1
|
Zhou ZY, Ma J, Zhao WR, Shi WT, Zhang J, Hu YY, Yue MY, Zhou WL, Yan H, Tang JY, Wang Y. Qiangxinyin formula protects against isoproterenol-induced cardiac hypertrophy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155717. [PMID: 38810550 DOI: 10.1016/j.phymed.2024.155717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/08/2024] [Accepted: 05/05/2024] [Indexed: 05/31/2024]
Abstract
Heart failure is a life-threatening cardiovascular disease and characterized by cardiac hypertrophy, inflammation and fibrosis. The traditional Chinese medicine formula Qiangxinyin (QXY) is effective for the treatment of heart failure while the underlying mechanism is not clear. This study aims to identify the active ingredients of QXY and explore its mechanisms protecting against cardiac hypertrophy. We found that QXY significantly protected against isoproterenol (ISO)-induced cardiac hypertrophy and dysfunction in zebrafish. Eight compounds, including benzoylmesaconine (BMA), atractylenolide I (ATL I), icariin (ICA), quercitrin (QUE), psoralen (PRN), kaempferol (KMP), ferulic acid (FA) and protocatechuic acid (PCA) were identified from QXY. PRN, KMP and icaritin (ICT), an active pharmaceutical ingredient of ICA, prevented ISO-induced cardiac hypertrophy and dysfunction in zebrafish. In H9c2 cardiomyocyte treated with ISO, QXY significantly blocked the calcium influx, reduced intracellular lipid peroxidative product MDA, stimulated ATP production and increased mitochondrial membrane potential. QXY also inhibited ISO-induced cardiomyocyte hypertrophy and cytoskeleton reorganization. Mechanistically, QXY enhanced the phosphorylation of Smad family member 2 (SMAD2) and myosin phosphatase target subunit-1 (MYPT1), and suppressed the phosphorylation of myosin light chain (MLC). In conclusion, PRN, KMP and ICA are the main active ingredients of QXY that protect against ISO-induced cardiac hypertrophy and dysfunction largely via the blockage of calcium influx and inhibition of mitochondrial dysfunction as well as cytoskeleton reorganization.
Collapse
Affiliation(s)
- Zhong-Yan Zhou
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong Special Administrative Regions of China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administrative Regions of China
| | - Jie Ma
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; School of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Wai-Rong Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wen-Ting Shi
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Zhang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan-Yan Hu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mei-Yan Yue
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wen-Long Zhou
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Yan
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing-Yi Tang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yu Wang
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong Special Administrative Regions of China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administrative Regions of China.
| |
Collapse
|
2
|
Hou W, Dai W, Huang H, Liu SL, Liu J, Huang LJ, Huang XH, Zeng JL, Gan ZW, Zhang ZY, Lan JX. Pharmacological activity and mechanism of pyrazines. Eur J Med Chem 2023; 258:115544. [PMID: 37300915 DOI: 10.1016/j.ejmech.2023.115544] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/25/2023] [Accepted: 06/03/2023] [Indexed: 06/12/2023]
Abstract
Heterocycles are common in the structure of drugs used clinically to deal with diseases. Such drugs usually contain nitrogen, oxygen and sulfur, which possess electron-accepting capacity and can form hydrogen bonds. These properties often bring enhanced target binding ability to these compounds when compared to alkanes. Pyrazine is a nitrogen-containing six-membered heterocyclic ring and many of its derivatives are identified as bioactive molecules. We review here the most active pyrazine compounds in terms of their structure, activity in vitro and in vivo (mainly antitumor activity) and the reported mechanisms of action. References have been downloaded through Web of Science, PubMed, Science Direct, Google Scholar and SciFinder Scholar. Publications reporting only the chemistry of pyrazine derivatives are beyond the scope of this review and have not been included. We found that compounds in which a pyrazine ring was fused into other heterocycles especially pyrrole or imidazole were the highly studied pyrazine derivatives, whose antineoplastic activity had been widely investigated. To the best of our knowledge, this is the first review of pyrazine derivatives and their bioactivity, especially their antitumor activity. This review should be useful for those engaged in development of medications based on heterocyclic compounds especially those based on pyrazine.
Collapse
Affiliation(s)
- Wen Hou
- College of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China
| | - Wei Dai
- College of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China
| | - Hao Huang
- College of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China
| | - Sheng-Lan Liu
- College of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China
| | - Jun Liu
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Le-Jun Huang
- College of Rehabilitation, Gannan Medical University, Ganzhou, 341000, PR China
| | - Xian-Hua Huang
- College of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China
| | - Jun-Lin Zeng
- HuanKui Academy, Nanchang University, Nanchang, 330006, PR China
| | - Zhi-Wei Gan
- College of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China
| | - Zhen-Yu Zhang
- College of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China
| | - Jin-Xia Lan
- College of Public Health and Health Management, Gannan Medical University, Ganzhou, 341000, PR China.
| |
Collapse
|
3
|
Moossavi M, Lu X, Herrmann J, Xu X. Molecular mechanisms of anthracycline induced cardiotoxicity: Zebrafish come into play. Front Cardiovasc Med 2023; 10:1080299. [PMID: 36970353 PMCID: PMC10036604 DOI: 10.3389/fcvm.2023.1080299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/22/2023] [Indexed: 03/12/2023] Open
Abstract
Anthracyclines are among the most potent chemotherapeutics; however, cardiotoxicity significantly restricts their use. Indeed, anthracycline-induced cardiotoxicity (AIC) fares among the worst types of cardiomyopathy, and may only slowly and partially respond to standard heart failure therapies including β-blockers and ACE inhibitors. No therapy specifically designed to treat anthracycline cardiomyopathy at present, and neither is it known if any such strategy could be developed. To address this gap and to elucidate the molecular basis of AIC with a therapeutic goal in mind, zebrafish has been introduced as an in vivo vertebrate model about a decade ago. Here, we first review our current understanding of the basic molecular and biochemical mechanisms of AIC, and then the contribution of zebrafish to the AIC field. We summarize the generation of embryonic zebrafish AIC models (eAIC) and their use for chemical screening and assessment of genetic modifiers, and then the generation of adult zebrafish AIC models (aAIC) and their use for discovering genetic modifiers via forward mutagenesis screening, deciphering spatial-temporal-specific mechanisms of modifier genes, and prioritizing therapeutic compounds via chemical genetic tools. Several therapeutic target genes and related therapies have emerged, including a retinoic acid (RA)-based therapy for the early phase of AIC and an autophagy-based therapy that, for the first time, is able to reverse cardiac dysfunction in the late phase of AIC. We conclude that zebrafish is becoming an important in vivo model that would accelerate both mechanistic studies and therapeutic development of AIC.
