1
|
Zhao S, Dong G, Guo Y, Sun Y, Li M, Sha B, Huang W, Zhang Y, Du Y, Yan J, Ma Y, Yang R, Shi J, Li P, Hu T, Chen P. NSC632839 suppresses esophageal squamous cell carcinoma cell proliferation in vitro by triggering spindle assembly checkpoint-mediated mitotic arrest and CREB-Noxa-dependent apoptosis. Cancer Cell Int 2025; 25:198. [PMID: 40450316 DOI: 10.1186/s12935-025-03831-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 05/15/2025] [Indexed: 06/03/2025] Open
Abstract
OBJECTIVE Esophageal cancer is one of the most common digestive cancers in the world. Because of the limitation and resistence of the traditional chemotherapy drugs, it is important to explore new therapeutic targets and strategies for this refractory cancer. Recently, targeting deubiquitinases has emerged as a promising avenue for the development of anti-tumor drugs. However, the role and underlying mechanism of NSC632839, a broad-spectrum deubiquitinases inhibitor, in esophageal squamous cell carcinoma in vitro remain elusive. METHODS Cell Counting Kit-8 assay, colony formation assay, EdU proliferation experiment and cell morphology observation were used to detect the effect of NSC632839 on cell growth. Flow cytometry was employed to detect cell apoptosis and cell cycle arrest. Immunoblot and immunofluorescence was used to evaluate the expression level of cell cycle-, apoptosis-, and autophagy-related proteins. RESULTS NSC632839 inhibited the proliferation of Kyse30 and Kyse450 cells. Mechanistically, NSC632839 induced the formation of multipolar spindles, and its concomitant spindle assembly checkpoint-dependent mitotic arrest, followed by CREB-Noxa-mediated apoptosis. Reversine, a classical MPS1 kinase inhibitor known for its ability to inhibit the spindle assembly checkpoint, could rescue NSC632839-induced cell cycle arrest and apoptosis. Additionally, NSC632839 could trigger pro-survival autophagy. Combination of autophagy inhibitor, CQ and BafA1, with NSC632839 could induce stronger cell proliferation inhibition and apoptosis than NSC632839 alone. CONCLUSIONS These findings provided a novel anti-cancer mechanism of NSC632839 and highlighted it as a potential anti-tumor agent for the treatment of esophageal cancer.
Collapse
Affiliation(s)
- Shan Zhao
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- XinJiang Hotan College, Xinjiang, 848000, China
| | - Guihong Dong
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yixuan Guo
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yaxin Sun
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Sanquan College of Xinxiang Medical University, Xinxiang, 453003, China
| | - Miaomiao Li
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Beibei Sha
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- The Second Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450014, China
| | - Wenjing Huang
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yuan Zhang
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yue Du
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jie Yan
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, China
| | - Yangcheng Ma
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, China
| | - Ruiyi Yang
- The Nursing College of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, China
| | - Jianxiang Shi
- Precision Medicine Center, Henan Institute of Medical and Pharmaceutical Sciences & BGI College, Zhengzhou University, Zhengzhou, 450052, China
| | - Pei Li
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Tao Hu
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| | - Ping Chen
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
2
|
Cimmino A, Gioia M, Clementi ME, Faraoni I, Marini S, Ciaccio C. Polydatin-Induced Shift of Redox Balance and Its Anti-Cancer Impact on Human Osteosarcoma Cells. Curr Issues Mol Biol 2024; 47:21. [PMID: 39852136 PMCID: PMC11764470 DOI: 10.3390/cimb47010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/26/2025] Open
Abstract
Cancer cells demonstrate remarkable resilience by adapting to oxidative stress and undergoing metabolic reprogramming, making oxidative stress a critical target for cancer therapy. This study explores, for the first time, the redox-dependent anticancer effects of Polydatin (PD), a glucoside derivative of resveratrol, on the human Osteosarcoma (OS) cells SAOS-2 and U2OS. Using cell-based biochemical assays, we found that cytotoxic doses of PD (100-200 µM) promote ROS production, deplete glutathione (GSH), and elevate levels of both total iron and intracellular malondialdehyde (MDA), which are key markers of ferroptosis. Notably, the ROS scavenger N-acetylcysteine (NAC) and the ferroptosis inhibitor ferrostatin-1 (Fer-1) partially reverse PD's cytotoxic effects. Interestingly, PD's ability to hinder cell adhesion and migration appears independent of its pro-oxidant effect. Analysis of the oxidative stress regulators SIRT1 and Nrf2 at the gene and protein levels using real-time PCR and Western blot indicates an early oxidative response to PD treatment. PD remains effective under tumor-like conditions of hypoxia and serum starvation, and sensitizes OS cells to ROS-inducing chemotherapeutics like doxorubicin (DOX) and cisplatin (CIS). Importantly, PD exhibits minimal toxicity to non-tumorigenic cells (hFOB), suggesting a favorable therapeutic profile. Overall, our findings underscore that PD-induced redox imbalance plays a crucial role in its anti-OS effects, warranting further exploration into the molecular mechanisms behind its pro-oxidant activity.
Collapse
Affiliation(s)
- Alessio Cimmino
- Department of Clinical Sciences and Translational Medicine, University of Rome ‘Tor Vergata’, Via Montpellier 1, 00133 Rome, Italy; (A.C.); (M.G.); (S.M.)
| | - Magda Gioia
- Department of Clinical Sciences and Translational Medicine, University of Rome ‘Tor Vergata’, Via Montpellier 1, 00133 Rome, Italy; (A.C.); (M.G.); (S.M.)
| | - Maria Elisabetta Clementi
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta” SCITEC-CNR, Largo Francesco Vito 1, 00168 Rome, Italy;
| | - Isabella Faraoni
- Department of Systems Medicine, University of Rome ‘Tor Vergata’, Via Montpellier 1, 00133 Rome, Italy;
| | - Stefano Marini
- Department of Clinical Sciences and Translational Medicine, University of Rome ‘Tor Vergata’, Via Montpellier 1, 00133 Rome, Italy; (A.C.); (M.G.); (S.M.)
| | - Chiara Ciaccio
- Department of Clinical Sciences and Translational Medicine, University of Rome ‘Tor Vergata’, Via Montpellier 1, 00133 Rome, Italy; (A.C.); (M.G.); (S.M.)
| |
Collapse
|
3
|
Wu S, Zhao Q, Liu S, Kuang J, Zhang J, Onga A, Shen Y, Wang J, Sui H, Ni L, Ye Y, Tu X, Le HB, Zheng Y, Cui R, Zhu W. Polydatin, a potential NOX5 agonist, synergistically enhances antitumor activity of cisplatin by stimulating oxidative stress in non‑small cell lung cancer. Int J Oncol 2024; 65:77. [PMID: 38873997 PMCID: PMC11251743 DOI: 10.3892/ijo.2024.5665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 05/10/2024] [Indexed: 06/15/2024] Open
Abstract
Non‑small cell lung cancer (NSCLC) is one of the major causes of cancer‑related death worldwide. Cisplatin is a front‑line chemotherapeutic agent in NSCLC. Nevertheless, subsequent harsh side effects and drug resistance limit its further clinical application. Polydatin (PD) induces apoptosis in various cancer cells by generating reactive oxygen species (ROS). However, underlying molecular mechanisms of PD and its effects on cisplatin‑mediated antitumor activity in NSCLC remains unknown. MTT, colony formation, wound healing analyses and flow cytometry was employed to investigate the cell phenotypic changes and ROS generation. Relative gene and protein expressions were evaluated by reverse transcription‑quantitative PCR and western blot analyses. The antitumor effects of PD, cisplatin and their combination were evaluated by mouse xenograft model. In the present study, it was found that PD in combination with cisplatin synergistically enhances the antitumor activity in NSCLC by stimulating ROS‑mediated endoplasmic reticulum stress, and the C‑Jun‑amino‑terminal kinase and p38 mitogen‑activated protein kinase signaling pathways. PD treatment elevated ROS generation by promoting expression of NADPH oxidase 5 (NOX5), and NOX5 knockdown attenuated ROS‑mediated cytotoxicity of PD in NSCLC cells. Mice xenograft model further confirmed the synergistic antitumor efficacy of combined therapy with PD and cisplatin. The present study exhibited a superior therapeutic strategy for some patients with NSCLC by combining PD and cisplatin.
