1
|
Kaur G, Devi S, Sharma A, Sood P. Pharmacological insights and role of bufalin (bufadienolides) in inflammation modulation: a narrative review. Inflammopharmacology 2024:10.1007/s10787-024-01517-9. [PMID: 39012431 DOI: 10.1007/s10787-024-01517-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 06/24/2024] [Indexed: 07/17/2024]
Abstract
Bufadienolides, specifically bufalin, have garnered attention for their potential therapeutic application in modulating inflammatory pathways. Bufalin is derived from toad venom and exhibits promising anti-inflammatory properties. Its anti-inflammatory effects have been demonstrated by influencing crucial signaling pathways like NF-B, MAPK, and JAK-STAT, resulting in the inhibition of pro-inflammatory substances like cytokines, chemokines, and adhesion molecules. Bufalin blocks inflammasome activation and reduces oxidative stress, hence increasing its anti-inflammatory properties. Bufalin has shown effectiveness in reducing inflammation-related diseases such as cancer, cardiovascular problems, and autoimmune ailments in preclinical investigations. Furthermore, producing new approaches of medication delivery and combining therapies with bufalin shows potential for improving its effectiveness and reducing adverse effects. This review explores the pharmacological effects and mechanistic approaches of bufalin as an anti-inflammatory agent, which further highlights its potential for therapy and offers the basis for further study on its therapeutic application in inflammation-related disorders.
Collapse
Affiliation(s)
- Gagandeep Kaur
- Chitkara University School of Pharmacy, Chitkara University, Baddi, Himachal Pradesh, India
| | - Sushma Devi
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Akhil Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Parul Sood
- Chitkara University School of Pharmacy, Chitkara University, Baddi, Himachal Pradesh, India
| |
Collapse
|
2
|
Miao L, Liu Y, Ali NM, Dong Y, Zhang B, Cui X. Bufalin serves as a pharmaceutic that mitigates drug resistance. Drug Metab Rev 2023:1-10. [PMID: 37114332 DOI: 10.1080/03602532.2023.2206065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Intrinsic or acquired drug resistance of tumor cells is the main cause of tumor chemotherapy failure and tumor-related death. Bufalin (BF) is the main active monomer component extracted from the Traditional Chinese Medicine Toad venom (secretions of glands behind the ears and epidermis of bufo gargarizans and Bufo Melanostictus Schneider). It is a cardiotonic steroid with broad-spectrum anti-cancer effects and has been widely used against various malignant tumors in clinical practice. Pharmacological studies also found that BF has the effect of reversing drug resistance, which provides a new perspective for the application of Traditional Chinese Medicine as a chemosensitizer in cancer therapy. This article provides an extensive search and summary of published research on mitigating drug resistance to BF and reviews its potential mechanisms.
Collapse
Affiliation(s)
- Linxuan Miao
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, P.R. China
| | - Ying Liu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, P.R. China
- Department of Oncology, Affiliated Zhongshan Hospital of Dalian University, Dalian, P.R. China
| | - Nasra Mohamoud Ali
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, P.R. China
| | - Yan Dong
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, P.R. China
| | - Bin Zhang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, P.R. China
| | - Xiaonan Cui
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, P.R. China
| |
Collapse
|
3
|
Shao H, Li B, Li H, Gao L, Zhang C, Sheng H, Zhu L. Novel Strategies for Solubility and Bioavailability Enhancement of Bufadienolides. Molecules 2021; 27:51. [PMID: 35011278 PMCID: PMC8746454 DOI: 10.3390/molecules27010051] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 02/07/2023] Open
Abstract
Toad venom contains a large number of bufadienolides, which have a variety of pharmacological activities, including antitumor, cardiovascular, anti-inflammatory, analgesic and immunomodulatory effects. The strong antitumor effect of bufadienolides has attracted considerable attention in recent years, but the clinical application of bufadienolides is limited due to their low solubility and poor bioavailability. In order to overcome these shortcomings, many strategies have been explored, such as structural modification, solid dispersion, cyclodextrin inclusion, microemulsion and nanodrug delivery systems, etc. In this review, we have tried to summarize the pharmacological activities and structure-activity relationship of bufadienolides. Furthermore, the strategies for solubility and bioavailability enhancement of bufadienolides also are discussed. This review can provide a basis for further study on bufadienolides.