Collapse
Affiliation(s)
- Maryam Moossavi
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Xiaoguang Lu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Correspondence: Xiaolei Xu
| |
Collapse
|
4
|
Wu J, Li K, Liu Y, Feng A, Liu C, Adu-Amankwaah J, Ji M, Ma Y, Hao Y, Bu H, Sun H. Daidzein ameliorates doxorubicin-induced cardiac injury by inhibiting autophagy and apoptosis in rats. Food Funct 2023; 14:934-945. [PMID: 36541083 DOI: 10.1039/d2fo03416f] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Backgrounds: Doxorubicin (Dox) is a classical antitumor antibiotic widely restricted for use due to its cardiotoxicity. Daidzein (Daid) is a soy isoflavone that enhances antioxidant enzyme systems and inhibits apoptosis to prevent cardiovascular diseases. In this study, we intended to assess whether Daid protects against Dox-induced cardiotoxicity and explored its underlying mechanisms. Methods: Male Sprague-Dawley (SD) rats were divided into five groups: control (Ctrl), 40 mg per kg per day Daidzein (Daid), 3 mg per kg per week doxorubicin (Dox), 20 mg per kg per day Daidzein + 3 mg per kg per week doxorubicin (Daid20 + Dox) and 40 mg per kg per day Daidzein + 3 mg per kg per week doxorubicin (Daid40 + Dox) groups. Cardiac function assessments, immunohistochemistry (IHC) and immunofluorescence (IF) analyses were initially performed in each group of rats. Secondly, the cell proliferative capacity analysis, AO staining, and LC3 puncta analysis were employed to evaluate the cellular response to Dox in H9c2 cells. Ultimately, the protein expressions of cleaved caspase3, LC3 II, Bcl-2, Bax, Akt, p-Akt, and cyclin D1 were examined by western blotting. Results: Pretreatment with a low dose of Daid rather than a high dose significantly enhanced cardiac function and alleviated histopathological deterioration of cardiomyocytes induced by Dox. Daid downregulated the protein levels of Bax, LC3 II, cleaved caspase3 and p-Akt, while up-regulating Bcl-2 and cyclin D1. The Akt agonist SC79 could invalidate all the protective effects of Daid both in vivo and in vitro. Conclusions: Daid reduced autophagy and apoptosis by inhibiting the PI3K/Akt pathway, thereby protecting the hearts from Dox-induced cardiac damage.
Collapse
Affiliation(s)
- Jinxia Wu
- Department of Physiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Kexue Li
- Department of Physiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Yan Liu
- Department of Physiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Ailu Feng
- Department of Physiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Chunyang Liu
- Department of Physiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Joseph Adu-Amankwaah
- Department of Physiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Miaojin Ji
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, School of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Yanhong Ma
- Department of Physiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Yanling Hao
- Department of Physiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Huimin Bu
- Department of Physiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Hong Sun
- Department of Physiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| |
Collapse
|
5
|
Yi X, Wang F, Feng Y, Zhu J, Wu Y. Danhong injection attenuates doxorubicin-induced cardiotoxicity in rats via suppression of apoptosis: network pharmacology analysis and experimental validation. Front Pharmacol 2022; 13:929302. [PMID: 36071840 PMCID: PMC9441549 DOI: 10.3389/fphar.2022.929302] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 07/18/2022] [Indexed: 12/06/2022] Open
Abstract
Doxorubicin (DOX) is a potent chemotherapeutic agent that is used against various types of human malignancies. However, the associated risk of cardiotoxicity has limited its clinical application. Danhong injection (DHI) is a Chinese medicine with multiple pharmacological activities and is widely used for treating cardiovascular diseases. The aim of the present study was to evaluate the potential protective effect of DHI on DOX-induced cardiotoxicity in vivo and to investigate the possible underlying mechanisms. First, a sensitive and reliable HPLC−ESI-Q-TOF-MS/MS method was developed to comprehensively analyze the chemical compositions of DHI. A total of 56 compounds were identified, including phenolic acids, tanshinones, and flavonoids. Then, a DOX-induced chronic cardiotoxicity rat model was established to assess the therapeutic effect of DHI. As a result, DHI administration prevented the reduction in body weight and heart weight, and improved electrocardiogram performance. Additionally, the elevated levels of serum biochemical indicators were reduced, and the activities of oxidative enzymes were restored in the DOX-DHI group. Network pharmacology analysis further revealed that these effects might be attributed to 14 active compounds (e.g., danshensu, salvianolic acid A, salvianolic acid B, rosmarinic acid, and tanshinone IIA) and 15 potential targets (e.g., CASP3, SOD1, NOS3, TNF, and TOP2A). The apoptosis pathway was highly enriched according to the KEGG analysis. Molecular docking verified the good binding affinities between the active compounds and the corresponding apoptosis targets. Finally, experimental validation demonstrated that DHI treatment significantly increased the Bcl-2 level and suppressed DOX-induced Bax and caspase-3 expression in rat heart tissue. Furthermore, DHI treatment obviously decreased the apoptosis rate of DOX-treated H9c2 cells. These results indicate that DHI attenuated DOX-induced cardiotoxicity via regulating the apoptosis pathway. The present study suggested that DHI is a promising agent for the prevention of DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Xiaojiao Yi
- Department of Pharmacy, Affiliated Hangzhou Xixi Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fugen Wang
- Department of Pharmacy, Affiliated Hangzhou Xixi Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Feng
- Department of Pharmacy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Junfeng Zhu
- Department of Pharmacy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- *Correspondence: Junfeng Zhu, ; Yongjiang Wu,
| | - Yongjiang Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- *Correspondence: Junfeng Zhu, ; Yongjiang Wu,
| |
Collapse
|
6
|
Ren C, Liu K, Zhao X, Guo H, Luo Y, Chang J, Gao X, Lv X, Zhi X, Wu X, Jiang H, Chen Q, Li Y. Research Progress of Traditional Chinese Medicine in Treatment of Myocardial fibrosis. Front Pharmacol 2022; 13:853289. [PMID: 35754495 PMCID: PMC9213783 DOI: 10.3389/fphar.2022.853289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 05/02/2022] [Indexed: 11/13/2022] Open
Abstract
Effective drugs for the treatment of myocardial fibrosis (MF) are lacking. Traditional Chinese medicine (TCM) has garnered increasing attention in recent years for the prevention and treatment of myocardial fibrosis. This Article describes the pathogenesis of myocardial fibrosis from the modern medicine, along with the research progress. Reports suggest that Chinese medicine may play a role in ameliorating myocardial fibrosis through different regulatory mechanisms such as reduction of inflammatory reaction and oxidative stress, inhibition of cardiac fibroblast activation, reduction in extracellular matrix, renin-angiotensin-aldosterone system regulation, transforming growth Factor-β1 (TGF-β1) expression downregulation, TGF-β1/Smad signalling pathway regulation, and microRNA expression regulation. Therefore, traditional Chinese medicine serves as a valuable source of candidate drugs for exploration of the mechanism of occurrence and development, along with clinical prevention and treatment of MF.