Collapse
Affiliation(s)
- Siyuan Wu
- Cellular and Molecular Biology Laboratory, Affiliated Zhoushan Hospital of Wenzhou Medical University, Zhoushan, Zhejiang 316020, P.R. China
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Qi Zhao
- Cellular and Molecular Biology Laboratory, Affiliated Zhoushan Hospital of Wenzhou Medical University, Zhoushan, Zhejiang 316020, P.R. China
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Shengjuan Liu
- Cellular and Molecular Biology Laboratory, Affiliated Zhoushan Hospital of Wenzhou Medical University, Zhoushan, Zhejiang 316020, P.R. China
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Jiayang Kuang
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Ji Zhang
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Annabeth Onga
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Yiwei Shen
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Jiaying Wang
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Hehuan Sui
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Lianli Ni
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Yuxin Ye
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Xinyue Tu
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Han-Bo Le
- Cellular and Molecular Biology Laboratory, Affiliated Zhoushan Hospital of Wenzhou Medical University, Zhoushan, Zhejiang 316020, P.R. China
| | - Yihu Zheng
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Ri Cui
- Cellular and Molecular Biology Laboratory, Affiliated Zhoushan Hospital of Wenzhou Medical University, Zhoushan, Zhejiang 316020, P.R. China
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Wangyu Zhu
- Cellular and Molecular Biology Laboratory, Affiliated Zhoushan Hospital of Wenzhou Medical University, Zhoushan, Zhejiang 316020, P.R. China
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| |
Collapse
|
4
|
Zhang Z, Sun Z, Jia R, Jiang D, Xu Z, Zhang Y, Wu YQ, Wang X. Protective effects of polydatin against bone and joint disorders: the in vitro and in vivo evidence so far. Nutr Res Rev 2024; 37:96-107. [PMID: 37088535 DOI: 10.1017/s0954422423000082] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Polydatin is an active polyphenol displaying multifaceted benefits. Recently, growing studies have noticed its potential therapeutic effects on bone and joint disorders (BJDs). Therefore, this article reviews recent in vivo and in vitro progress on the protective role of polydatin against BJDs. An insight into the underlying mechanisms is also presented. It was found that polydatin could promote osteogenesis in vitro, and symptom improvements have been disclosed with animal models of osteoporosis, osteosarcoma, osteoarthritis and rheumatic arthritis. These beneficial effects obtained in laboratory could be mainly attributed to the bone metabolism-regulating, anti-inflammatory, antioxidative, apoptosis-regulating and autophagy-regulating functions of polydatin. However, studies on human subjects with BJDs that can lead to early identification of the clinical efficacy and adverse effects of polydatin have not been reported yet. Accordingly, this review serves as a starting point for pursuing clinical trials. Additionally, future emphasis should also be devoted to the low bioavailability and prompt metabolism nature of polydatin. In summary, well-designed clinical trials of polydatin in patients with BJD are in demand, and its pharmacokinetic nature must be taken into account.
Collapse
Affiliation(s)
- Zhen Zhang
- Department of Orthopaedics, Hunan Engineering Laboratory of Advanced Artificial Osteo-materials, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
- Department of Spine Surgery, Youyang Tujia and Miao Autonomous County People's Hospital, Chongqing, 409899, People's Republic of China
| | - Zhicheng Sun
- Department of Orthopaedics, Hunan Engineering Laboratory of Advanced Artificial Osteo-materials, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Runze Jia
- Department of Orthopaedics, Hunan Engineering Laboratory of Advanced Artificial Osteo-materials, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Dingyu Jiang
- Department of Orthopaedics, Hunan Engineering Laboratory of Advanced Artificial Osteo-materials, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Zhenchao Xu
- Department of Orthopaedics, Hunan Engineering Laboratory of Advanced Artificial Osteo-materials, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Yilu Zhang
- Department of Orthopaedics, Hunan Engineering Laboratory of Advanced Artificial Osteo-materials, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Yun-Qi Wu
- Department of Orthopaedics, Hunan Engineering Laboratory of Advanced Artificial Osteo-materials, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Xiyang Wang
- Department of Orthopaedics, Hunan Engineering Laboratory of Advanced Artificial Osteo-materials, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| |
Collapse
|
5
|
Ke J, Li MT, Xu S, Ma J, Liu MY, Han Y. Advances for pharmacological activities of Polygonum cuspidatum - A review. PHARMACEUTICAL BIOLOGY 2023; 61:177-188. [PMID: 36620922 PMCID: PMC9833411 DOI: 10.1080/13880209.2022.2158349] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 10/22/2022] [Accepted: 12/08/2022] [Indexed: 06/01/2023]
Abstract
CONTEXT Polygonum cuspidatum Sieb. et Zucc (Polygonaceae), the root of which is included in the Chinese Pharmcopoeia under the name 'Huzhang', has a long history as a medicinal plant and vegetable. Polygonum cuspidatum has been used in traditional Chinese medicine for the treatment of inflammation, hyperlipemia, etc. OBJECTIVE This article reviews the pharmacological action and the clinical applications of Polygonum cuspidatum and its extracts, whether in vivo or in vitro. We also summarized the main phytochemical constituents and pharmacokinetics of Polygonum cuspidatum and its extracts. METHODS The data were retrieved from major medical databases, such as CNKI, PubMed, and SinoMed, from 2014 to 2022. Polygonum cuspidatum, pharmacology, toxicity, clinical application, and pharmacokinetics were used as keywords. RESULTS The rhizomes, leaves, and flowers of Polygonum cuspidatum have different phytochemical constituents. The plant contains flavonoids, anthraquinones, and stilbenes. Polygonum cuspidatum and the extracts have anti-inflammatory, antioxidation, anticancer, heart protection, and other pharmacological effects. It is used in the clinics to treat dizziness, headaches, traumatic injuries, and water and fire burns. CONCLUSIONS Polygonum cuspidatum has the potential to treat many diseases, such as arthritis, ulcerative colitis, asthma, and cardiac hypertrophy. It has a broad range of medicinal applications, but mainly focused on root medication; its aerial parts should receive more attention. Pharmacokinetics also need to be further investigated.
Collapse
Affiliation(s)
- Jia Ke
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Meng-Ting Li
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuyang Xu
- Monteverde Academy Shanghai, Shanghai, China
| | - Jianpeng Ma
- Multiscale Research Institute of Complex Systems, Fudan University, Shanghai, China
| | - Ming-Yuan Liu
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Han
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
6
|
Cimmino A, Fasciglione GF, Gioia M, Marini S, Ciaccio C. Multi-Anticancer Activities of Phytoestrogens in Human Osteosarcoma. Int J Mol Sci 2023; 24:13344. [PMID: 37686148 PMCID: PMC10487502 DOI: 10.3390/ijms241713344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/19/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
Phytoestrogens are plant-derived bioactive compounds with estrogen-like properties. Their potential health benefits, especially in cancer prevention and treatment, have been a subject of considerable research in the past decade. Phytoestrogens exert their effects, at least in part, through interactions with estrogen receptors (ERs), mimicking or inhibiting the actions of natural estrogens. Recently, there has been growing interest in exploring the impact of phytoestrogens on osteosarcoma (OS), a type of bone malignancy that primarily affects children and young adults and is currently presenting limited treatment options. Considering the critical role of the estrogen/ERs axis in bone development and growth, the modulation of ERs has emerged as a highly promising approach in the treatment of OS. This review provides an extensive overview of current literature on the effects of phytoestrogens on human OS models. It delves into the multiple mechanisms through which these molecules regulate the cell cycle, apoptosis, and key pathways implicated in the growth and progression of OS, including ER signaling. Moreover, potential interactions between phytoestrogens and conventional chemotherapy agents commonly used in OS treatment will be examined. Understanding the impact of these compounds in OS holds great promise for developing novel therapeutic approaches that can augment current OS treatment modalities.
Collapse
Affiliation(s)
| | | | | | | | - Chiara Ciaccio
- Department of Clinical Sciences and Translational Medicine, University of Rome ‘Tor Vergata’, Via Montpellier 1, I-00133 Rome, Italy; (A.C.); (G.F.F.); (M.G.); (S.M.)
| |
Collapse
|
7
|
Li J, Zhang J, Zhu Y, Afolabi LO, Chen L, Feng X. Natural Compounds, Optimal Combination of Brusatol and Polydatin Promote Anti-Tumor Effect in Breast Cancer by Targeting Nrf2 Signaling Pathway. Int J Mol Sci 2023; 24:ijms24098265. [PMID: 37175972 PMCID: PMC10179160 DOI: 10.3390/ijms24098265] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/18/2023] [Accepted: 04/23/2023] [Indexed: 05/15/2023] Open
Abstract
Triple-negative breast cancer (TNBC) has been clearly recognized as a heterogeneous tumor with the worst prognosis among the subtypes of breast cancer (BC). The advent and application of current small-molecule drugs for treating TNBC, as well as other novel inhibitors, among others, have made treatment options for TNBC more selective. However, there are still problems, such as poor patient tolerance, large administration doses, high dosing frequency, and toxic side effects, necessitating the development of more efficient and less toxic treatment strategies. High expression of Nrf2, a vital antioxidant transcription factor, often promotes tumor progression, and it is also one of the most effective targets in BC therapy. We found that in MDA-MB-231 cells and SUM159 cells, brusatol (BRU) combined with polydatin (PD) could significantly inhibit cell proliferation in vitro, significantly downregulate the expression of Nrf2 protein as well as the expression of downstream related target genes Heme Oxygenase-1 (HO-1) and NAD(P)H dehydrogenase, quinone 1 (NQO1), and promote reactive oxygen species (ROS) levels to further strengthen the anti-tumor effect. Furthermore, we discovered in our in vivo experiments that by reducing the drug dosage three times, we could significantly reduce tumor cell growth while avoiding toxic side effects, providing a treatment method with greater clinical application value for TNBC treatment.
