enhance anti-cancer effect of IR or other therapies.
Collapse
Affiliation(s)
- Jinsheng Hong
- Department of Radiation Oncology, First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Division of Radiation Biology, Central Research Lab, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Zhenhuan Zhang
- Department of Radiation Oncology, UF Shands Cancer Center, Gainesville, Florida, United States of America
| | - Wenlong Lv
- Department of Radiation Oncology, First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Mei Zhang
- Department of Radiation Oncology, UF Shands Cancer Center, Gainesville, Florida, United States of America
| | - Chun Chen
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Shanmin Yang
- Department of Radiation Oncology, UF Shands Cancer Center, Gainesville, Florida, United States of America
| | - Shan Li
- Department of Radiation Oncology, First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Lurong Zhang
- Department of Radiation Oncology, UF Shands Cancer Center, Gainesville, Florida, United States of America
| | - Deping Han
- Division of Radiation Biology, Central Research Lab, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- * E-mail: (WZ); (DH)
| | - Weijian Zhang
- Department of Radiation Oncology, First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Division of Radiation Biology, Central Research Lab, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- * E-mail: (WZ); (DH)
| |
Collapse
|
46
|
Wang XL, Wang N, Zheng LZ, Xie XH, Yao D, Liu MY, Yao ZH, Dai Y, Zhang G, Yao XS, Qin L. Phytoestrogenic molecule desmethylicaritin suppressed adipogenesis via Wnt/β-catenin signaling pathway. Eur J Pharmacol 2013; 714:254-60. [PMID: 23792141 PMCID: PMC7094326 DOI: 10.1016/j.ejphar.2013.06.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Revised: 06/03/2013] [Accepted: 06/08/2013] [Indexed: 11/18/2022]
Abstract
Epimedium flavonoids inhibit extravascular lipid deposition during prevention of steroid-associated osteonecrosis. Desmethylicaritin is a bioactive metabolite of Epimedium flavonoids in serum. As it is well known that estrogen inhibits aidpogenesis, so we hypothesized that desmethylicaritin as a phytoestrogen might have the potential to inhibit lipid deposition. This study was designed to investigate the effect of desmethylicaritin on adipogenesis and its underlying mechanism in vitro. Adipogenesis was assessed by Oil Red O staining in 3T3-L1 preadipocytes. Bromodeoxyuridine was used to test the clonal expansion. Further, the mRNA level and protein expression of adipgenic and related factors were detected by qRT-PCR and western blot, respectively. The nuclear location of β-catenin was identified using immunofluoresence assay. Our results showed that desmethylicaritin suppressed the adipogenesis in 3T3-L1 cells in a dose-dependent manner. In addition, desmethylicaritin inhibited clonal expansion during adipogenesis. Desmethylicaritin did not affect CCAAT/enhancer binding protein δ and β mRNA expression, but decreased the mRNA expression of CCAAT/enhancer binding protein α, peroxisome proliferator-activated receptor γ, adipocyte lipid-binding protein and lipoprotein lipase. Desmethylicaritin up-regulated the mRNA expression of Wnt10b that was however down-regulated after adipogenic induction. Desmethylicaritin increased the protein expression of β-catenin both in the cytoplasm and nuclei and immunofluorescence results confirmed that desmethylicaritin increased nuclear translocation of β-catenin. Above findings implied that desmethylicaritin was able to inhibit adipogenesis and Wnt/β-catenin signaling pathway was regulated by desmethylicaritin in the process of suppression of adipogenesis. Above findings supported desmethylicaritin as a novel phytochemical agent for potential prevention of disorders involving lipid metabolism.
Collapse
Affiliation(s)
- Xin-Luan Wang
- Translational Medicine R&D Center, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Yan J, Liu Q, Dou Y, Hsieh Y, Liu Y, Tao R, Zhu D, Lou Y. Activating glucocorticoid receptor-ERK signaling pathway contributes to ginsenoside Rg1 protection against β-amyloid peptide-induced human endothelial cells apoptosis. JOURNAL OF ETHNOPHARMACOLOGY 2013; 147:456-466. [PMID: 23538162 DOI: 10.1016/j.jep.2013.03.039] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 03/10/2013] [Accepted: 03/11/2013] [Indexed: 06/02/2023]
Abstract
The deposition of β-amyloid (Aβ) in neurons and vascular cells of the brain has been characterized in Alzheimer's disease. Ginsenoside Rg1 (Rg1) is an active components in Panax ginseng, a famous traditional Chinese medicines recorded in Compendium of Materia Medica. Present study attempted to evaluate the potential mechanisms of Aβ-mediated insult and the protective effects of Rg1 on human endothelial cells. Rg1 attenuated the Aβ25-35-associated mitochondrial apoptotic events, accompanied by inhibiting HIF-1α expression followed by intracellular reactive nitrogen species generation, and protein nitrotyrosination. These protective effects were abolished by glucocorticoid receptor (GR) antagonist RU486 or p-ERK inhibitor U0126 rather than estrogen receptor α antagonist ICI 82,780. Taken together, our results suggested that Rg1 protected against Aβ25-35-induced apoptosis at least in part by two complementary GR-dependent ERK phosphorylation pathways: (1) down-regulating HIF-1α initiated protein nitrotyrosination, and (2) inhibiting mitochondrial apoptotic cascades. These data provided a novel insight to the mechanisms of Rg1protective effects on Aβ25-35-induced endothelial cells apoptosis, suggesting that GR-ERK signaling pathway might play an important role in it.