Collapse
Affiliation(s)
- Chunzhen Ren
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Kai Liu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Xinke Zhao
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Huan Guo
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yali Luo
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Juan Chang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Gansu Provincial People’s Hospital, Lanzhou, China
| | - Xiang Gao
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Xinfang Lv
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Xiaodong Zhi
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Xue Wu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- The Second Hospital of Lanzhou University, Lanzhou, China
| | - Hugang Jiang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Qilin Chen
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yingdong Li
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
7
|
Construction and Screening of Fractional Library of Salviae Miltiorrhizae Radix et Rhizoma for the Rapid Identification of Active Compounds against Prostate Cancer. JOURNAL OF ONCOLOGY 2022; 2022:9955834. [PMID: 35251179 PMCID: PMC8894037 DOI: 10.1155/2022/9955834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/12/2022] [Indexed: 11/24/2022]
Abstract
Efficient screening of anticancer agents is in urgent need to develop new drugs that combat malignant tumors and drug resistance. In this study, a combined strategy composed by solvent partition and HPLC fractionation was developed to generate an herbal fraction library of Salviae Miltiorrhizae Radix et Rhizoma to quickly and efficiently screen anticancer agents. All library entries are directed into 96 well plates which are well mapped with HPLC chromatograms. The cell proliferation assay revealed seven active subfractions. Then, the major active ten peaks in these subfractions were prepared and isolated by semipreparative HPLC, and their inhibitory activities against prostate cancer cells were then tested at the same concentration level, leading to the identification of several active compounds. In addition, the structures of compounds arucadiol (2), 15,16-dihydrotanshinone I (4), methyl tanshinonate (5), cryptanshinone (7), 1,2-dihydrotanshinquinone I (9), and tanshinone IIA (10) were characterized by mass spectrometry and X-ray crystallographic analysis, and they were confirmed to be active in suppressing prostate cancer cell proliferation at 7.5 or 15 μg/mL, among which, the minor compounds 2, 4, and 5 showed higher activities than 9 and 10. This study provided a rapid strategy of identifying new anticancer agents in Salviae Miltiorrhizae Radix et Rhizoma, which can be applied in other herbal medicines.
Collapse
|
8
|
Shi Y, Li F, Shen M, Sun C, Hao W, Wu C, Xie Y, Zhang S, Gao H, Yang J, Zhou Z, Gao D, Qin Y, Han X, Liu S. Luteolin Prevents Cardiac Dysfunction and Improves the Chemotherapeutic Efficacy of Doxorubicin in Breast Cancer. Front Cardiovasc Med 2021; 8:750186. [PMID: 34722681 PMCID: PMC8548634 DOI: 10.3389/fcvm.2021.750186] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/16/2021] [Indexed: 12/19/2022] Open
Abstract
Background: Doxorubicin (Dox) is one of the most effective chemotherapy agents used in the treatment of solid tumors and hematological malignancies. However, it causes dose-related cardiotoxicity that may lead to heart failure in patients. Luteolin (Lut) is a common flavonoid that exists in many types of plants. It has been studied for treating various diseases such as hypertension, inflammatory disorders, and cancer. In this study, we evaluated the cardioprotective and anticancer effects of Lut on Dox-induced cardiomyopathy in vitro and in vivo to explore related mechanisms in alleviating dynamin-related protein (Drp1)-mediated mitochondrial apoptosis. Methods: MTT and LDH assay were used to determine the viability and toxicity of cardiomyocytes treated with Dox and Lut. Flow cytometry was used to examine ROS levels, and electron and confocal microscopy was employed to assess the mitochondrial morphology. The level of apoptosis was examined by Hoechst 33258 staining. The protein levels of myocardial fission protein and apoptosis-related protein were examined using Western blot. Transcriptome analysis of the protective effect of Lut against Dox-induced cardiac toxicity in myocardial cells was performed using RNA sequencing technology. The protective effects of Lut against cardiotoxicity mediated by Dox in zebrafish were quantified. The effect of Lut increase the antitumor activity of Dox in breast cancer both in vitro and in vivo were further employed. Results: Lut ameliorated Dox-induced toxicity in H9c2 and AC16 cells. The level of oxidative stress was downregulated by Lut after Dox treatment of myocardial cells. Lut effectively reduced the increased mitochondrial fission post Dox stimulation in cardiomyocytes. Apoptosis, fission protein Drp1, and Ser616 phosphorylation were also increased post Dox and reduced by Lut. In the zebrafish model, Lut significantly preserved the ventricular function of zebrafish after Dox treatment. Moreover, in the mouse model, Lut prevented Dox-induced cardiotoxicity and enhanced the cytotoxicity in triple-negative breast cancer by inhibiting proliferation and metastasis and inducing apoptosis.
Collapse
Affiliation(s)
- Youyang Shi
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Feifei Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Man Shen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenpin Sun
- Department of Breast Surgery (Integrated Traditional and Western Medicine), Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Hao
- Department of Breast Surgery (Integrated Traditional and Western Medicine), Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chunyu Wu
- Department of Breast Surgery (Integrated Traditional and Western Medicine), Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Xie
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuai Zhang
- Department of Breast Surgery (Integrated Traditional and Western Medicine), Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongzhi Gao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianfeng Yang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhongyan Zhou
- Department of Cardiovascular Research Laboratory, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dongwen Gao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuenong Qin
- Department of Breast Surgery (Integrated Traditional and Western Medicine), Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xianghui Han
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Sheng Liu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
9
|
Lu X, Lu L, Gao L, Wang Y, Wang W. Calycosin attenuates doxorubicin-induced cardiotoxicity via autophagy regulation in zebrafish models. Biomed Pharmacother 2021; 137:111375. [PMID: 33761601 DOI: 10.1016/j.biopha.2021.111375] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/02/2021] [Accepted: 02/08/2021] [Indexed: 10/22/2022] Open
Abstract
Anthracyclines are highly effective chemotherapeutics for antineoplastic treatment. However, cumulative cardiotoxicity is the main side effect with poor prognosis. No mechanism-based therapy is currently available to reverse chronic anthracycline-induced cardiotoxicity (AIC) after the deterioration of cardiac function. Calycosin (CA) is the main compound extracted from the traditional Chinese medicine Astragalus, and it has diverse beneficial effects, including autophagy modulation, anti-inflammatory and anti-tumor effects. Autophagy dysregulation is an important pathological event in AIC. Our study demonstrated a cardioprotective effect of CA in a zebrafish embryonic AIC model. To assess the effect of CA on late-onset chronic AIC, adult zebrafish were treated with CA 28 days after doxorubicin (DOX) injection, at which point heart function was obviously impaired. The results demonstrated that DOX blocked autophagic activity in adult zebrafish 8 weeks post-injection, and CA treatment improved heart function and restored autophagy. Further in vitro experiments demonstrated that atg7, which encodes an E1-like activating enzyme, may play an essential role in the CA regulation of autophagy. In conclusion, we used a rapid pharmacological screening system in embryo-adult zebrafish in vivo and elucidated the mechanism of gene targeting in vitro.