Collapse
Affiliation(s)
- Jing Li
- Shenzhen Laboratory of Tumor Cell Biology, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Jianchao Zhang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yan Zhu
- Shenzhen Laboratory of Tumor Cell Biology, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Lukman O Afolabi
- Shenzhen Laboratory of Tumor Cell Biology, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Liang Chen
- Shenzhen Laboratory of Tumor Cell Biology, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xuesong Feng
- School of Pharmacy, China Medical University, Shenyang 110122, China
| |
Collapse
|
8
|
Mohammadhosseinpour S, Weaver A, Sudhakaran M, Ho LC, Le T, Doseff AI, Medina-Bolivar F. Arachidin-1, a Prenylated Stilbenoid from Peanut, Enhances the Anticancer Effects of Paclitaxel in Triple-Negative Breast Cancer Cells. Cancers (Basel) 2023; 15:399. [PMID: 36672351 PMCID: PMC9856928 DOI: 10.3390/cancers15020399] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is one of the deadliest forms of breast cancer. Investigating alternative therapies to increase survival rates for this disease is essential. To this end, the cytotoxic effects of the prenylated stilbenoids arachidin-1 (A-1) and arachidin-3 (A-3), and non-prenylated resveratrol (RES) were evaluated in human TNBC cell lines as potential adjuvants for paclitaxel (Pac). A-1, alone or in combination with Pac, showed the highest cytotoxicity in TNBC cells. Apoptosis was further evaluated by measuring key apoptosis marker proteins, cell cycle arrest, and intracellular reactive oxygen species (ROS) generation. Furthermore, the cytotoxic effect of A-1 combined with Pac was also evaluated in a 3D spheroid TNBC model. The results showed that A-1 decreased the Pac IC50 approximately 2-fold in TNBC cells. The synergistic combination of A-1 and Pac arrested cells in G2/M phase and activated p53 expression. In addition, the combined treatment increased intracellular ROS generation and induced apoptosis. Importantly, the combination of A-1 with Pac inhibited TNBC spheroid growth. Our results demonstrated that A-1 in combination with Pac inhibited cell proliferation, induced apoptosis through mitochondrial oxidative stress, and reduced TNBC spheroid growth. These findings underscore the impactful effects of the prenylated stilbenoid A-1 as a novel adjuvant for Pac chemotherapy in TNBC treatment.
Collapse
Affiliation(s)
| | - Alexx Weaver
- Arkansas Biosciences Institute, Arkansas State University, Jonesboro, AR 72401, USA
| | - Meenakshi Sudhakaran
- Molecular, Cellular, and Integrative Physiology Graduate Program, Michigan State University, East Lansing, MI 48824, USA
| | - Linh-Chi Ho
- Arkansas Biosciences Institute, Arkansas State University, Jonesboro, AR 72401, USA
| | - Tra Le
- Arkansas Biosciences Institute, Arkansas State University, Jonesboro, AR 72401, USA
| | - Andrea I. Doseff
- Department of Physiology, and Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Fabricio Medina-Bolivar
- Arkansas Biosciences Institute, Arkansas State University, Jonesboro, AR 72401, USA
- Department of Biological Sciences, Arkansas State University, Jonesboro, AR 72401, USA
| |
Collapse
|
9
|
Farooq U, Wang H, Hu J, Li G, Jehan S, Shi J, Li D, Sui G. Polydatin Inhibits Hepatocellular Carcinoma Cell Proliferation and Sensitizes Doxorubicin and Cisplatin through Targeting Cell Mitotic Machinery. Cells 2023; 12:cells12020222. [PMID: 36672157 PMCID: PMC9856937 DOI: 10.3390/cells12020222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/22/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
Polydatin (PD) is a natural compound with anticancer activities, but the underlying mechanisms remain largely unclear. To understand how PD inhibited hepatocellular carcinoma (HCC), we studied PD treatments in HCC HepG2 and SK-HEP1 cells, and normal liver HL-7702 cells. PD selectively blocked the proliferation of HCC cells but showed low toxicity in normal cells, while the effects of doxorubicin (DOX) and cisplatin (DDP) on HCC and normal liver cells were opposite. In the cotreatment studies, PD synergistically improved the inhibitory activities of DOX and DDP in HCC cells but alleviated their toxicity in HL-7702 cells. Furthermore, RNA-seq studies of PD-treated HepG2 cells revealed multiple altered signaling pathways. We identified 1679 Differentially Expressed Genes (DEGs) with over a 2.0-fold change in response to PD treatment. Integrative analyses using the DEGs in PD-treated HepG2 cells and DEGs in a TCGA dataset of HCC patients revealed five PD-repressed DEGs regulating mitotic spindle midzone formation. The expression of these genes showed significantly positive correlation with poor clinical outcomes of HCC patients, suggesting that mitotic machinery was likely a primary target of PD. Our findings improve the understanding of PD's anticancer mechanisms and provide insights into developing effective clinical approaches in HCC therapies.
Collapse
Affiliation(s)
- Umar Farooq
- College of Life Sciences, Northeast Forestry University, 26 Hexing Road, Harbin 150040, China
| | - Hao Wang
- College of Life Sciences, Northeast Forestry University, 26 Hexing Road, Harbin 150040, China
| | - Jingru Hu
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Guangyue Li
- College of Life Sciences, Northeast Forestry University, 26 Hexing Road, Harbin 150040, China
| | - Shah Jehan
- College of Life Sciences, Northeast Forestry University, 26 Hexing Road, Harbin 150040, China
| | - Jinming Shi
- College of Life Sciences, Northeast Forestry University, 26 Hexing Road, Harbin 150040, China
| | - Dangdang Li
- College of Life Sciences, Northeast Forestry University, 26 Hexing Road, Harbin 150040, China
- Correspondence: (D.L.); (G.S.)
| | - Guangchao Sui
- College of Life Sciences, Northeast Forestry University, 26 Hexing Road, Harbin 150040, China
- Correspondence: (D.L.); (G.S.)
| |
Collapse
|
10
|
Shah MA, Hamid A, Faheem HI, Rasul A, Baokbah TAS, Haris M, Yousaf R, Saleem U, Iqbal S, Alves MS, Khan Z, Hussain G, Alsharfi I, Khan H, Jeandet P. Uncovering the Anticancer Potential of Polydatin: A Mechanistic Insight. Molecules 2022; 27:7175. [PMID: 36364001 PMCID: PMC9656535 DOI: 10.3390/molecules27217175] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/17/2022] [Accepted: 10/17/2022] [Indexed: 11/05/2022] Open
Abstract
Polydatin or 3-O-β-d-resveratrol-glucopyranoside (PD), a stilbenoid component of Polygonum cuspicadum (Polygonaceae), has a variety of biological roles. In traditional Chinese medicine, P. cuspicadum extracts are used for the treatment of infections, inflammation, and cardiovascular disorders. Polydatin possesses a broad range of biological activities including antioxidant, anti-inflammatory, anticancer, and hepatoprotective, neuroprotective, and immunostimulatory effects. Currently, a major proportion of the population is victimized with cervical lung cancer, ovarian cancer and breast cancer. PD has been recognized as a potent anticancer agent. PD could effectively inhibit the migration and proliferation of ovarian cancer cells, as well as the expression of the PI3K protein. The malignancy of lung cancer cells was reduced after PD treatments via targeting caspase 3, arresting cancer cells at the S phase and inhibiting NLRP3 inflammasome by downregulation of the NF-κB pathway. This ceases cell cycle, inhibits VEGF, and counteracts ROS in breast cancer. It also prevents cervical cancer by regulating epithelial-to-mesenchymal transition (EMT), apoptosis, and the C-Myc gene. The objective of this review is thus to unveil the polydatin anticancer potential for the treatment of various tumors, as well as to examine the mechanisms of action of this compound.
Collapse
Affiliation(s)
| | - Ayesha Hamid
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Hafiza Ishmal Faheem
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Azhar Rasul
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Tourki A. S. Baokbah
- Department of Medical Emergency Services, College of Health Sciences-AlQunfudah, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Muhammad Haris
- Faculty of Pharmaceutical Sciences, Universiteit Gent, Ghent 9000, Belgium
| | - Rimsha Yousaf
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Uzma Saleem
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Shabnoor Iqbal
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Maria Silvana Alves
- Laboratory of Cellular and Molecular Bioactivity, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Zahid Khan
- Department of Pharmacognosy, Faculty of Pharmacy, Federal Urdu University of Arts, Science & Technology, Karachi 75300, Pakistan
| | - Ghulam Hussain
- Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Ifat Alsharfi
- Department of Biology, Jamoum University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| | - Philippe Jeandet
- Research Unit Induced Resistance and Plant Bioprotection, University of Reims Champagne-Ardenne, USC INRAe 1488, 51100 Reims, France
| |
Collapse
|
11
|
Karami A, Fakhri S, Kooshki L, Khan H. Polydatin: Pharmacological Mechanisms, Therapeutic Targets, Biological Activities, and Health Benefits. Molecules 2022; 27:6474. [PMID: 36235012 PMCID: PMC9572446 DOI: 10.3390/molecules27196474] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/22/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022] Open
Abstract
Polydatin is a natural potent stilbenoid polyphenol and a resveratrol derivative with improved bioavailability. Polydatin possesses potential biological activities predominantly through the modulation of pivotal signaling pathways involved in inflammation, oxidative stress, and apoptosis. Various imperative biological activities have been suggested for polydatin towards promising therapeutic effects, including anticancer, cardioprotective, anti-diabetic, gastroprotective, hepatoprotective, neuroprotective, anti-microbial, as well as health-promoting roles on the renal system, the respiratory system, rheumatoid diseases, the skeletal system, and women's health. In the present study, the therapeutic targets, biological activities, pharmacological mechanisms, and health benefits of polydatin are reviewed to provide new insights to researchers. The need to develop further clinical trials and novel delivery systems of polydatin is also considered to reveal new insights to researchers.
