1
|
Gu J, Zhang H, Wang M, Zhou Y, Deng Z, Shi R. Typical and atypical metabolic characteristics of three iridaceae isoflavone components: in vitro and in silico studies. Front Pharmacol 2025; 16:1522857. [PMID: 40008132 PMCID: PMC11850249 DOI: 10.3389/fphar.2025.1522857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Background Belamcanda chinensis (L.) DC (Chinese name Shegan) has been widely used because of its pharmacological activity and remarkable therapeutic effects in sore throat. Tectorigenin, irigenin, and irisflorentin have been recognized as important quality indicators in Shegan. However, the metabolic characteristics of isoflavone aglycones remain unclear. Methods In this study, human liver microsomes (HLMs) and Cytochrome P450 (CYP) recombinant enzymes were used to study the metabolic stability, identify the metabolic pathways and enzyme kinetics of these three components, and elucidate their possible binding sites through molecular docking. Results When tectorigenin, irigenin, and irisflorentin were co-incubated with HLMs and CYP recombinant enzymes, hydroxylation metabolite for tectorigenin, demethylated metabolite for irigenin, and 6,7-dihydroxy-5,3',4',5'-tetramethoxy isoflavone originating from irisflorentin were identified. CYP2E1 and CYP3A4 have high metabolic rates for tectorigenin, whereas CYP2C19 and CYP1A2 are the most important metabolic enzymes for irigenin and irisflorentin, respectively. The kinetics showed that the metabolism of tectorigenin and irigenin conformed to the Michaelis-Menten model, while the Eadie-Hofstee plot of irisflorentin yielded a convex curve with a unique "hooked" characteristic, and it conformed to the sigmoidal kinetics characteristic. Furthermore, molecular simulations showed that tectorigenin and irigenin bind to the orthosteric site of CYP isoforms via hydrogen bonds and π-π stacking, whereas irisflorentin is principally bound to CYP1A2 via π-π stacking and hydrophobic interactions. Conclusion Collectively, these Iridaceae isoflavone aglycones can be metabolized by CYP enzymes with typical or atypical kinetic characteristics. These results lay a foundation for a better understanding of the in vivo processes of these components.
Collapse
Affiliation(s)
- Jifeng Gu
- Department of Pharmacy, Eye and ENT Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Huishan Zhang
- Science and Technology Experimental Center, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mei Wang
- Science and Technology Experimental Center, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuyang Zhou
- Science and Technology Experimental Center, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Pharmacology, Addiction Science, Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Zhipeng Deng
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Rong Shi
- Science and Technology Experimental Center, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
2
|
Liu X, Li Z. The role and mechanism of epigenetics in anticancer drug-induced cardiotoxicity. Basic Res Cardiol 2025; 120:11-24. [PMID: 38724618 DOI: 10.1007/s00395-024-01054-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/20/2024] [Accepted: 05/03/2024] [Indexed: 05/23/2024]
Abstract
Cardiovascular disease is the main factor contributing to the global burden of diseases, and the cardiotoxicity caused by anticancer drugs is an essential component that cannot be ignored. With the development of anticancer drugs, the survival period of cancer patients is prolonged; however, the cardiotoxicity caused by anticancer drugs is becoming increasingly prominent. Currently, cardiovascular disease has emerged as the second leading cause of mortality among long-term cancer survivors. Anticancer drug-induced cardiotoxicity has become a frontier and hot topic. The discovery of epigenetics has given the possibility of environmental changes in gene expression, protein synthesis, and traits. It has been found that epigenetics plays a pivotal role in promoting cardiovascular diseases, such as heart failure, coronary heart disease, and hypertension. In recent years, increasing studies have underscored the crucial roles played by epigenetics in anticancer drug-induced cardiotoxicity. Here, we provide a comprehensive overview of the role and mechanisms of epigenetics in anticancer drug-induced cardiotoxicity.
Collapse
Affiliation(s)
- Xuening Liu
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Zijian Li
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China.