Collapse
Affiliation(s)
- Xiaoguang Lu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Linghui Lu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Li Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yong Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Wei Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
10
|
Ng PCI, Chan JYW, Leung RKK, Li J, Ren Z, Chan AWH, Xu Y, Lee SS, Wang R, Ji X, Zheng J, Chan DPC, Yew WW, Lee SMY. Role of oxidative stress in clofazimine-induced cardiac dysfunction in a zebrafish model. Biomed Pharmacother 2020; 132:110749. [PMID: 33017766 DOI: 10.1016/j.biopha.2020.110749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/19/2020] [Accepted: 09/07/2020] [Indexed: 10/23/2022] Open
Abstract
BACKGROUND Clofazimine (CFZ), a riminophenazine, is now commonly used in the treatment of multidrug-resistant tuberculosis. However, its use may be potentially associated with cardiac dysfunction in some individuals. In this study, the zebrafish heart, by merit of its developmental and genetic characteristics being in homology with that of human, was chosen as an animal model for evaluation of such dysfunction. METHODS Morphological and physiological parameters were used to assess cardiac dysfunction. Transcriptome analysis was performed, followed by validation with real-time quantitative PCR, for delineation of the relevant genomics. RESULTS Exposure of 2 dpf zebrafish to 4 mg/L CFZ for 2 days, adversely affected cardiac functions including significant decreases in HR, SV, CO, and FS, with observable pathophysiological developments of pericardial effusion and blood accumulation in the heart, in comparison with the control group. In addition, genes which respond to xenobiotic stimulus, related to oxygen transport, glutathione metabolism and extracellular matrix -receptor interactions, were significantly enriched among the differentially up-regulated genes. Antioxidant response element motif was enriched in the 5000 base pair upstream regions of the differentially expressed genes. Co-administration of N-acetylcysteine was shown to protect zebrafish against the development of CFZ-induced cardiac dysfunction. CONCLUSIONS This study suggests an important role of oxidative stress as a major pathogenetic mechanism of riminophenazine-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Phoebe C I Ng
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Judy Y W Chan
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Ross K K Leung
- School of Public Health, University of Hong Kong, Hong Kong, China; Stanley Ho Centre for Emerging Infectious Diseases, Chinese University of Hong Kong, Hong Kong, China
| | - J Li
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Z Ren
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Anthony W H Chan
- Department of Anatomical & Cellular Pathology, Chinese University of Hong Kong, Hong Kong, China
| | - Y Xu
- Stanley Ho Centre for Emerging Infectious Diseases, Chinese University of Hong Kong, Hong Kong, China
| | - S S Lee
- Stanley Ho Centre for Emerging Infectious Diseases, Chinese University of Hong Kong, Hong Kong, China
| | - R Wang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Xia Ji
- Faculty of Health Sciences, University of Macau, Macao, China
| | - Jun Zheng
- Faculty of Health Sciences, University of Macau, Macao, China
| | - Denise P C Chan
- Stanley Ho Centre for Emerging Infectious Diseases, Chinese University of Hong Kong, Hong Kong, China.
| | - W W Yew
- Stanley Ho Centre for Emerging Infectious Diseases, Chinese University of Hong Kong, Hong Kong, China.
| | - Simon M Y Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| |
Collapse
|
11
|
Functional Redox Proteomics Reveal That Salvia miltiorrhiza Aqueous Extract Alleviates Adriamycin-Induced Cardiomyopathy via Inhibiting ROS-Dependent Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5136934. [PMID: 32963697 PMCID: PMC7501560 DOI: 10.1155/2020/5136934] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/14/2020] [Accepted: 08/19/2020] [Indexed: 01/03/2023]
Abstract
The anticancer agent adriamycin (ADR) has long been recognized to induce a dose-limiting cardiotoxicity, while Salvia miltiorrhiza (SM) is a Chinese herb widely used for the treatment of cardiovascular disorders and its aqueous extract (SMAE) has shown anticancer as well as antioxidant effects. In the current study, we aimed at investigating the synergistic effect and potent molecular mechanisms of SMAE with a focus on the cardioprotective benefit observed under ADR adoption. Histopathological analysis indicated that SMAE could substantially alleviate cardiomyopathy and cell apoptosis caused by ADR. Meanwhile, the two-dimensional electrophoresis (2-DE) oxyblots demonstrated that SMAE treatment could effectively reduce carbonylation of specific proteins associated with oxidative stress response and various metabolic pathways in the presence of ADR. SMAE application also showed protective efficacy against ADR-mediated H9c2 cell death in a dose-dependent manner without causing any cytotoxicity and significantly attenuated the reactive oxygen species production. Particularly, the simultaneous administration of ADR and SMAE could remarkably suppress the growth of breast cancer cells. We also noticed that there was a marked upregulation of detoxifying enzyme system in the presence of SMAE, and its exposure also contributed to an increase in Nrf2 and HO-1 content as well. SMAE also amended the ERK/p53/Bcl-xL/caspase-3 signaling pathways and the mitochondrial dysfunction, which eventually attribute to apoptotic cathepsin B/AIF cascades. Correspondingly, both the ERK1/2 inhibitor (U0126) and pan-caspase inhibitor (Z-VAD-FMK) could at least partially abolish the ADR-associated cytotoxicity in H9c2 cells. Collectively, these results support that ROS apoptosis-inducing molecule release is closely involved in ADR-induced cardiotoxicity while SMAE could prevent or mitigate the causative cardiomyopathy through controlling multiple targets without compromising the efficacy of chemotherapy.
Collapse
|
12
|
Liang X, Wang S, Wang L, Ceylan AF, Ren J, Zhang Y. Mitophagy inhibitor liensinine suppresses doxorubicin-induced cardiotoxicity through inhibition of Drp1-mediated maladaptive mitochondrial fission. Pharmacol Res 2020; 157:104846. [PMID: 32339784 DOI: 10.1016/j.phrs.2020.104846] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/17/2020] [Accepted: 04/16/2020] [Indexed: 01/04/2023]
Abstract
Doxorubicin (DOX) is one of the most effective antineoplastic drugs. However, its clinical application has been greatly limited due to the development of cardiotoxicity with DOX utilization. A number of theories have been postulated for DOX-induced cardiotoxicity with a pivotal contribution from unchecked (excess) mitophagy and mitochondrial fission. Liensinine (LIEN), a newly identified mitophagy inhibitor, strengthens the antineoplastic efficacy of DOX although its action on hearts remains elusive. This study was designed to examine the effect of LIEN on DOX-induced cardiotoxicity and the underlying mechanisms involved with a focus on mitochondrial dynamics. Our data revealed that LIEN alleviated DOX-induced cardiac dysfunction and apoptosis through inhibition of dynamin-related protein 1 (Drp1)-mediated excess (unchecked) mitochondrial fission. LIEN treatment decreased Drp1 phosphorylation at Ser616 site, inhibited mitochondrial fragmentation, mitophagy (assessed by TOM20 and TIM23), oxidative stress, cytochrome C leakage, cardiomyocyte apoptosis, as well as improved mitochondrial function and cardiomyocyte contractile function in DOX-induced cardiac injury. In DOX-challenged neonatal mouse ventricular myocytes (NMVMs), LIEN-suppressed Drp1 phosphorylation, mitochondrial fragmentation, and apoptosis were blunted by Rab7 overexpression, the effect of which was reversed by the ERK inhibitor U0126. Moreover, activation of ERK or Drp1 abolished the protective effects of LIEN on cardiomyocyte mechanical anomalies. These data shed some lights towards understanding the role of LIEN as a new protective agent against DOX-associated cardiotoxicity without compromising its anti-tumor effects.