Collapse
Affiliation(s)
- Ahmad Karami
- Student Research Committee, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah 6714415153, Iran
| | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Leila Kooshki
- Student Research Committee, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah 6714415153, Iran
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| |
Collapse
|
12
|
Identification of Molecular Targets and Underlying Mechanisms of Xiaoji Recipe against Pancreatic Cancer Based on Network Pharmacology. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:4640849. [PMID: 36118824 PMCID: PMC9477627 DOI: 10.1155/2022/4640849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022]
Abstract
Traditional Chinese medicine (TCM) is applied in the anticancer adjuvant therapy of various malignancies and pancreatic cancer included. Xiaoji recipe consists several TCM materials with anticancer activities. In our work, we intended to analyze the molecular targets as well as the underlying mechanisms of Xiaoji recipe against pancreatic cancer. A total of 32 active components and 522 potential targets of Xiaoji recipe were selected using the TCMSP and SwissTargetPrediction databases. The potential target gene prediction in pancreatic cancer was performed using OMIM, Disgenet, and Genecards databases, and totally, 998 target genes were obtained. The component-disease network was constructed using the Cytoscape software, and 116 shared targets of pancreatic cancer and Xiaoji recipe were screened out. As shown in the protein–protein interaction (PPI) network, the top 20 hub genes such as TP53, HRAS, AKT1, VEGFA, STAT3, EGFR, and SRC were further selected by degree. GO and KEGG functional enrichment analysis revealed that Xiaoji recipe may affect pancreatic cancer progression by targeting the PI3K/AKT and MAPK signaling pathways. Moreover, we performed in vitro assays to explore the effect of Xiaoji recipe on pancreatic cancer cells. The results revealed that Xiaoji recipe suppressed the viability and migration and promoted the apoptosis of pancreatic cancer cells via the inactivation of PI3K/AKT, MAPK, and STAT3 pathways. The findings of our study suggested the potential of Xiaoji recipe in the targeting therapy of pancreatic cancer.
Collapse
|
13
|
Liu M, Li Y, Kong B, Zhang G, Zhang Q. Polydatin down-regulates the phosphorylation level of STAT3 and induces pyroptosis in triple-negative breast cancer mice with a high-fat diet. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:173. [PMID: 35280371 PMCID: PMC8908165 DOI: 10.21037/atm-22-73] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/11/2022] [Indexed: 12/26/2022]
Abstract
Background To explore the impact of polydatin on mice with triple-negative breast cancer (TNBC) receiving a high-fat diet, as well as the underlying processes. Methods A total of 40 female Balb/c mice were randomly separated into 4 groups (4T1 + polydatin + fat diet group, 4T1 + high-fat diet group, 4T1 + polydatin group, and 4T1 group). To establish the obese TNBC mouse model, TNBC was xenografted 1×105 4T1 cells/50 µL per mouse at the right fourth mammary fat pad under anesthesia and the mice were fed a high fat diet. When the experiment was completed, total plasma cholesterol (TC) and cancer antigen (CA)15-3 were measured. The enzyme-linked immunosorbent assay (ELISA) method was used detect CA15-3. Oil red O staining was used to observe the morphological changes. Western blot analysis and reverse transcription polymerase chain reaction (RT-PCR) were used to detect the corresponding protein expression and the messenger RNA (mRNA) level. Results Polydatin decreased the degree of fatty liver, as determined by oil red O staining. The TC level in the 4T1 + fat diet group was significantly higher, and it was decreased in the 4T1 + polydatin group. The results of ELISA showed that compared with the 4T1 group, CA15-3 was significantly increased in the 4T1 + fat diet group, and polydatin was shown to significantly reduce the expression of CA15-3. Polydatin inhibited p-JAK2 and p-STAT3 mRNA and protein levels. Polydatin increased pyroptosis-related gene mRNA and protein level. Conclusions We believe that polydatin can effectively reduce blood lipid levels in TNBC mice with a high-fat diet, and play an anticancer role in TNBC. The underlying mechanism may be related to the JAK2/STAT3 signaling pathway and pyroptosis in TNBC. Our results contribute to validating the traditional use of polydatin in the treatment of TNBC with hyperlipidemia.
Collapse
Affiliation(s)
- Min Liu
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yinan Li
- Beijing University of Chinese Medicine, Beijing, China
| | - Bingtan Kong
- Beijing University of Chinese Medicine, Beijing, China
| | - Ganlin Zhang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Qing Zhang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
14
|
Gutierrez DA, Contreras L, Villanueva PJ, Borrego EA, Morán-Santibañez K, Hess JD, DeJesus R, Larragoity M, Betancourt AP, Mohl JE, Robles-Escajeda E, Begum K, Roy S, Kirken RA, Varela-Ramirez A, Aguilera RJ. Identification of a Potent Cytotoxic Pyrazole with Anti-Breast Cancer Activity That Alters Multiple Pathways. Cells 2022; 11:254. [PMID: 35053370 PMCID: PMC8773755 DOI: 10.3390/cells11020254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/14/2021] [Accepted: 01/07/2022] [Indexed: 11/26/2022] Open
Abstract
In this study, we identified a novel pyrazole-based derivative (P3C) that displayed potent cytotoxicity against 27 human cancer cell lines derived from different tissue origins with 50% cytotoxic concentrations (CC50) in the low micromolar and nanomolar range, particularly in two triple-negative breast cancer (TNBC) cell lines (from 0.25 to 0.49 µM). In vitro assays revealed that P3C induces reactive oxygen species (ROS) accumulation leading to mitochondrial depolarization and caspase-3/7 and -8 activation, suggesting the participation of both the intrinsic and extrinsic apoptotic pathways. P3C caused microtubule disruption, phosphatidylserine externalization, PARP cleavage, DNA fragmentation, and cell cycle arrest on TNBC cells. In addition, P3C triggered dephosphorylation of CREB, p38, ERK, STAT3, and Fyn, and hyperphosphorylation of JNK and NF-kB in TNBC cells, indicating the inactivation of both p38MAPK/STAT3 and ERK1/2/CREB signaling pathways. In support of our in vitro assays, transcriptome analyses of two distinct TNBC cell lines (MDA-MB-231 and MDA-MB-468 cells) treated with P3C revealed 28 genes similarly affected by the treatment implicated in apoptosis, oxidative stress, protein kinase modulation, and microtubule stability.
Collapse
Affiliation(s)
- Denisse A. Gutierrez
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Lisett Contreras
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Paulina J. Villanueva
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Edgar A. Borrego
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Karla Morán-Santibañez
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Jessica D. Hess
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Rebecca DeJesus
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Manuel Larragoity
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Ana P. Betancourt
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Jonathon E. Mohl
- Department of Bioinformatics, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA;
| | - Elisa Robles-Escajeda
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Khodeza Begum
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Sourav Roy
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Robert A. Kirken
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Armando Varela-Ramirez
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Renato J. Aguilera
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| |
Collapse
|
15
|
Ye P, Wu H, Jiang Y, Xiao X, Song D, Xu N, Ma X, Zeng J, Guo Y. Old dog, new tricks: Polydatin as a multitarget agent for current diseases. Phytother Res 2021; 36:214-230. [PMID: 34936712 DOI: 10.1002/ptr.7306] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/19/2021] [Accepted: 09/22/2021] [Indexed: 12/24/2022]
Abstract
Polydatin (PD) is a natural single-crystal product that is primarily extracted from the traditional plant Polygonum cuspidatum Sieb. et Zucc. Early research showed that PD exhibited a variety of biological activities. PD has attracted increasing research interest since 2014, but no review comprehensively summarized the new findings. A great gap between its biological activities and drug development remains. It is necessary to summarize new findings on the pharmacological effects of PD on current diseases. We propose that PD will most likely be used in cardiac and cerebral ischaemia/reperfusion-related diseases and atherosclerosis in the future. The present work classified these new findings according to diseases and summarized the main effects of PD via specific mechanisms of action. In summary, we found that PD played a therapeutic role in a variety of diseases, primarily via five mechanisms: antioxidative effects, antiinflammatory effects, regulation of autophagy and apoptosis, maintenance of mitochondrial function, and lipid regulation.
Collapse
Affiliation(s)
- Penghui Ye
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hefei Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yinxiao Jiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaolin Xiao
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dan Song
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Nuo Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinhao Zeng
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yaoguang Guo
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
16
|
Bae H, Lee W, Song J, Hong T, Kim MH, Ham J, Song G, Lim W. Polydatin Counteracts 5-Fluorouracil Resistance by Enhancing Apoptosis via Calcium Influx in Colon Cancer. Antioxidants (Basel) 2021; 10:antiox10091477. [PMID: 34573109 PMCID: PMC8469995 DOI: 10.3390/antiox10091477] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022] Open
Abstract
Colon cancer is a disease with a high prevalence rate worldwide, and for its treatment, a 5-fluorouracil (5-FU)-based chemotherapeutic strategy is generally used. However, conventional anticancer agents have some limitations, including the development of drug resistance. Therefore, there has recently been a demand for the improvement of antitumor agents using natural products with low side effects and high efficacy. Polydatin is a natural active compound extracted from an annual plant, and widely known for its anticancer effects in diverse types of cancer. However, it is still not clearly understood how polydatin ameliorates several drawbacks of standard anticancer drugs by reinforcing the chemosensitivity against 5-FU, and neither are the intrinsic mechanisms behind this process. In this study, we examined how polydatin produces anticancer effects in two types of colon cancer, called HCT116 and HT-29 cells. Polydatin has the ability to repress the progression of colon cancer, and causes a modification of distribution in the cell cycle by a flow cytometry analysis. It also induces mitochondrial dysfunctions through oxidative stress and the loss of mitochondrial membrane potential. The present study investigated the apoptosis caused by the disturbance of calcium regulation and the expression levels of related proteins through flow cytometry and immunoblotting analysis. It was revealed that polydatin suppresses the signaling pathways of the mitogen-activated protein kinase (MAPK) and PI3K/AKT. In addition, it was shown that polydatin combined with 5-FU counteracts drug resistance in 5-FU-resistant cells. Therefore, this study suggests that polydatin has the potential to be developed as an innovative medicinal drug for the treatment of colon cancer.