Collapse
Affiliation(s)
| | | | | | | | | | - Huagang Sheng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road, Jinan 250355, China; (H.S.); (B.L.); (H.L.); (L.G.); (C.Z.)
| | - Liqiao Zhu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road, Jinan 250355, China; (H.S.); (B.L.); (H.L.); (L.G.); (C.Z.)
| |
Collapse
|
4
|
Zhang Y, Yuan B, Bian B, Zhao H, Kiyomi A, Hayashi H, Iwatani Y, Sugiura M, Takagi N. Cytotoxic Effects of Hellebrigenin and Arenobufagin Against Human Breast Cancer Cells. Front Oncol 2021; 11:711220. [PMID: 34513690 PMCID: PMC8427765 DOI: 10.3389/fonc.2021.711220] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022] Open
Abstract
Development of new therapeutic strategies for breast cancer is urgently needed due to the sustained emergence of drug resistance, tumor recurrence and metastasis. To gain a novel insight into therapeutic approaches to fight against breast cancer, the cytocidal effects of hellebrigenin (Helle) and arenobufagin (Areno) were investigated in human estrogen receptor (ER)-positive breast cancer cell line MCF-7 and triple-negative breast cancer cell line MDA-MB-231. Helle exhibited more potent cytotoxicity than Areno in both cancer cells, and MCF-7 cells were more susceptible to both drugs in comparison with MDA-MB-231 cells. Apoptotic-like morphological characteristics, along with the downregulation of the expression level of Bcl-2 and Bcl-xL and the upregulation of the expression level of Bad, were observed in Helle-treated MCF-7 cells. Helle also caused the activation of caspase-8, caspase-9, along with the cleavage of poly(ADP-ribose) polymerase in MCF-7 cells. Helle-mediated necrosis-like phenotype, as evidenced by the increased propidium iodide (PI)-positive cells was further observed. G2/M cell cycle arrest was also induced by Helle in the cells. Upregulation of the expression level of p21 and downregulation of the expression level of cyclin D1, cyclin E1, cdc25C and survivin were observed in MCF-7 cells treated with Helle and occurred in parallel with G2/M arrest. Autophagy was triggered in MCF-7 cells and the addition of wortmannin or 3-MA, two well-known autophagy inhibitors, slightly but significantly rescued the cells. Furthermore, similar alterations of some key molecules associated with the aforementioned biological phenomena were observed in MDA-MB-231 cells. Intriguingly, the numbers of PI-positive cells in Helle-treated MCF-7 cells were significantly reduced by wortmannin and 3-MA, respectively. In addition, Helle-triggered G2/M arrest was significantly corrected by wortmannin, suggesting autophagy induction contributed to Helle-induced cytotoxicity of breast cancer cells by modulating necrosis and cell cycle arrest. Collectively, our results suggested potential usefulness of both Helle and Areno in developing therapeutic strategies to treat patients with different types of breast cancer, especially ER-positive breast cancer.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Applied Biochemistry, Tokyo University of Pharmacy & Life Sciences, Hachioji, Japan.,Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bo Yuan
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Pharmaceutical Sciences, Josai University, Sakado, Japan
| | - Baolin Bian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haiyu Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Anna Kiyomi
- Department of Drug Safety and Risk Management, Tokyo University of Pharmacy & Life Sciences, Hachioji, Japan
| | - Hideki Hayashi
- Department of Applied Biochemistry, Tokyo University of Pharmacy & Life Sciences, Hachioji, Japan
| | - Yui Iwatani
- Department of Applied Biochemistry, Tokyo University of Pharmacy & Life Sciences, Hachioji, Japan
| | - Munetoshi Sugiura
- Department of Drug Safety and Risk Management, Tokyo University of Pharmacy & Life Sciences, Hachioji, Japan
| | - Norio Takagi
- Department of Applied Biochemistry, Tokyo University of Pharmacy & Life Sciences, Hachioji, Japan
| |
Collapse
|
5
|