Collapse
Affiliation(s)
- Xinyue Liang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Shuyi Wang
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China; Center for Cardiovascular Research and Alternative Medicine, Laramie, WY 82071, USA
| | - Lifeng Wang
- Center for Cardiovascular Research and Alternative Medicine, Laramie, WY 82071, USA; Department of Physiology, Basic Medicine College, Xinjiang Medical University, Urumqi, Xinjiang 830011, China
| | - Asli F Ceylan
- Center for Cardiovascular Research and Alternative Medicine, Laramie, WY 82071, USA
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China; Center for Cardiovascular Research and Alternative Medicine, Laramie, WY 82071, USA.
| | - Yingmei Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| |
Collapse
|
13
|
Lu P, Xing Y, Peng H, Liu Z, Zhou Q(T, Xue Z, Ma Z, Kebebe D, Zhang B, Liu H. Physicochemical and Pharmacokinetic Evaluation of Spray-Dried Coformulation of Salvia miltiorrhiza Polyphenolic Acid and L-Leucine with Improved Bioavailability. J Aerosol Med Pulm Drug Deliv 2020; 33:73-82. [DOI: 10.1089/jamp.2019.1538] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Peng Lu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin, China
| | - Yue Xing
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin, China
| | - Hui Peng
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin, China
| | - Zhidong Liu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin, China
| | - Qi (Tony) Zhou
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana
| | - Zhifeng Xue
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin, China
| | - Zhe Ma
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin, China
| | - Dereje Kebebe
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin, China
- School of Pharmacy, Institute of Health Science, Jimma University, Jimma, Ethiopia
| | - Bing Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin, China
| | - Hongfei Liu
- College of Pharmacy, Jiangsu University, Zhenjiang, China
| |
Collapse
|
14
|
Jia J, Mo X, Liu J, Yan F, Wang N, Lin Y, Li H, Zheng Y, Chen D. Mechanism of danshensu-induced inhibition of abnormal epidermal proliferation in psoriasis. Eur J Pharmacol 2019; 868:172881. [PMID: 31866405 DOI: 10.1016/j.ejphar.2019.172881] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 12/15/2019] [Accepted: 12/17/2019] [Indexed: 01/16/2023]
Abstract
Psoriasis is a chronic, inflammatory skin disease with high incidence and high rates of relapse, for which no satisfactory treatments are currently available. Yes-associated protein (YAP) is highly expressed in psoriasis and may regulate the proliferation and apoptosis of keratinocytes. Danshen is a traditional Chinese medicine, commonly used in the treatment of psoriasis. Danshensu is the most abundant water-soluble component of Danshen, but its therapeutic mechanism is still unclear. In this study, MTT was used to detect the effects of different danshensu concentrations (0.125, 0.25, 0.5 mmol/l) on the proliferation of an M5-based psoriasis cell model. The effects of danshensu on cell cycle and apoptosis were detected by flow cytometry. Cyclins and apoptosis-related proteins were evaluated by Western blot. Danshensu (20, 40, 80 mg/kg/day) was administered intraperitoneally to the imiquimod (IMQ) psoriasis mouse model. After 7 days, the expression of YAP in the lesions was detected by immunohistochemistry and Western blot. We found that danshensu reduced the expression of YAP in the M5 psoriasis cell model, inhibited cell proliferation, induced cell cycle arrest in G0/G1 phase, and promoted cell apoptosis. All these effects were partly reverted by YAP overexpression. The skin lesions of IMQ mice were thinned and the scales reduced after intragastric administration of danshensu, which also resulted in dose-dependent inhibition of YAP expression. We concluded that danshensu prevents abnormal epidermis proliferation in psoriasis possibly by modulating YAP expression. Our work can provide a theoretical basis for the clinical application of Danshen in the treatment of psoriasis.
Collapse
Affiliation(s)
- Jinjing Jia
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Xiumei Mo
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Junfeng Liu
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Fenggen Yan
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Ning Wang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, School of Medicine, Xi'an, China
| | - Ying Lin
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Hongyi Li
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Yan Zheng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, School of Medicine, Xi'an, China.
| | - Dancan Chen
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.
| |
Collapse
|
15
|
Bao XY, Zheng Q, Tong Q, Zhu PC, Zhuang Z, Zheng GQ, Wang Y. Danshensu for Myocardial Ischemic Injury: Preclinical Evidence and Novel Methodology of Quality Assessment Tool. Front Pharmacol 2018; 9:1445. [PMID: 30618743 PMCID: PMC6297803 DOI: 10.3389/fphar.2018.01445] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 11/22/2018] [Indexed: 12/09/2022] Open
Abstract
Background: Danshensu (DSS) possesses unique bioactivity on the cardiovascular system. However, there is a lack of systematical summary of DSS for acute myocardial ischemia injury and no quality assessment tool for the systematical review of cell experiments. Here, we aimed to assess the preclinical evidences and possible mechanisms of DSS for myocardial ischemia injury, and to develop a quality assessment tool for the systematical review of cell experiments. Methods: Thirty-two studies with 473 animals and 134 cells were identified by searching seven databases. All data analysis was performed using RevMan 5.3. CAMARADES 10-item checklist was used to assess the methodological quality of animal experiments. A new 10-item checklist was first developed to assess the methodological quality of cell studies. Results: The score of study quality ranged from 3 to 7 points in animal studies, while the cell studies scored 3–6 points. Meta-analysis showed that DSS had significant effects on reducing myocardial infarct (MI) size in vivo, and increasing cell viability and reducing apoptosis rate in vitro compared with controls (P < 0.01). The possible mechanisms of DSS for MI are improving circulation, antioxidant, anti-apoptosis, anti-inflammatory, promoting angiogenesis, anti-excessive autophagy, anti-calcium overload, and improving energy metabolism. Conclusions: DSS could exert cardioprotective effect on myocardial ischemia injury, and thus is a probable candidate for further clinical trials andtreatment of AMI. In addition, the newly devloped 10-item checklist for assessing methodological quality of cell study that recommened to use the sysmatic review of cell studies.
Collapse
Affiliation(s)
- Xiao-Yi Bao
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qun Zheng
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qiang Tong
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Peng-Chong Zhu
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhuang Zhuang
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guo-Qing Zheng
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yan Wang
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
16
|
Zhang Y, Deng H, Zhou H, Lu Y, Shan L, Lee SM, Cui G. A novel agent attenuates cardiotoxicity and improves antitumor activity of doxorubicin in breast cancer cells. J Cell Biochem 2018; 120:5913-5922. [DOI: 10.1002/jcb.27880] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 09/19/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Ying Zhang
- Department of Bioengineering Zhuhai Campus of Zunyi Medical University Zhuhai China
| | - Hongkuan Deng
- Department of Pharmaceutical Engineering, School of Life Sciences, Shandong University of Technology Zibo China
| | - Hefeng Zhou
- Department of Bioengineering Zhuhai Campus of Zunyi Medical University Zhuhai China
| | - Yucong Lu
- Department of Bioengineering Zhuhai Campus of Zunyi Medical University Zhuhai China
| | - Luchen Shan
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio‐cerebrovascular Diseases, Jinan University College of Pharmacy Guangzhou China
| | - Simon Ming‐Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau Macao China
| | - Guozhen Cui
- Department of Bioengineering Zhuhai Campus of Zunyi Medical University Zhuhai China
| |
Collapse
|
17
|
Shi W, Deng H, Zhang J, Zhang Y, Zhang X, Cui G. Mitochondria-Targeting Small Molecules Effectively Prevent Cardiotoxicity Induced by Doxorubicin. Molecules 2018; 23:E1486. [PMID: 29921817 PMCID: PMC6099719 DOI: 10.3390/molecules23061486] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/13/2018] [Accepted: 06/13/2018] [Indexed: 02/07/2023] Open
Abstract
Doxorubicin (Dox) is a chemotherapeutic agent widely used for the treatment of numerous cancers. However, the clinical use of Dox is limited by its unwanted cardiotoxicity. Mitochondrial dysfunction has been associated with Dox-induced cardiotoxicity. To mitigate Dox-related cardiotoxicity, considerable successful examples of a variety of small molecules that target mitochondria to modulate Dox-induced cardiotoxicity have appeared in recent years. Here, we review the related literatures and discuss the evidence showing that mitochondria-targeting small molecules are promising cardioprotective agents against Dox-induced cardiac events.