Collapse
Affiliation(s)
- Hyocheol Bae
- Department of Oriental Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Korea;
| | - Woonghee Lee
- Institute of Animal Molecular Biotechnology, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea; (W.L.); (J.H.)
| | - Jisoo Song
- Department of Food and Nutrition, College of Science and Technology, Kookmin University, Seoul 02707, Korea; (J.S.); (T.H.)
| | - Taeyeon Hong
- Department of Food and Nutrition, College of Science and Technology, Kookmin University, Seoul 02707, Korea; (J.S.); (T.H.)
| | - Myung Hyun Kim
- Department of Food and Nutrition, Sookmyung Women’s University, Seoul 04310, Korea;
| | - Jiyeon Ham
- Institute of Animal Molecular Biotechnology, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea; (W.L.); (J.H.)
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea; (W.L.); (J.H.)
- Correspondence: (G.S.); (W.L.); Tel.: +82-2-3290-3881 (G.S.); +82-2-910-4773 (W.L.)
| | - Whasun Lim
- Department of Food and Nutrition, College of Science and Technology, Kookmin University, Seoul 02707, Korea; (J.S.); (T.H.)
- Correspondence: (G.S.); (W.L.); Tel.: +82-2-3290-3881 (G.S.); +82-2-910-4773 (W.L.)
| |
Collapse
|
17
|
Bang TH, Park BS, Kang HM, Kim JH, Kim IR. Polydatin, a Glycoside of Resveratrol, Induces Apoptosis and Inhibits Metastasis Oral Squamous Cell Carcinoma Cells In Vitro. Pharmaceuticals (Basel) 2021; 14:ph14090902. [PMID: 34577602 PMCID: PMC8468100 DOI: 10.3390/ph14090902] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 01/07/2023] Open
Abstract
Although various methods, such as surgery and chemotherapy, are applied to the treatment of OSCC, there are problems, such as functional and aesthetic limitations of the mouth and face, drug side effects, and lymph node metastasis. Many researchers are making efforts to develop new therapeutic agents from plant-derived substances to overcome the side effects that occur in oral cancer treatment. Polydatin is known as a natural precursor of resveratrol, and research on its efficacy is being actively conducted recently. Therefore, we investigated whether polydatin can induce apoptosis and whether it affects cell migration and invasion through the regulation of EMT-related factors in OSCC. Polydatin decreased the survival and proliferation rates of CAL27 and Ca9-22 cells, and induced the release of cytochrome c, a factor related to apoptosis, and fragmentation of procaspase-3 and PARP. Another form of cell death, autophagy, was observed in polydatin-treated cells. In addition, polydatin inhibits cell migration and invasion, and it has been shown to occur through increased expression of E-cadherin, an EMT related factor, and decreased expression of N-cadherin and Slug and Snail proteins and genes. These findings suggest that polydatin is a potential oral cancer treatment.
Collapse
Affiliation(s)
- Tae-Hyun Bang
- Department of Oral Anatomy, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si 50612, Korea; (T.-H.B.); (B.-S.P.); (H.-M.K.)
| | - Bong-Soo Park
- Department of Oral Anatomy, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si 50612, Korea; (T.-H.B.); (B.-S.P.); (H.-M.K.)
- Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Hae-Mi Kang
- Department of Oral Anatomy, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si 50612, Korea; (T.-H.B.); (B.-S.P.); (H.-M.K.)
- Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Jung-Han Kim
- Department of Oral and Maxillofacial Surgery, Medical Center, Dong-A University, Daesingongwon-ro, 26, Seo-gu, Busan 49201, Korea;
| | - In-Ryoung Kim
- Department of Oral Anatomy, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si 50612, Korea; (T.-H.B.); (B.-S.P.); (H.-M.K.)
- Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Correspondence: ; Tel.: +82-51-510-8552
| |
Collapse
|
18
|
Lee J, Han Y, Wang W, Jo H, Kim H, Kim S, Yang KM, Kim SJ, Dhanasekaran DN, Song YS. Phytochemicals in Cancer Immune Checkpoint Inhibitor Therapy. Biomolecules 2021; 11:1107. [PMID: 34439774 PMCID: PMC8393583 DOI: 10.3390/biom11081107] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/08/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022] Open
Abstract
The interaction of immune checkpoint molecules in the tumor microenvironment reduces the anti-tumor immune response by suppressing the recognition of T cells to tumor cells. Immune checkpoint inhibitor (ICI) therapy is emerging as a promising therapeutic option for cancer treatment. However, modulating the immune system with ICIs still faces obstacles with severe immunogenic side effects and a lack of response against many cancer types. Plant-derived natural compounds offer regulation on various signaling cascades and have been applied for the treatment of multiple diseases, including cancer. Accumulated evidence provides the possibility of efficacy of phytochemicals in combinational with other therapeutic agents of ICIs, effectively modulating immune checkpoint-related signaling molecules. Recently, several phytochemicals have been reported to show the modulatory effects of immune checkpoints in various cancers in in vivo or in vitro models. This review summarizes druggable immune checkpoints and their regulatory factors. In addition, phytochemicals that are capable of suppressing PD-1/PD-L1 binding, the best-studied target of ICI therapy, were comprehensively summarized and classified according to chemical structure subgroups. It may help extend further research on phytochemicals as candidates of combinational adjuvants. Future clinical trials may validate the synergetic effects of preclinically investigated phytochemicals with ICI therapy.
Collapse
Affiliation(s)
- Juwon Lee
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Korea; (J.L.); (Y.H.); (W.W.); (H.J.); (H.K.)
- WCU Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea
| | - Youngjin Han
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Korea; (J.L.); (Y.H.); (W.W.); (H.J.); (H.K.)
- SK Biopharmaceuticals Co., Ltd., Seongnam-si 13494, Korea
| | - Wenyu Wang
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Korea; (J.L.); (Y.H.); (W.W.); (H.J.); (H.K.)
- Interdisciplinary Program in Cancer Biology, Seoul National University, Seoul 03080, Korea
| | - HyunA Jo
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Korea; (J.L.); (Y.H.); (W.W.); (H.J.); (H.K.)
- WCU Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea
| | - Heeyeon Kim
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Korea; (J.L.); (Y.H.); (W.W.); (H.J.); (H.K.)
- WCU Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea
| | - Soochi Kim
- Department of Neurology and Neurological Sciences, School of Medicine, Stanford University, Stanford, CA 94304, USA;
| | - Kyung-Min Yang
- MedPacto Inc., 92, Myeongdal-ro, Seocho-gu, Seoul 06668, Korea; (K.-M.Y.); (S.-J.K.)
| | - Seong-Jin Kim
- MedPacto Inc., 92, Myeongdal-ro, Seocho-gu, Seoul 06668, Korea; (K.-M.Y.); (S.-J.K.)
- Precision Medicine Research Center, Advanced Institute of Convergence Technology, Seoul National University, Suwon 16229, Korea
- Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Suwon 16229, Korea
| | - Danny N. Dhanasekaran
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Yong Sang Song
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Korea; (J.L.); (Y.H.); (W.W.); (H.J.); (H.K.)
- WCU Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea
- Interdisciplinary Program in Cancer Biology, Seoul National University, Seoul 03080, Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
19
|
Steven A, Friedrich M, Jank P, Heimer N, Budczies J, Denkert C, Seliger B. What turns CREB on? And off? And why does it matter? Cell Mol Life Sci 2020; 77:4049-4067. [PMID: 32347317 PMCID: PMC7532970 DOI: 10.1007/s00018-020-03525-8] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/21/2020] [Accepted: 04/06/2020] [Indexed: 12/16/2022]
Abstract
Altered expression and function of the transcription factor cyclic AMP response-binding protein (CREB) has been identified to play an important role in cancer and is associated with the overall survival and therapy response of tumor patients. This review focuses on the expression and activation of CREB under physiologic conditions and in tumors of distinct origin as well as the underlying mechanisms of CREB regulation by diverse stimuli and inhibitors. In addition, the clinical relevance of CREB is summarized, including its use as a prognostic and/or predictive marker as well as a therapeutic target.