Gong M, Wang X, Mu L, Wang Y, Pan J, Yuan X, Zhou H, Xing J, Wang R, Sun J, Liu Q, Zhang X, Wang L, Chen Y, Pei Y, Li S, Liu L, Zhao Y, Yuan Y. Steroid receptor coactivator-1 enhances the stemness of glioblastoma by activating long noncoding RNA XIST/miR-152/KLF4 pathway. Cancer Sci 2021; 112:604-618. [PMID: 33090636 PMCID: PMC7894023 DOI: 10.1111/cas.14685] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 09/25/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) recurrence is attributed to the presence of therapy-resistant glioblastoma stem cells. Steroid receptor coactivator-1 (SRC-1) acts as an oncogenic regulator in many human tumors. The relationship between SRC-1 and GBM has not yet been studied. Herein, we investigate the role of SRC-1 in GBM. In this study, we found that SRC-1 expression is positively correlated with grades of glioma and inversely correlated with glioma patient's prognosis. Steroid receptor coactivator-1 promotes the proliferation, migration, and tumor growth of GBM cells. Notably, SRC-1 knockdown suppresses the stemness of GBM cells. Mechanistically, long noncoding RNA X-inactive specific transcript (XIST) is regulated by SRC-1 at the posttranscriptional level and mediates the function of SRC-1 in promoting stemness-like properties of GBM. Steroid receptor coactivator-1 can promote the expression of Kruppel-like factor 4 (KLF4) through the XIST/microRNA (miR)-152 axis. Additionally, arenobufagin and bufalin, SRC small molecule inhibitors, can reduce the proliferation and stemness of GBM cells. This study reveals SRC-1 promotes the stemness of GBM by activating the long noncoding RNA XIST/miR-152/KLF4 pathway and provides novel markers for diagnosis and therapy of GBM.
Collapse
Affiliation(s)
- Miaomiao Gong
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Xun Wang
- Department of NeurosurgeryThe Third People’s Hospital of DalianDalianChina
| | - Lin Mu
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Yueyue Wang
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Jinjin Pan
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Xiaocheng Yuan
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Haoran Zhou
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Jinshan Xing
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Rui Wang
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Jian Sun
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Qiwang Liu
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Xiya Zhang
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Lin Wang
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Yiying Chen
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Yandong Pei
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Shao Li
- College of Basic Medical SciencesDalian Medical UniversityDalianChina
| | - Liang Liu
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Yongshun Zhao
- The First Affiliated HospitalDalian Medical UniversityDalianChina
| | - Yuhui Yuan
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| |
Collapse
|
6
|
Chen B, Wang C, Ma J, Ma H, Wang Y, Zhang H, Zhu Y, Yao J, Luo C, Miao Z, Wu Y. Discovery of 3-peptide substituted arenobufagin derivatives as potent antitumor agents with low cardiotoxicity. Steroids 2021; 166:108772. [PMID: 33271132 DOI: 10.1016/j.steroids.2020.108772] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 09/17/2020] [Accepted: 11/09/2020] [Indexed: 11/21/2022]
Abstract
Active natural productscan be valuable lead compounds and numerous drugs derived from natural products have successfully entered the clinic. Arenobufagin, one of the important active components of toad venom, indicates significant antitumor activities with limited preclinical development for its strong cardiotoxicity. Ten 3-monopeptide substituted arenobufagin derivatives have been designed and synthesized. Antitumor activity and cardiotoxicity assays lead to the discovery of compound ZM226 as a potent antitumor agent with low cardiotoxicity. These findings suggest optimization of arenobufagin on position 3 maybe an efficacious strategy for the development of antitumor drug candidates derived from arenobufagin.
Collapse
Affiliation(s)
- Baobao Chen
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, People's Republic of China
| | - Chuanhao Wang
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, People's Republic of China
| | - Jianjiang Ma
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, People's Republic of China
| | - Haijun Ma
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750004, People's Republic of China
| | - Yuan Wang
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750004, People's Republic of China
| | - Hui Zhang
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, People's Republic of China
| | - Yazhao Zhu
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, People's Republic of China
| | - Jianzhong Yao
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, People's Republic of China
| | - Chuan Luo
- Anhui China Resources Jinchan Pharmaceutical Co., Ltd., 39 Longfa Road, Huaibei, Anhui 235000, People's Repubilic of China.
| | - Zhenyuan Miao
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, People's Republic of China.
| | - Yuelin Wu
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, People's Republic of China.