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China.
| |
Collapse
|
3
|
Yin B, Ren J, Liu X, Zhang Y, Zuo J, Wen R, Pei H, Lu M, Zhu S, Zhang Z, Wang Z, Zhai Y, Ma Y. Astaxanthin mitigates doxorubicin-induced cardiotoxicity via inhibiting ferroptosis and autophagy: a study based on bioinformatic analysis and in vivo/ vitro experiments. Front Pharmacol 2025; 16:1524448. [PMID: 39906141 PMCID: PMC11790656 DOI: 10.3389/fphar.2025.1524448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/02/2025] [Indexed: 02/06/2025] Open
Abstract
Background Doxorubicin (DOX), a widely employed chemotherapeutic agent in cancer treatment, has seen restricted use in recent years owing to its associated cardiotoxicity. Current reports indicate that doxorubicin-induced cardiotoxicity (DIC) is a complex phenomenon involving various modes of cell death. Astaxanthin (ASX), a natural carotenoid pigment, has garnered significant attention for its numerous health benefits. Recent studies have shown that ASX has a broad and effective cardiovascular protective effect. Our study aims to investigate the protective effects of ASX against DIC and elucidate its underlying mechanisms. This has substantial practical significance for the clinical application of DOX. Methods Bioinformatic analyses were conducted using transcriptomic data from the gene expression omnibus (GEO) database to identify key mechanisms underlying DIC. Network pharmacology was employed to predict the potential pathways and targets through which ASX exerts its effects on DIC. In vitro experiments, following pretreatment with ASX, H9C2 cells were exposed to DOX. Cell viability, injury and the protein expression levels associated with ferroptosis and autophagy were assessed. In the animal experiments, rats underwent 4 weeks of gavage treatment with various doses of ASX, followed by intraperitoneal injections of DOX every 2 days during the final week. Histological, serum, and protein analyses were conducted to evaluate the effects of ASX on DIC. Results The bioinformatics analysis revealed that ferroptosis and autophagy are closely associated with the development of DIC. ASX may exert an anti-DIC effect by modulating ferroptosis and autophagy. The experimental results show that ASX significantly mitigates DOX-induced myocardial tissue damage, inflammatory response, oxidative stress, and damage to H9C2 cells. Mechanistically, ASX markedly ameliorates levels of ferroptosis and autophagy both in vitro and in vivo. Specifically, ASX upregulates solute carrier family 7 member 11 (SLC7A11) and glutathione peroxidase 4 (GPX4), while downregulating the expression of transferrin receptor 1 (TFRC), ferritin heavy chain (FTH1) and ferritin light chain (FTL). Additionally, ASX enhances the expression of P62 and decreases levels of Beclin1 and microtubule-associated proteins light chain 3 (LC3). Conclusion Our results indicate that ferroptosis and autophagy are critical factors influencing the occurrence and progression of DOX-induced cardiotoxicity. ASX can alleviate DIC by inhibiting ferroptosis and autophagy.
Collapse
Affiliation(s)
- Bowen Yin
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Jingyi Ren
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Xuanyi Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Yadong Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Jinshi Zuo
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Rui Wen
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Huanting Pei
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Miaomiao Lu
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Siqi Zhu
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Zhenao Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Ziyi Wang
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Yanyi Zhai
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Yuxia Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| |
Collapse
|
4
|
Jie S, Wenying G, Lebo S. Dehydroevodiamine Alleviates Doxorubicin-Induced Cardiomyocyte Injury by Regulating Neuregulin-1/ErbB Signaling. Cardiovasc Ther 2024; 2024:5538740. [PMID: 39742014 PMCID: PMC11646148 DOI: 10.1155/cdr/5538740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/19/2024] [Indexed: 01/03/2025] Open
Abstract
Background: Doxorubicin (DOX) is a widely used antitumor drug; however, its use is limited by the risk of serious cardiotoxicity. Dehydroevodiamine (DHE) is a quinazoline alkaloid which has antiarrhythmic effects. The aim of this study was to investigate the protective effect of DHE on doxorubicin-induced cardiotoxicity (DIC) and its potential mechanism. Materials and Methods: Rat H9c2 cardiomyocytes were exposed to DOX for 24 h to establish a DOX-induced cardiomyocyte injury model. DHE and ErbB inhibitor AG1478 were used to treat H9c2 cells to investigate their effects. Cell counting kit-8 (CCK-8) and lactate dehydrogenase (LDH) release assays were used to evaluate cell viability. Flow cytometry and caspase-3 activity assay were used to detect apoptosis. Western blot was used to detect the expression levels of apoptosis-related proteins and neuregulin-1 (NRG1)/ErbB pathway-related proteins. The levels of proinflammatory cytokines and markers of oxidative stress were also detected, respectively. Quantitative polymerase chain reaction (qPCR) was used to detect mRNA expression levels of hub genes. Results: DHE enhanced cardiomyocyte viability and decreased LDH release in a concentration- and time-dependent manner. DHE also significantly inhibited DOX-induced cardiomyocyte apoptosis, inflammation, and oxidative stress. Bioinformatics analysis showed that the protective mechanism of DHE against DIC was related to ErbB signaling pathway. DOX treatment significantly reduced NRG1, p-ErbB2, and p-ErbB4 protein expression levels in cardiomyocytes, while DHE pretreatment reversed this effect. ErbB inhibitor AG1478 reversed the protective effect of DHE on cardiomyocytes. Conclusion: DHE protects cardiomyocytes against DOX by regulating NRG1/ErbB pathway. DHE may be a potential agent for the prevention and treatment of DIC.