Collapse
Affiliation(s)
- André Steven
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Michael Friedrich
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Paul Jank
- Institute of Pathology, Philipps University Marburg, 35043, Marburg, Germany
| | - Nadine Heimer
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Jan Budczies
- Institute of Pathology, University Clinic Heidelberg, 69120, Heidelberg, Germany
| | - Carsten Denkert
- Institute of Pathology, Philipps University Marburg, 35043, Marburg, Germany
| | - Barbara Seliger
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany.
| |
Collapse
|
20
|
Le Naour A, Koffi Y, Diab M, Le Guennec D, Rougé S, Aldekwer S, Goncalves-Mendes N, Talvas J, Farges MC, Caldefie-Chezet F, Vasson MP, Rossary A. EO771, the first luminal B mammary cancer cell line from C57BL/6 mice. Cancer Cell Int 2020; 20:328. [PMID: 32699527 PMCID: PMC7372867 DOI: 10.1186/s12935-020-01418-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Background Despite decades of therapeutic trials, effective diagnosis, many drugs available and numerous studies on breast cancer, it remains the deadliest cancer in women. In order to choose the most appropriate treatment and to understand the prognosis of the patients, breast cancer is divided into different subtypes using a molecular classification. Just as there remains a need to discover new effective therapies, models to test them are also required. Methods The EO771 (also named E0771 or EO 771) murine mammary cancer cell line was originally isolated from a spontaneous tumour in C57BL/6 mouse. Although frequently used, this cell line remains poorly characterized. Therefore, the EO771 phenotype was investigated. The phenotype was compared to that of MCF-7 cells, known to be of luminal A subtype and to express estrogen receptors, as well as MDA-MB-231 cells, which are triple negative. Their sensitivity to hormonal treatment was evaluated by viability tests. Results The EO771 were estrogen receptor α negative, estrogen receptor β positive, progesterone receptor positive and ErbB2 positive. This phenotype was associated with a sensitivity to anti-estrogen treatments such as tamoxifen, 4-hydroxy-tamoxifen, endoxifen and fulvestrant. Conclusions On account of the numerous results published with the EO771 cell line, it is important to know its classification, to facilitate comparisons with corresponding types of tumours in patients. Transcriptomic and protein analysis of the EO771 cell line classified it within the luminal B subtype. Luminal B cancers correspond to one of the subtypes most frequently encountered in patients and associated with a poor prognosis.
Collapse
Affiliation(s)
- Augustin Le Naour
- Human Nutrition Unit, ECREIN team, UMR 1019, University of Clermont Auvergne, INRAE, CRNH-Auvergne, TSA 50400, 28 place Henri Dunant, 63000 Clermont-Ferrand Cedex 1, France
| | - Yvonne Koffi
- Human Nutrition Unit, ECREIN team, UMR 1019, University of Clermont Auvergne, INRAE, CRNH-Auvergne, TSA 50400, 28 place Henri Dunant, 63000 Clermont-Ferrand Cedex 1, France
| | - Mariane Diab
- Human Nutrition Unit, ECREIN team, UMR 1019, University of Clermont Auvergne, INRAE, CRNH-Auvergne, TSA 50400, 28 place Henri Dunant, 63000 Clermont-Ferrand Cedex 1, France
| | - Delphine Le Guennec
- Human Nutrition Unit, ECREIN team, UMR 1019, University of Clermont Auvergne, INRAE, CRNH-Auvergne, TSA 50400, 28 place Henri Dunant, 63000 Clermont-Ferrand Cedex 1, France
| | - Stéphanie Rougé
- Human Nutrition Unit, ECREIN team, UMR 1019, University of Clermont Auvergne, INRAE, CRNH-Auvergne, TSA 50400, 28 place Henri Dunant, 63000 Clermont-Ferrand Cedex 1, France
| | - Sahar Aldekwer
- Human Nutrition Unit, ECREIN team, UMR 1019, University of Clermont Auvergne, INRAE, CRNH-Auvergne, TSA 50400, 28 place Henri Dunant, 63000 Clermont-Ferrand Cedex 1, France
| | - Nicolas Goncalves-Mendes
- Human Nutrition Unit, ECREIN team, UMR 1019, University of Clermont Auvergne, INRAE, CRNH-Auvergne, TSA 50400, 28 place Henri Dunant, 63000 Clermont-Ferrand Cedex 1, France
| | - Jérémie Talvas
- Human Nutrition Unit, ECREIN team, UMR 1019, University of Clermont Auvergne, INRAE, CRNH-Auvergne, TSA 50400, 28 place Henri Dunant, 63000 Clermont-Ferrand Cedex 1, France
| | - Marie-Chantal Farges
- Human Nutrition Unit, ECREIN team, UMR 1019, University of Clermont Auvergne, INRAE, CRNH-Auvergne, TSA 50400, 28 place Henri Dunant, 63000 Clermont-Ferrand Cedex 1, France
| | - Florence Caldefie-Chezet
- Human Nutrition Unit, ECREIN team, UMR 1019, University of Clermont Auvergne, INRAE, CRNH-Auvergne, TSA 50400, 28 place Henri Dunant, 63000 Clermont-Ferrand Cedex 1, France
| | - Marie-Paule Vasson
- Human Nutrition Unit, ECREIN team, UMR 1019, University of Clermont Auvergne, INRAE, CRNH-Auvergne, TSA 50400, 28 place Henri Dunant, 63000 Clermont-Ferrand Cedex 1, France.,Department of Nutrition, Gabriel Montpied University Hospital, Jean Perrin Cancer Centre, 58 rue Montalembert, 63011 Clermont-Ferrand, France
| | - Adrien Rossary
- Human Nutrition Unit, ECREIN team, UMR 1019, University of Clermont Auvergne, INRAE, CRNH-Auvergne, TSA 50400, 28 place Henri Dunant, 63000 Clermont-Ferrand Cedex 1, France
| |
Collapse
|
21
|
High-throughput screening suggests glutathione synthetase as an anti-tumor target of polydatin using human proteome chip. Int J Biol Macromol 2020; 161:1230-1239. [PMID: 32544581 DOI: 10.1016/j.ijbiomac.2020.06.061] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/26/2020] [Accepted: 06/07/2020] [Indexed: 12/23/2022]
Abstract
Polydatin (PD) is a bio-active ingredient with known anti-tumor effects. However, its specific protein targets yet have not been systematically screened, and the molecular anti-tumor mechanism is still unclear. Here, proteomic-chip was efficiently used to screen potential targets of PD. First, we investigated through animal experiment and proteomics studies, and found that polydatin play an important role in tumor cells. Then, the red-green fluorescent of polydatin was compared comprehensively to screen its targets on chip, followed by bioinformatics analysis. Glutathione synthetase (GSS) was selected as candidate research target. After a series of molecular biological experiments GSS was confirmed a target protein for PD in vitro. Moreover, we also found that PD can significantly inhibit the activity of GSS in vitro and live cells. Our findings reveal that PD could be a selective small-molecule GSS enzyme activity inhibitor and GSS could be a potential therapeutic target in cancer.
Collapse
|
22
|
Jin Y, Zhan X, Zhang B, Chen Y, Liu C, Yu L. Polydatin Exerts an Antitumor Effect Through Regulating the miR-382/PD-L1 Axis in Colorectal Cancer. Cancer Biother Radiopharm 2020; 35:83-91. [PMID: 31755739 DOI: 10.1089/cbr.2019.2999] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Ye Jin
- Department of General Surgery, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Xiaobo Zhan
- Department of General Surgery, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Bin Zhang
- Department of General Surgery, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Yun Chen
- Department of General Surgery, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Changfeng Liu
- Department of General Surgery, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Lingli Yu
- Department of Anesthesiology, The Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
23
|
Platella C, Raucci U, Rega N, D'Atri S, Levati L, Roviello GN, Fuggetta MP, Musumeci D, Montesarchio D. Shedding light on the interaction of polydatin and resveratrol with G-quadruplex and duplex DNA: a biophysical, computational and biological approach. Int J Biol Macromol 2019; 151:1163-1172. [PMID: 31747572 DOI: 10.1016/j.ijbiomac.2019.10.160] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/09/2019] [Accepted: 10/18/2019] [Indexed: 12/17/2022]
Abstract
Among polyphenols, trans-resveratrol (tRES) and trans-polydatin (tPD) exert multiple biological effects, particularly antioxidant and antiproliferative. In this work, we have investigated the interaction of tPD with three cancer-related DNA sequences able to form G-quadruplex (G4) structures, as well as with a model duplex, and compared its behaviour with tRES. Interestingly, fluorescence analysis evidenced the ability of tPD to bind all the studied DNA systems, similarly to tRES, with tRES displaying a higher ability to discriminate G4 over duplex with respect to tPD. However, neither tRES nor tPD produced significant conformational changes of the analyzed DNA upon binding, as determined by CD-titration analysis. Computational analysis and biological data confirmed the biophysical results: indeed, molecular docking evidenced the stronger interaction of tRES with the promoter of c-myc oncogene, and immunoblotting assays revealed a reduction of c-myc expression, more effective for tRES than tPD. Furthermore, in vitro assays on melanoma cells proved that tPD was able to significantly reduce telomerase activity, and inhibit cell proliferation, with tRES producing higher effects than tPD.