Collapse
Affiliation(s)
- Wei Shi
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China.
| | - Hongkuan Deng
- School of Life Sciences, Shandong University of Technology, Zibo 255000, China.
| | - Jianyong Zhang
- Pharmacy School, Zunyi Medical University, Zunyi 563003, China.
| | - Ying Zhang
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China.
| | - Xiufang Zhang
- School of Life Sciences, Shandong University of Technology, Zibo 255000, China.
| | - Guozhen Cui
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China.
| |
Collapse
|
18
|
Yu J, Gao H, Wu C, Xu QM, Lu JJ, Chen X. Diethyl Blechnic, a Novel Natural Product Isolated from Salvia miltiorrhiza Bunge, Inhibits Doxorubicin-Induced Apoptosis by Inhibiting ROS and Activating JNK1/2. Int J Mol Sci 2018; 19:ijms19061809. [PMID: 29921821 PMCID: PMC6032151 DOI: 10.3390/ijms19061809] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 06/10/2018] [Indexed: 11/24/2022] Open
Abstract
Doxorubicin (DOX) is a widely used antineoplastic agent in clinics. However, its clinical application is largely limited by its cardiotoxicity. Diethyl blechnic (DB) is a novel compound isolated from Salvia miltiorrhiza Bunge. Here, we study the effect of DB on DOX-induced cardiotoxicity and its underlying mechanisms. Cellular viability was tested by 3-[-4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) and protein level was evaluated by Western blotting. 5,5’,6,6’-tetrachloro-1,1’,3,3’-tetraethylbenzimidazolylcarbocyanine iodide (JC-1) staining was performed to determine the mitochondrial membrane potential (MMP). Hoechst 33342 staining and TUNEL staining was performed to test the apoptosis. Reactive oxygen species (ROS) generation was investigated by using flow cytometry. DB significantly inhibited DOX-induced apoptosis in H9c2 cells and primary cultured cardiomyocytes. Moreover, DB decreased cell apoptotic morphological changes and reversed the mitochondrial membrane potential induced by DOX. Meanwhile, pre-treatment with DB increased the expression levels of B-cell lymphoma 2 (Bcl-2), B-cell lymphoma-extra-large (Bcl-xl), and survivin and reduced the expression levels of Bcl-2-associated X protein (Bax), p-p53, cytochrome c (cyt c), and cleaved-caspase 3, 7, 8, 9 in the protein levels in DOX-treated H9c2 cells. Furthermore, DB suppressed ROS generation. The DB-mediated protective effects were accompanied by increased c-Jun N-terminal kinase1/2 (JNK1/2) expression. In addition, SP600125, the inhibitor of JNK1/2, abolished the protective effect of DB. We concluded that DB protected cardiomyocytes against DOX-induced cytotoxicity by inhibiting ROS and activating the JNK1/2 pathway. Therefore, DB is a promising candidate as a cardioprotective agent against DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Jie Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau China.
| | - Hongwei Gao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau China.
| | - Chuanhong Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau China.
| | - Qiong-Ming Xu
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China.
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau China.
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau China.
| |
Collapse
|
19
|
Liao Q, Gong G, Siu SWI, Wong CTT, Yu H, Tse YC, Rádis-Baptista G, Lee SMY. A Novel ShK-Like Toxic Peptide from the Transcriptome of the Cnidarian Palythoa caribaeorum Displays Neuroprotection and Cardioprotection in Zebrafish. Toxins (Basel) 2018; 10:toxins10060238. [PMID: 29895785 PMCID: PMC6024583 DOI: 10.3390/toxins10060238] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 06/07/2018] [Accepted: 06/08/2018] [Indexed: 12/25/2022] Open
Abstract
Palythoa caribaeorum (class Anthozoa) is a zoantharian which, together with other cnidarians, like jellyfishes, hydra, and sea anemones, possesses specialized structures in its tissues, the cnidocytes, which deliver an array of toxins in order to capture prey and deter predators. The whole transcriptome of P. caribaeroum was deep sequenced, and a diversity of toxin-related peptide sequences were identified, and some retrieved for functional analysis. In this work, a peptide precursor containing a ShK domain, named PcShK3, was analyzed by means of computational processing, comprising structural phylogenetic analysis, model prediction, and dynamics simulation of peptide-receptor interaction. The combined data indicated that PcShK3 is a distinct peptide which is homologous to a cluster of peptides belonging to the ShK toxin family. In vivo, PcShK3 distributed across the vitelline membrane and accumulated in the yolk sac stripe of zebrafish larvae. Notably, it displayed a significant cardio-protective effect in zebrafish in concentrations inferior to the IC50 (<43.53 ± 6.45 µM), while in high concentrations (>IC50), it accumulated in the blood and caused pericardial edema, being cardiotoxic to zebrafish larvae. Remarkably, PcShK3 suppressed the 6-OHDA-induced neurotoxicity on the locomotive behavior of zebrafish. The present results indicated that PcShK3 is a novel member of ShK toxin family, and has the intrinsic ability to induce neuro- and cardio-protective effects or cause cardiac toxicity, according to its effective concentration.
Collapse
Affiliation(s)
- Qiwen Liao
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China.
| | - Guiyi Gong
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China.
| | - Shirley Weng In Siu
- Department of Computer and Information Science, Faculty of Science and Technology, University of Macau, Macau, China.
| | | | - Huidong Yu
- Shenzhen Rongxin Biotechnology Co., Ltd., Shenzhen 518054, China.
| | - Yu Chung Tse
- Department of Biology, South University of Science and Technology of China, Shenzhen 518055, China.
| | - Gandhi Rádis-Baptista
- Laboratory of Biochemistry and Biotechnology, Institute for Marine Sciences, Federal University of Ceará, Fortaleza 60165-081, Brazil.