Collapse
Affiliation(s)
- Jieping Yan
- Institute of Pharmacology, Toxicology and Biochemical Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Hu B, El Haj AJ. Methionine sulfoxide reductase A as a marker for isolating subpopulations of stem and progenitor cells used in regenerative medicine. Med Hypotheses 2013; 80:663-5. [DOI: 10.1016/j.mehy.2013.01.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 01/19/2013] [Indexed: 10/27/2022]
|
49
|
Zhou L, Huang Y, Zhang Y, Zhao Q, Zheng B, Lou Y, Zhu D. mGluR5 stimulating Homer-PIKE formation initiates icariin induced cardiomyogenesis of mouse embryonic stem cells by activating reactive oxygen species. Exp Cell Res 2013; 319:1505-14. [PMID: 23524143 DOI: 10.1016/j.yexcr.2013.03.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 01/25/2013] [Accepted: 03/02/2013] [Indexed: 10/27/2022]
Abstract
Icariin (ICA) has been reported to facilitate cardiac differentiation of mouse embryonic stem (ES) cells; however, the mechanism by which ICA induced cardiomyogenesis has not been fully elucidated yet. Here, an underlying signaling network including metabotropic glutamate receptor 5 (mGluR5), Homer, phosphatidylinositol 3-Kinase Enhancer (PIKE), phosphatidylinositol 3-Kinase (PI3K), reactive oxygen species (ROS) and nuclear factor-kappaB (NF-κB) was investigated in ICA induced cardiomyogenesis. Our results showed that the co-expression of mGluR5 together with α-actinin or Troponin T in embryoid bodies (EBs) treated with ICA was elevated to 10.86% and 9.62%, compared with the case in the control (4.04% and 3.45%, respectively). Exposure of EBs to ICA for 2 h remarkably increased the dimeric form of mGluR5, which was inhibited by small interfering RNA targeting mGluR5 (si-mGluR5). Moreover, the extracellular glutamate concentration in ICA treatment medium was elevated to 28.9±3.5 μM. Furthermore, the activation of mGluR5 by ICA triggered the formation of Homer-PIKE complex and activated PI3K, stimulating ROS generation and NF-κB nuclear translocation. Knockdown of mGluR5 or inhibition of PI3K by LY294002 blocked ICA induced cardiomyogenesis via repressing mGluR5 pathway, reducing ROS and NF-κB activation. These results revealed that the inducible mechanisms of ICA were related to activate mGluR5 pathway.
Collapse
Affiliation(s)
- Limin Zhou
- Institute of Pharmacology, Toxicology and Biochemical Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, No. 866, Yu Hang Tang Road, Hangzhou 310058, China
| | | | | | | | | | | | | |
Collapse
|
50
|
Sun J, He W, Bai SZ, Peng X, Zhang N, Li HX, Zhang WH, Wang LN, Shao XQ, He YQ, Yang GD, Wu LY, Wang R, Xu CQ. The expression of calcium-sensing receptor in mouse embryonic stem cells (mESCs) and its influence on differentiation of mESC into cardiomyocytes. Differentiation 2013; 85:32-40. [PMID: 23314289 DOI: 10.1016/j.diff.2012.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 11/16/2012] [Accepted: 11/27/2012] [Indexed: 01/22/2023]
Abstract
The calcium-sensing receptor (CaSR), a G protein coupled receptor, is involved in a number of physiological and pathological processes. Embryonic stem cells (ESCs) have a potential role to differentiate into all types of cells. Whether CaSR is functionally expressed in ESCs is unclear. In this study, the expression and distribution of CaSR in 129 mouse ES-D3 cell lines were detected by Western blotting and immunofluorescence; and the intracellular calcium concentration ([Ca(2+)]i) was measured using Laser Confocal Scanning Microscopy. Mouse embryonic stem cells (mESCs) were cultured to embryoid bodies (EBs) and the differentiation of EBs into cardiomyocytes was induced by icariin (ICA). The cardiac specific proteins, a-Actinin and cardiac troponin-I (cTnI), were analyzed by immunofluorescence, and the differentiation rate was analyzed by flow cytometry. The expression of cardiac-specific transcription factors, Nkx2.5 and GATA-4, was detected by Western blotting. We found that the CaSR protein exists in both mESCs and mESC-derived cardiomyocytes (mESC-CMs). Increasing extracellular calcium or neomycin (an agonist of CaSR) increased [Ca(2+)]i and the differentiation rate. These effects were abolished by inhibition of CaSR, phospholipase C, IP3 receptor and Ca(2+) ATPase, or by depletion of the sarcoplasmic reticulum Ca(2+) store, respectively. Activation of CaSR up-regulated protein expression of Nkx2.5 and GATA4 in EBs at an early stage of ICA-induced differentiation. In conclusion, CaSR is functionally expressed in mESCs, and activation of CaSR is involved in the differentiation of mESCs into cardiomyocytes by facilitating the expression of NKx2.5 and GATA-4.
Collapse
Affiliation(s)
- Jian Sun
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|