Collapse
Affiliation(s)
- Chiara Platella
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, Naples I-80126, Italy
| | - Umberto Raucci
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, Naples I-80126, Italy
| | - Nadia Rega
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, Naples I-80126, Italy; Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Piazzale Tecchio 80, Naples I-80125, Italy
| | - Stefania D'Atri
- Laboratory of Molecular Oncology, IDI-IRCCS, Via dei Monti di Creta 104, Rome I-00167, Italy
| | - Lauretta Levati
- Laboratory of Molecular Oncology, IDI-IRCCS, Via dei Monti di Creta 104, Rome I-00167, Italy
| | - Giovanni N Roviello
- Institute of Biostructures and Bioimages, CNR, Via Mezzocannone 16, Naples I-80134, Italy
| | - Maria Pia Fuggetta
- Institute of Translational Pharmacology, CNR, Via Fosso del Cavaliere 100, Rome I-00133, Italy.
| | - Domenica Musumeci
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, Naples I-80126, Italy; Institute of Biostructures and Bioimages, CNR, Via Mezzocannone 16, Naples I-80134, Italy.
| | - Daniela Montesarchio
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, Naples I-80126, Italy
| |
Collapse
|
24
|
Pan B, Shi X, Ding T, Liu L. Unraveling the action mechanism of polygonum cuspidatum by a network pharmacology approach. Am J Transl Res 2019; 11:6790-6811. [PMID: 31814888 PMCID: PMC6895524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 11/02/2019] [Indexed: 06/10/2023]
Abstract
As a popular Chinese herbal medicine (CHM), polygonum cuspidatum is widely used to treat various diseases in China. However, its biological function and action mechanism have yet to be systematically explored. In the present study, we first identified 14 potential active ingredients of polygonum cuspidatum using the TCMSP server and then conducted an in silico target prediction for these ingredients using PharmMapper. The subsequent KEGG pathway enrichment analysis of the 57 identified potential targets revealed that they were closely associated with cancer and gynecological disorders. Furthermore, a protein-protein interaction network of these targets was constructed using STRING and Cytoscape, through which 11 core targets were excavated according to degree, a key topological parameter. Meanwhile, we developed a novel formula, in which the "R value" is determined by average shortest path length and closeness centrality, two other key topological parameters, to evaluate the reliability of these predicted core targets. Intriguingly, among the top 10 core targets excavated using this new formula, 7 overlapped with the former 11 core targets, showing a good consistency in these core targets between the different prediction algorithms. Next, 7 ingredients were identified/validated from the crude extract of polygonum cuspidatum using UPLC-MS/MS. Noteworthy, 6 potential targets predicted for these 7 ingredients overlapped with the 7 core targets excavated from the previous in silico analyses. Further molecular docking and druggability analyses suggested that polydatin may play a pivotal role in manifesting the therapeutic effects of polygonum cuspidatum. Finally, we carried out a series of cell functional assays, which validated the anti-proliferative effects of polygonum cuspidatum on gynecological cancer cells, thus demonstrating our network pharmacology approach is reliable and powerful enough to guide the CHM mechanism study.
Collapse
Affiliation(s)
- Boyu Pan
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for CancerTianjin 300060, China
| | - Xiaona Shi
- Tianjin International Joint Academy of Biotechnology and Medicine Analytical Testing CenterTianjin 300457, China
| | - Tingting Ding
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for CancerTianjin 300060, China
| | - Liren Liu
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for CancerTianjin 300060, China
| |
Collapse
|
25
|
Vijayalakshmi S, Mariadoss AVA, Ramachandran V, Shalini V, Agilan B, Sangeetha CC, Balu P, Kotakadi VS, Karthikkumar V, Ernest D. Polydatin Encapsulated Poly [Lactic-co-glycolic acid] Nanoformulation Counteract the 7,12-Dimethylbenz[a] Anthracene Mediated Experimental Carcinogenesis through the Inhibition of Cell Proliferation. Antioxidants (Basel) 2019; 8:E375. [PMID: 31491872 PMCID: PMC6770361 DOI: 10.3390/antiox8090375] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/22/2019] [Accepted: 08/23/2019] [Indexed: 12/24/2022] Open
Abstract
In the present study, the authors have attempted to fabricate Polydatin encapsulated Poly [lactic-co-glycolic acid] (POL-PLGA-NPs) to counteract 7,12-dimethyl benzyl anthracene (DMBA) promoted buccal pouch carcinogenesis in experimental animals. The bio-formulated POL-PLGA-NPs were characterized by dynamic light scattering (DLS), Fourier transform infrared (FTIR) spectroscopy, X-ray powder diffraction (XRD) pattern analysis, and transmission electron microscope (TEM). In addition, the nano-chemopreventive potential of POL-PLGA-NPs was assessed by scrutinizing the neoplastic incidence and analyzing the status of lipid peroxidation, antioxidants, phase I, phase II detoxification status, and histopathological changes and in DMBA-treated animals. In golden Syrian hamsters, oral squamous cell carcinoma (OSCC) was generated by painting with 0.5% DMBA in liquid paraffin three times a week for 14 weeks. After 100% tumor formation was observed, high tumor volume, tumor burden, and altered levels of biochemical status were observed in the DMBA-painted hamsters. Intra-gastric administration of varying concentration of POL-PLGA-NPs (7.5, 15, and 30 mg/kg b.wt) to DMBA-treated hamsters assumedly prevents oncological incidences and restores the status of the biochemical markers. It also significantly enhances the apoptotic associated and inhibits the cancer cell proliferative markers expression (p53, Bax, Bcl-2, cleaved caspase 3, cyclin-D1). The present study reveals that POL-PLGA-NPs is a penitential candidate for nano-chemopreventive, anti-lipid peroxidative, and antioxidant potential, and also has a modulating effect on the phase I and Phase II detoxification system, which is associated with reduced cell proliferation and induced apoptosis in experimental oral carcinogenesis.
Collapse
Affiliation(s)
- Sankaran Vijayalakshmi
- Department of Biotechnology, Thiruvalluvar University, Serkadu, Vellore 632 115, Tamilnadu, India
| | | | - Vinayagam Ramachandran
- Department of Biotechnology, Thiruvalluvar University, Serkadu, Vellore 632 115, Tamilnadu, India
| | - Vijayakumar Shalini
- Department of Biotechnology, Thiruvalluvar University, Serkadu, Vellore 632 115, Tamilnadu, India
| | - Balupillai Agilan
- Department of Biotechnology, Thiruvalluvar University, Serkadu, Vellore 632 115, Tamilnadu, India
| | - Casimeer C Sangeetha
- Department of Physics, Sri Padmavati Mahila Visvavidyalayam, Tirupati 517502, Andra Pradesh, India
| | - Periyasamy Balu
- Department of Chemistry, Thiruvalluvar University, Serkadu, Vellore 632 115, Tamilnadu, India
| | | | - Venkatachalam Karthikkumar
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, UAE University, Al Ain 17666, UAE.
| | - David Ernest
- Department of Biotechnology, Thiruvalluvar University, Serkadu, Vellore 632 115, Tamilnadu, India.
| |
Collapse
|
26
|
Zhang T, Zhu X, Wu H, Jiang K, Zhao G, Shaukat A, Deng G, Qiu C. Targeting the ROS/PI3K/AKT/HIF-1α/HK2 axis of breast cancer cells: Combined administration of Polydatin and 2-Deoxy-d-glucose. J Cell Mol Med 2019; 23:3711-3723. [PMID: 30920152 PMCID: PMC6484306 DOI: 10.1111/jcmm.14276] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 11/19/2018] [Accepted: 02/25/2019] [Indexed: 12/28/2022] Open
Abstract
It is well established that cancer cells depend upon aerobic glycolysis to provide the energy they need to survive and proliferate. However, anti-glycolytic agents have yielded few positive results in human patients, in part due to dose-limiting side effects. Here, we discovered the unexpected anti-cancer efficacy of Polydatin (PD) combined with 2-deoxy-D-glucose (2-DG), which is a compound that inhibits glycolysis. We demonstrated in two breast cell lines (MCF-7 and 4T1) that combination treatment with PD and 2-DG induced cell apoptosis and inhibited cell proliferation, migration and invasion. Furthermore, we determined the mechanism of PD in synergy with 2-DG, which decreased the intracellular reactive oxygen (ROS) levels and suppressed the PI3K/AKT pathway. In addition, the combined treatment inhibited the glycolytic phenotype through reducing the expression of HK2. HK2 deletion in breast cancer cells thus improved the anti-cancer activity of 2-DG. The combination treatment also resulted in significant tumour regression in the absence of significant morphologic changes in the heart, liver or kidney in vivo. In summary, our study demonstrates that PD synergised with 2-DG to enhance its anti-cancer efficacy by inhibiting the ROS/PI3K/AKT/HIF-1α/HK2 signalling axis, providing a potential anti-cancer strategy.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Huazhong Agricultural UniversityWuhanPeople's Republic of China
| | - Xinying Zhu
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Huazhong Agricultural UniversityWuhanPeople's Republic of China
| | - Haichong Wu
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Huazhong Agricultural UniversityWuhanPeople's Republic of China
| | - Kangfeng Jiang
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Huazhong Agricultural UniversityWuhanPeople's Republic of China
| | - Gan Zhao
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Huazhong Agricultural UniversityWuhanPeople's Republic of China
| | - Aftab Shaukat
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Huazhong Agricultural UniversityWuhanPeople's Republic of China
| | - Ganzhen Deng
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Huazhong Agricultural UniversityWuhanPeople's Republic of China
| | - Changwei Qiu
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Huazhong Agricultural UniversityWuhanPeople's Republic of China
| |
Collapse
|
27
|
Fuggetta MP, Migliorino MR, Ricciardi S, Osman G, Iacono D, Leone A, Lombardi A, Ravagnan G, Greco S, Remotti D, Romano MCP. Prophylactic Dermatologic Treatment of Afatinib-Induced Skin Toxicities in Patients with Metastatic Lung Cancer: A Pilot Study. SCIENTIFICA 2019; 2019:9136249. [PMID: 30838155 PMCID: PMC6374781 DOI: 10.1155/2019/9136249] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 10/02/2018] [Accepted: 12/16/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Severe skin rash is listed among important side effects of EGFR tyrosine kinase inhibitors. Polydatin (PD), a glycosylated polyphenol, is endowed with anti-inflammatory activity in human epidermal keratinocytes. OBJECTIVE This study evaluated the effect of topical application of a moisturizer containing PD to prevent skin rash in patients with mutated non-small cell lung cancer (NSCLC) treated with afatinib. MATERIALS AND METHODS Eligible NSCLC patients with metastatic disease were treated with first-line afatinib 40 mg/die. One day before starting systemic therapy, all patients received topical administration of a 1.5% PD-based cream b.i.d. every day until the end of afatinib treatment. RESULTS Out of 34 treated patients, the incidence of skin rash (all grades) was 41.2% and grade 2 rash was 20.6%, and grade 3 rash was not observed in any of the patients. None of the patients discontinued therapy for toxicity. The mean duration of treatment was 6.4 months, calculated from the time treatment was started to the date treatment was stopped. CONCLUSION The results showed that a PD-based cream can reduce the incidence of grade ≥2 skin toxicities in patients treated with afatinib. Clinical study registration number: Prot. No. 130/CE Lazio 1 Italy.