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China.
| |
Collapse
|
20
|
A Theoretical Investigation About the Excited State Dynamical Mechanism for Doxorubicin Sensor. J CLUST SCI 2018. [DOI: 10.1007/s10876-018-1388-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
21
|
Li ZM, Xu SW, Liu PQ. Salvia miltiorrhizaBurge (Danshen): a golden herbal medicine in cardiovascular therapeutics. Acta Pharmacol Sin 2018; 39:802-824. [PMID: 29698387 PMCID: PMC5943903 DOI: 10.1038/aps.2017.193] [Citation(s) in RCA: 314] [Impact Index Per Article: 44.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 12/31/2017] [Indexed: 02/07/2023] Open
Abstract
Salvia miltiorrhiza Burge (Danshen) is an eminent medicinal herb that possesses broad cardiovascular and cerebrovascular protective actions and has been used in Asian countries for many centuries. Accumulating evidence suggests that Danshen and its components prevent vascular diseases, in particular, atherosclerosis and cardiac diseases, including myocardial infarction, myocardial ischemia/reperfusion injury, arrhythmia, cardiac hypertrophy and cardiac fibrosis. The published literature indicates that lipophilic constituents (tanshinone I, tanshinone IIa, tanshinone IIb, cryptotanshinone, dihydrotanshinone, etc) as well as hydrophilic constituents (danshensu, salvianolic acid A and B, protocatechuic aldehyde, etc) contribute to the cardiovascular protective actions of Danshen, suggesting a potential synergism among these constituents. Herein, we provide a systematic up-to-date review on the cardiovascular actions and therapeutic potential of major pharmacologically active constituents of Danshen. These bioactive compounds will serve as excellent drug candidates in small-molecule cardiovascular drug discovery. This article also provides a scientific rationale for understanding the traditional use of Danshen in cardiovascular therapeutics.
Collapse
Affiliation(s)
- Zhuo-ming Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences; National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-Sen University, Guangzhou 510006, China
| | - Suo-wen Xu
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, 14642, USA
| | - Pei-qing Liu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences; National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-Sen University, Guangzhou 510006, China
| |
Collapse
|
22
|
Wang L, Zhang X, Cui G, Chan JYW, Wang L, Li C, Shan L, Xu C, Zhang Q, Wang Y, Di L, Lee SMY. A novel agent exerts antitumor activity in breast cancer cells by targeting mitochondrial complex II. Oncotarget 2017; 7:32054-64. [PMID: 27081033 PMCID: PMC5077996 DOI: 10.18632/oncotarget.8410] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 03/02/2016] [Indexed: 12/24/2022] Open
Abstract
The mitochondrial respiratory chain, including mitochondrial complex II, has emerged as a potential target for cancer therapy. In the present study, a novel conjugate of danshensu (DSS) and tetramethylpyrazine (TMP), DT-010, was synthesized. Our results showed that DT-010 is more potent than its parental compounds separately or in combination, in inhibiting the proliferation of MCF-7 and MDA-MB-231 cells by inducing cytotoxicity and promoting cell cycle arrest. It also inhibited the growth of 4T1 breast cancer cells in vivo. DT-010 suppressed the fundamental parameters of mitochondrial function in MCF-7 cells, including basal respiration, ATP turnover, maximal respiration. Treatment with DT-010 in MCF-7 and MDA-MB-231 cells resulted in the loss of mitochondrial membrane potential and decreased ATP production. DT-010 also promoted ROS generation, while treatment with ROS scavenger, NAC (N-acetyl-L-cysteine), reversed DT-010-induced cytotoxicity. Further study showed that DT-010 suppressed succinate-induced mitochondrial respiration and impaired mitochondrial complex II enzyme activity indicating that DT-010 may inhibit mitochondrial complex II. Overall, our results suggested that the antitumor activity of DT-010 is associated with inhibition of mitochondrial complex II, which triggers ROS generation and mitochondrial dysfunction in breast cancer cells.
Collapse
Affiliation(s)
- Liang Wang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Xiaojing Zhang
- Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Guozhen Cui
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Judy Yuet-Wa Chan
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Li Wang
- Faculty of Health Sciences, University of Macau, Macao, China
| | - Chuwen Li
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Luchen Shan
- Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Changjiang Xu
- Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Qingwen Zhang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Yuqiang Wang
- Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Lijun Di
- Faculty of Health Sciences, University of Macau, Macao, China
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| |
Collapse
|
23
|
Tang F, Zhou X, Wang L, Shan L, Li C, Zhou H, Lee SMY, Hoi MPM. A novel compound DT-010 protects against doxorubicin-induced cardiotoxicity in zebrafish and H9c2 cells by inhibiting reactive oxygen species-mediated apoptotic and autophagic pathways. Eur J Pharmacol 2017; 820:86-96. [PMID: 29229534 DOI: 10.1016/j.ejphar.2017.12.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 12/03/2017] [Accepted: 12/08/2017] [Indexed: 01/25/2023]
Abstract
Doxorubicin (Dox) is an effective anti-cancer agent but limited by its cardiotoxicity, thus the search for pharmacological agents for enhancing anti-cancer activities and protecting against cardiotoxicity has been a subject of great interest. We have previously reported the synergistic anti-cancer effects of a novel compound DT-010. In the present study, we further investigated the cardioprotective effects of DT-010 in zebrafish embryos in vivo and the molecular underlying mechanisms in H9c2 cardiomyocytes in vitro. We showed that DT-010 prevented the Dox-induced morphological distortions in the zebrafish heart and the associated cardiac impairments, and especially improved ventricular functions. By using H9c2 cells model, we showed that DT-010 directly inhibited the generation of reactive oxygen species by Dox and protected cell death and cellular damage. We further observed that DT-010 protected against Dox-induced myocardiopathy via inhibiting downstream molecular pathways in response to oxidative stress, including reactive oxygen species-mediated MAPK signaling pathways ERK and JNK, and apoptotic pathways involving the activation of caspase 3, caspase 7, and PARP signaling. Recent studies also suggest the importance of alterations in cardiac autophagy in Dox cardiotoxicity. We further showed that DT-010 could inhibit the induction of autophagosomes formation by Dox via regulating the upstream Akt/AMPK/mTOR signaling. Since Dox-induced cardiotoxicity is multifactorial, our results suggest that multi-functional agent such as DT-010 might be an effective therapeutic agent for combating cardiotoxicity associated with chemotherapeutic agents such as Dox.