| |
Collapse
|
7
|
Deng LJ, Li Y, Qi M, Liu JS, Wang S, Hu LJ, Lei YH, Jiang RW, Chen WM, Qi Q, Tian HY, Han WL, Wu BJ, Chen JX, Ye WC, Zhang DM. Molecular mechanisms of bufadienolides and their novel strategies for cancer treatment. Eur J Pharmacol 2020; 887:173379. [PMID: 32758567 DOI: 10.1016/j.ejphar.2020.173379] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/05/2020] [Accepted: 07/13/2020] [Indexed: 12/16/2022]
Abstract
Bufadienolides are cardioactive C24 steroids with an α-pyrone ring at position C17. In the last ten years, accumulating studies have revealed the anticancer activities of bufadienolides and their underlying mechanisms, such as induction of autophagy and apoptosis, cell cycle disruption, inhibition of angiogenesis, epithelial-mesenchymal transition (EMT) and stemness, and multidrug resistance reversal. As Na+/K+-ATPase inhibitors, bufadienolides have inevitable cardiotoxicity. Short half-lives, poor stability, low plasma concentration and oral bioavailability in vivo are obstacles for their applications as drugs. To improve the drug potency of bufadienolides and reduce their side effects, prodrug strategies and drug delivery systems such as liposomes and nanoparticles have been applied. Therefore, systematic and recapitulated information about the antitumor activity of bufadienolides, with special emphasis on the molecular or cellular mechanisms, prodrug strategies and drug delivery systems, is of high interest. Here, we systematically review the anticancer effects of bufadienolides and the molecular or cellular mechanisms of action. Research advancements regarding bufadienolide prodrugs and their tumor-targeting delivery strategies are critically summarized. This work highlights recent scientific advances regarding bufadienolides as effective anticancer agents from 2011 to 2019, which will help researchers to understand the molecular pathways involving bufadienolides, resulting in a selective and safe new lead compound or therapeutic strategy with improved therapeutic applications of bufadienolides for cancer therapy.
Collapse
Affiliation(s)
- Li-Juan Deng
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China; School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Yong Li
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Ming Qi
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Jun-Shan Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Sheng Wang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Li-Jun Hu
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Yu-He Lei
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, 518034, China
| | - Ren-Wang Jiang
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Wei-Min Chen
- College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Qi Qi
- Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, PR China
| | - Hai-Yan Tian
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Wei-Li Han
- School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Bao-Jian Wu
- College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Jia-Xu Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Wen-Cai Ye
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China.
| | - Dong-Mei Zhang
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
8
|
Deng LJ, Qi M, Peng QL, Chen MF, Qi Q, Zhang JY, Yao N, Huang MH, Li XB, Peng YH, Liu JS, Fu DR, Chen JX, Ye WC, Zhang DM. Arenobufagin induces MCF-7 cell apoptosis by promoting JNK-mediated multisite phosphorylation of Yes-associated protein. Cancer Cell Int 2018; 18:209. [PMID: 30574018 PMCID: PMC6299615 DOI: 10.1186/s12935-018-0706-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 12/11/2018] [Indexed: 12/24/2022] Open
Abstract
Background It has been demonstrated that bufadienolides exert potent anti-cancer activity in various tumor types. However, the mechanisms that underlie their anti-cancer properties remain unclear. Yes-associated protein, a key effector of Hippo signaling, functions as a transcription coactivator, plays oncogenic and tumor suppressor roles under different conditions. Here, we report that arenobufagin (ABF), a representative bufadienolide, induced breast cancer MCF-7 cells to undergo apoptosis, which occurred through the JNK-mediated multisite phosphorylation of YAP. Methods Cytotoxicity was examined using an MTT assay. ABF-induced apoptosis was measured with a TUNEL assay and Annexin V-FITC/PI double staining assay. Western blotting, immunofluorescence, qRT-PCR and coimmunoprecipitation were employed to assess the expression levels of the indicated molecules. Lose-of-function experiments were carried out with siRNA transfection and pharmacological inhibitors. ABF-induced phosphopeptides were enriched with Ti4+-IMAC chromatography and further subjected to reverse-phase nano-LC–MS/MS analysis. Results ABF significantly reduced the viability of MCF-7 cells and increased the percentage of early and late apoptotic cells in a concentration- and time-dependent manner. Following ABF treatment, YAP accumulated in the nucleus