Collapse
Affiliation(s)
| | | | - Serena Ricciardi
- UOSD of Oncologic Pneumology, San Camillo Forlanini Hospital, Rome, Italy
| | - Giorgia Osman
- UOSD of Oncologic Pneumology, San Camillo Forlanini Hospital, Rome, Italy
| | - Daniela Iacono
- UOSD of Oncologic Pneumology, San Camillo Forlanini Hospital, Rome, Italy
| | - Alvaro Leone
- UOC of Anatomopathology, San Camillo Forlanini Hospital, Rome, Italy
| | | | | | - Stefania Greco
- UOSD of Oncologic Pneumology, San Camillo Forlanini Hospital, Rome, Italy
| | - Daniele Remotti
- UOC of Anatomopathology, San Camillo Forlanini Hospital, Rome, Italy
| | | |
Collapse
|
28
|
Ma Y, Jia X. Polydatin Alleviates Radiation-Induced Testes Injury by Scavenging ROS and Inhibiting Apoptosis Pathways. Med Sci Monit 2018; 24:8993-9000. [PMID: 30539937 PMCID: PMC6299782 DOI: 10.12659/msm.913725] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background Exposure to ionizing radiation (IR) induces severe damage in multiple human tissues. The testes are extremely sensitive to IR, and testes irradiation can result in infertility and abnormality. A novel and safe radioprotector for testes injury from IR is needed. Polydatin (PD) has been proved to have anti-oxidant and anti-inflammatory effects, indicating its potential application in radiation protection. Material/Methods Male wild-type C57BL/6 mice (8 weeks old) were exposed to ionizing radiation. At different times after irradiation, testes were isolated and subjected to hematoxylin-eosin (HE) staining and TUNEL staining, as well as related quantification. ELISA assay was used to measure the level of inflammatory cytokines, and apoptosis proteins were detected by Western blot assay. Intracellular ROS was measured by DCFH-DA flow cytometry method. Results In the present study, we demonstrated that polydatin effectively alleviated testes injury and retained sperm viability. PD pretreatment also inhibited cell apoptosis caused by irradiation. Radiation-induced decrease of FSH and testosterone was also inhibited by PD treatment. Finally, we showed that PD obviously reduced the ROS level, using DCFH-DA method. We also found that PD reduced the concentration of the oxidative products MDA and 8-OHdG. PD also inhibited apoptosis-related proteins such as Bax and caspase 3. Conclusions Our data proved that polydatin effectively alleviated testes injury after irradiation, mainly through reducing ROS and oxidative stress. Our findings suggest polydatin as a potential radioprotector for testes radiation damage.
Collapse
Affiliation(s)
- Yan Ma
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Xiaojing Jia
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin, China (mainland)
| |
Collapse
|
29
|
Antitumor Research of the Active Ingredients from Traditional Chinese Medical Plant Polygonum Cuspidatum. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:2313021. [PMID: 30584449 PMCID: PMC6280292 DOI: 10.1155/2018/2313021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/05/2018] [Accepted: 10/24/2018] [Indexed: 02/06/2023]
Abstract
In recent years, the utilization of Chinese native medicine and other plant extracts in the treatment of diseases has attracted extensive attention, especially in the area of malignant tumors. However, lots of herbal remedies active ingredients have not been found or have been discovered but not effectively developed and applied. Therefore, screening new Chinese medicine active components and determining their antitumor effects have become a new breakthrough in the prevention and treatment of tumor disease. In the past years, a large number of studies have demonstrated that Polygonum cuspidatum and its active components like resveratrol showed excellent antitumor activities, including our own antitumor studies about resveratrol in colorectal cancer. The purpose of this review is to summarize the research progress of Chinese herb Polygonum cuspidatum and its active components in tumor diseases and provide theoretical basis for further scientific experiments and clinical applications.
Collapse
|
30
|
Jin YL, Xin LM, Zhou CC, Ren Y. Polydatin exerts anti-tumor effects against renal cell carcinoma cells via induction of caspase-dependent apoptosis and inhibition of the PI3K/Akt pathway. Onco Targets Ther 2018; 11:8185-8195. [PMID: 30532558 PMCID: PMC6247951 DOI: 10.2147/ott.s180785] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Purpose Polydatin, a stilbenoid glucoside of a resveratrol derivative, has many biological functions, including antitumor effects. However, the antitumor effects of polydatin in renal cell carcinoma (RCC) have not been investigated. Materials and methods In the current study, MTT assays, transwell invasion assays and wound healing assays were performed to examine cell proliferation, invasion and migration. An apoptosis nucleosome ELISA was used to measure apoptosis. Caspase activity assays were applied to measure the activities of caspase-3/9. A Western blot assay was used to measure the change in protein levels. Results Our data demonstrated that polydatin inhibited the proliferation of RCC cells but not normal renal epithelial cells in a time- and dose-dependent manner. Polydatin also triggered apoptosis in a caspase-dependent manner. Moreover, polydatin treatment also led to the downregulation of Bcl-2 and Mcl-1 and to activation of Bax. Ectopic expression of Bcl-2 and Mcl-1 or silencing of Bax could repress the apoptosis that was induced by polydatin. Moreover, incubation with polydatin also suppressed the PI3K/Akt signaling pathway in RCC cells. Conclusion Taken together, our data indicated that polydatin may be applied as a potent agent against RCC.
Collapse
Affiliation(s)
- Yi-Li Jin
- Department of Urology, Dongyang People's Hospital, Wenzhou Medical University, Dongyang, Zhejiang 322100, People's Republic of China
| | - Li-Min Xin
- Laboratory of kidney Carcinoma, Ningbo Urology and Nephrology Hospital, Urology and Nephrology Institute of Ningbo University, Ningbo 315000, Zhejiang Province, People's Republic of China,
| | - Chang-Chun Zhou
- Department of Urology, Dongyang People's Hospital, Wenzhou Medical University, Dongyang, Zhejiang 322100, People's Republic of China
| | - Yu Ren
- Laboratory of kidney Carcinoma, Ningbo Urology and Nephrology Hospital, Urology and Nephrology Institute of Ningbo University, Ningbo 315000, Zhejiang Province, People's Republic of China,
| |
Collapse
|
31
|
Polydatin protects against acute myocardial infarction-induced cardiac damage by activation of Nrf2/HO-1 signaling. J Nat Med 2018; 73:85-92. [DOI: 10.1007/s11418-018-1241-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 08/22/2018] [Indexed: 12/31/2022]
|
32
|
Pan B, Ren Y, Liu L. Uncovering the action mechanism of polydatin via network pharmacological target prediction. RSC Adv 2018; 8:18851-18858. [PMID: 35539671 PMCID: PMC9080635 DOI: 10.1039/c8ra03124j] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 05/14/2018] [Indexed: 12/19/2022] Open
Abstract
Polydatin (PD), a small natural compound originally extracted from Polygonum cuspidatum, exerts distinct biological functions in a variety of diseases. However, the action mechanism of PD has yet to be systematically explored. In this study, we firstly corroborated the druggability of PD by evaluating the medicinal properties of PD using a TCMSP server. We then conducted in silico target-prediction for PD using PharmMapper and ChemMapper, which led to the identification of 15 potential targets overlapping in both approaches. These 15 targets were subsequently evaluated by GeneMANIA, GO biological process and KEGG pathway analysis, which finally contribute to the construction of a drug-target-pathway network for PD. The network analysis revealed that these targets were mainly associated with cancer, cell growth and apoptosis, hormones and other physiological processes, outlining the pharmacological influences of PD on multiple integrated pathways involved in a particular network.
Collapse
Affiliation(s)
- Boyu Pan
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute & Hospital Tianjin 300060 China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer Tianjin 300060 China
| | - Yuanyuan Ren
- Department of Pharmacy, Tianjin Medical University Cancer Institute & Hospital Tianjin 300060 China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer Tianjin 300060 China
| | - Liren Liu
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute & Hospital Tianjin 300060 China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer Tianjin 300060 China
| |
Collapse
|