Collapse
Key Words
- Anti-apoptosis
- Anti-oxidative stress
- Autophagy regulation
- Cardioprotection
- DT-010, 4-(3,5,6-trimethylpyrazin-2-yl) hepta-1,6-dien-4-yl (R)-3-(3,4-dihydroxyphenyl)-2-hydroxypropanoate
- Danshensu (PubChem CID: 11600642),(R)-3-(3,4-Dihydroxyphenyl)-2-hydroxypropanoic acid
- Doxorubicin (PubChem CID: 443939),(7S,9S)-7-[(2R,4S,5S,6S)-4-amino-5-hydroxy-6-methyloxan-2-yl] oxy-6,9,11-trihydroxy-9-(2-hydroxyacetyl)-4-methoxy-8,10-dihydro-7H-tetracene-5,12-dione
- H9c2 cardiomyocytes
- Tetramethylpyrazine (PubChem CID: 14296),2,3,5,6-tetramethylpyrazine
- Zebrafish cardiotoxicity
Collapse
Affiliation(s)
- Fan Tang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Xinhua Zhou
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Liang Wang
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Luchen Shan
- Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Chuwen Li
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Hefeng Zhou
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau.
| | - Maggie Pui-Man Hoi
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau.
| |
Collapse
|
24
|
Lin B, Zhang L, Li D, Sun H. MED23 in endocrinotherapy for breast cancer. Oncol Lett 2017; 13:4679-4684. [PMID: 28588722 PMCID: PMC5452902 DOI: 10.3892/ol.2017.6036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 02/21/2017] [Indexed: 12/29/2022] Open
Abstract
We investigated the role of the transcriptional mediator subunit 23 (MED23) in everolimus drug resistance, invasion and metastasis during breast cancer treatment and its molecular mechanism. We also evaluated the endocrinotherapy and prevention method for breast cancer. Breast cancer cell strains were established that can continuously express MED23, as well as inducible MED23-shRNA expression plasmids. The inductive agent, doxycycline (Dox), was added to the water for long-term silencing of MED23 in intratumoral cells. We conducted experiments on the role of MED23 in the regulation of invasion and metastasis of breast cancer using cell culture, western blotting, MTT proliferation experiment, fluorescent quantitative PCR and chromatin immunoprecipitation (ChIP). The silencing of MED23 significantly inhibited cellular growth and proliferation as well as soft agar cloning. Silencing of MED23 strengthened the sensitivity of the everolimus-resistant breast cancer cell strains BT474 and MCF-7/ADM cells to everolimus medication. The silencing of MED23, in combination with everolimus, inhibits the cell cycle progress of breast cancer cells. ChIP indicated that the mutual regulation of HER2 and MED23 also participates in the formation of the everolimus drug resistance mechanism. Therefore, MED23 plays an important role in everolimus drug resistance, invasion, and metastasis of breast cancer. As a potential molecular therapeutic target of breast cancer, MED23 overcomes drug resistance in clinical endocrinotherapy and controls the distal relapse and metastasis in breast cancer by the targeted silencing of MED23.
Collapse
Affiliation(s)
- Benrui Lin
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Lan Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Dinuo Li
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Hongzhi Sun
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| |
Collapse
|
25
|
Leung WK, Gao L, Siu PM, Lai CW. Diabetic nephropathy and endothelial dysfunction: Current and future therapies, and emerging of vascular imaging for preclinical renal-kinetic study. Life Sci 2016; 166:121-130. [PMID: 27765534 DOI: 10.1016/j.lfs.2016.10.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 10/13/2016] [Accepted: 10/14/2016] [Indexed: 02/07/2023]
Abstract
An explosion in global epidemic of type 2 diabetes mellitus poses major rise in cases with vascular endothelial dysfunction ranging from micro- (retinopathy, nephropathy and neuropathy) to macro-vascular (atherosclerosis and cardiomyopathy) conditions. Functional destruction of endothelium is regarded as an early event that lays the groundwork for the development of renal microangiopathy and subsequent clinical manifestation of nephropathic symptoms. Recent research has shed some light on the molecular mechanisms of type 2 diabetes-associated comorbidity of endothelial dysfunction and nephropathy. Stemming from currently proposed endothelium-centered therapeutic strategies for diabetic nephropathy, this review highlighted some most exploited pathways that involve the intricate coordination of vasodilators, vasoconstrictors and vaso-modulatory molecules in the pathogenesis of diabetic nephropathy. We also emphasized the emerging roles of oxidative and epigenetic modifications of microvasculature as our prospective therapeutics for diabetic renal diseases. Finally, this review in particular addressed the potential use of multispectral optoacoustic tomography in real-time, minimally-invasive vascular imaging of small experimental animals for preclinical renal-kinetic drug trials.
Collapse
Affiliation(s)
- Wilson Kc Leung
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hung Hom, HKSAR, China
| | - L Gao
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hung Hom, HKSAR, China
| | - Parco M Siu
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hung Hom, HKSAR, China
| | - Christopher Wk Lai
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hung Hom, HKSAR, China.
| |
Collapse
|
26
|
Wang L, Chan JY, Zhou X, Cui G, Yan Z, Wang L, Yan R, Di L, Wang Y, Hoi MP, Shan L, Lee SM. A Novel Agent Enhances the Chemotherapeutic Efficacy of Doxorubicin in MCF-7 Breast Cancer Cells. Front Pharmacol 2016; 7:249. [PMID: 27559313 PMCID: PMC4979254 DOI: 10.3389/fphar.2016.00249] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 07/27/2016] [Indexed: 12/20/2022] Open
Abstract
We have previously demonstrated that DT-010, a novel conjugate of danshensu (DSS) and tetramethylpyrazine (TMP), displays anti-tumor effects in breast cancer cells both in vitro and in vivo. In the present study, we investigated whether DT-010 enhances the chemotherapeutic effect of doxorubicin (Dox) in MCF-7 breast cancer cells and exerts concurrent cardioprotective benefit at the same time. Our findings showed that DT-010 was more potent than TMP, DSS, or their combination in potentiating Dox-induced toxicity in MCF-7 cells. Co-treatment with DT-010 and Dox increased apoptosis in MCF-7 cells relative to Dox alone. Further study indicated that glycolytic capacity, glycolytic reserve and lactate level of MCF-7 cells were significantly inhibited after DT-010 treatment. DT-010 also increased the expression of the pro-survival protein GRP78, which was inhibited by co-treatment with Dox. Both endoplasmic reticulum stress inhibitor 4-PBA and knockdown of the expression of GRP78 protein potentiated DT-010-mediated apoptosis in MCF-7 cells. Moreover, DT-010 inhibited Dox-induced cardiotoxicity in H9c2 myoblasts. In conclusion, DT-010 and Dox confer synergistic anti-tumor effect in MCF-7 breast cancer cells through downregulation of the glycolytic pathway and inhibition of the expression of GRP78. Meanwhile, DT-010 also protects against Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Liang Wang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau Macao, China
| | - Judy Y Chan
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau Macao, China
| | - Xinhua Zhou
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau Macao, China
| | - Guozhen Cui
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau Macao, China
| | - Zhixiang Yan
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau Macao, China
| | - Li Wang
- Faculty of Health sciences, University of Macau Macao, China
| | - Ru Yan
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau Macao, China
| | - Lijun Di
- Faculty of Health sciences, University of Macau Macao, China
| | - Yuqiang Wang
- Institute of New Drug Research, College of Pharmacy, Jinan University Guangzhou, China
| | - Maggie P Hoi
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau Macao, China
| | - Luchen Shan
- Institute of New Drug Research, College of Pharmacy, Jinan University Guangzhou, China
| | - Simon M Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau Macao, China
| |
Collapse
|
27
|
Xi L. Visnagin-a new protectant against doxorubicin cardiotoxicity? Inhibition of mitochondrial malate dehydrogenase 2 (MDH2) and beyond. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:65. [PMID: 27004212 DOI: 10.3978/j.issn.2305-5839.2015.10.43] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Lei Xi
- Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298-0204, USA
| |
Collapse
|