and bound to p73, which enhanced the transcription of the pro-apoptotic genes Bax and p53AIP1. YAP knock-down significantly attenuated ABF-induced apoptotic cell death. Importantly, we found that the mobility shift of YAP was derived from its phosphorylation at multiple sites, including Tyr357. Moreover, mass spectrometry analysis identified 19 potential phosphorylation sites in YAP, with a distribution of 14 phosphoserine and 5 phosphothreonine residues. Furthermore, we found that the JNK inhibitor SP600125 completely diminished the mobility shift of YAP and its phosphorylation at Tyr357, the binding of YAP and p73, the transcription of Bax and p53AIP1 as well as the apoptosis induced by ABF. These data indicate that ABF induced YAP multisite phosphorylation, which was associated with p73 binding, and that apoptosis was mediated by the JNK signaling pathway. Conclusions Our data demonstrate that ABF suppresses MCF-7 breast cancer proliferation by triggering the pro-apoptotic activity of YAP, which is mediated by JNK signaling-induced YAP multisite phosphorylation as well as its association with p73. The present work not only provides additional information on the use of ABF as an anti-breast cancer drug, but also offers evidence that the induction of the tumor suppressor role of YAP may be a therapeutic strategy. Electronic supplementary material The online version of this article (10.1186/s12935-018-0706-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Li-Juan Deng
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,2Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Ming Qi
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Qun-Long Peng
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Min-Feng Chen
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Qi Qi
- 4Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Jia-Yan Zhang
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Nan Yao
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Mao-Hua Huang
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Xiao-Bo Li
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Yin-Hui Peng
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Jun-Shan Liu
- 5School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515 People's Republic of China
| | - Deng-Rui Fu
- Guangzhou Yucai Middle School, Fujin Road 2#, Dongshan District, Guangzhou, China
| | - Jia-Xu Chen
- 2Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Wen-Cai Ye
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Dong-Mei Zhang
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| |
Collapse
|
9
|
Sun P, Feng LX, Zhang DM, Liu M, Liu W, Mi T, Wu WY, Jiang BH, Yang M, Hu LH, Guo DA, Liu X. Bufalin derivative BF211 inhibits proteasome activity in human lung cancer cells in vitro by inhibiting β1 subunit expression and disrupting proteasome assembly. Acta Pharmacol Sin 2016; 37:908-18. [PMID: 27238210 DOI: 10.1038/aps.2016.30] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/28/2016] [Indexed: 12/20/2022] Open
Abstract
AIM Bufalin is one of the active components in the traditional Chinese medicine ChanSu that is used to treat arrhythmia, inflammation and cancer. BF211 is a bufalin derivative with stronger cytotoxic activity in cancer cells. The aim of this study was to identify the putative target proteins of BF211 and the signaling pathways in cancer cells. METHODS A549 human lung cancer cells were treated with BF211. A SILAC-based proteomic analysis was used to detect the protein expression profiles of BF211-treated A549 cells. Cellular proteasome activities were examined using fluorogenic peptide substrates, and the binding affinities of BF211 to recombinant proteasome subunit proteins were evaluated using the Biacore assay. The expression levels of proteasome subunits were determined using RT-PCR and Western blotting, and the levels of the integral 26S proteasome were evaluated using native PAGE analysis. RESULTS The proteomic analysis revealed that 1282 proteins were differentially expressed in BF211-treated A549 cells, and the putative target proteins of BF211 were associated with various cellular functions, including transcription, translation, mRNA splicing, ribosomal protein synthesis and proteasome function. In A549 cells, BF211 (5, 10, and 20 nmol/L) dose-dependently inhibited the enzymatic activities of proteasome. But BF211 displayed a moderate affinity in binding to proteasome β1 subunit and no binding affinity to the β2 and β5 subunits. Moreover, BF211 (0.1, 1, and 10 nmol/L) did not inhibit the proteasome activities in the cell lysates. BF211 (5, 10, and 20 nmol/L) significantly decreased the expression level of proteasome β1 subunit and the levels of integral 26S proteasome in A549 cells. Similarly, knockdown of the β1 subunit with siRNA in A549 cells significantly decreased integral 26S proteasome and proteasome activity. CONCLUSION BF211 inhibits proteasome activity in A549 cells by decreasing β1 subunit expression and disrupting proteasome assembly.
Collapse
|