Collapse
Affiliation(s)
- Song Jie
- Department of Cardiothoracic Surgery, Ningbo Medical Center Lihuili Hospital of Ningbo University, No. 57, Xingning Rd, Ningbo City 315041, Zhejiang Province, China
| | - Guo Wenying
- Department of Digestive, Ningbo Medical Center Lihuili Hospital of Ningbo University, No. 57, Xingning Rd, Ningbo City 315041, Zhejiang Province, China
| | - Sun Lebo
- Department of Cardiothoracic Surgery, Ningbo Medical Center Lihuili Hospital of Ningbo University, No. 57, Xingning Rd, Ningbo City 315041, Zhejiang Province, China
| |
Collapse
|
5
|
Zhang Q, Pu Q, Jiang Z, Zhao J, Dai Y, Hu N, Han Y, Jiang N, Shi L, Zhao J, Ouyang K, Huang H, Jin K, Li J. Tectorigenin Reduces Dabie bandavirus-Induced Cytokine Storm by Regulating Toll-Like Receptor 7/Extracellular Signal-Regulated Kinase Pathway. Chem Biol Drug Des 2024; 104:e70005. [PMID: 39672675 DOI: 10.1111/cbdd.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 10/09/2024] [Accepted: 10/14/2024] [Indexed: 12/15/2024]
Abstract
Severe fever with thrombocytopenia syndrome (SFTS) is a severe emerging infectious disease caused by Dabie bandavirus (DBV). Tectorigenin has been demonstrated to exert anti-inflammatory effect. Here, we aimed to investigate the effects of tectorigenin on DBV-induced cytokine storm. Effects of tectorigenin on cytokines in DBV-infected THP-1 cells and plasma samples of Type I interferon receptor (IFNAR)-/- mice infected with DBV were detected. The changes in body weight and survival time of mice were recorded. The liver, spleen, kidney, and lymph node tissues were collected for hematoxylin-eosin staining. We demonstrated that tectorigenin reduced the expression levels of inflammatory cytokines in both DBV-infected THP-1 cells and plasma samples of IFNAR-/- mice infected with DBV. Tectorigenin attenuated DBV-induced histopathological changes in mice. Mechanistically, tectorigenin attenuated DBV-induced phosphorylation of inhibitor of kappa-B kinase alpha/beta (IKKα/β) of the nuclear factor-κB (NF-κB) signaling pathway, extracellular signal-regulated kinase (ERK) of the mitogen-activated protein kinase (MAPK) signaling pathway and might function by downregulation of Toll-like receptor. The result of this study suggested that tectorigenin exerted anti-inflammatory effects in vivo and in vitro and could serve as a novel potential therapeutic strategy for SFTS.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Infectious Disease, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qinqin Pu
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhengyi Jiang
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jie Zhao
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yan Dai
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Nannan Hu
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yaping Han
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Nan Jiang
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Luchen Shi
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiaying Zhao
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ke Ouyang
- Department of Infectious Disease, Nanjing Second Hospital, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Huaying Huang
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Ke Jin
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jun Li
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
6
|
Ononiwu CP, Joshua PE, Amah CC, Asomadu RO, Okorigwe EM, Nnemolisa CS, Ezeorba TPC, Nwanelo VO, Iyidiegwu FC, Duru JO, Okeke PN, Adiele OB. Cleistopholis patens root bark extract exerts cardioprotective effect against doxorubicin-induced myocardial toxicity in rats. Lab Anim Res 2024; 40:39. [PMID: 39551811 PMCID: PMC11572060 DOI: 10.1186/s42826-024-00225-3] [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: 07/22/2024] [Revised: 10/17/2024] [Accepted: 10/26/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND Myocardial Infarction still persists as the most prevalent cardiovascular disease and is a top cause of morbidity and mortality in doxorubicin treated cancer patients. This study evaluated the prophylactic effect of the ethanol root bark extract of Cleistopholis patens (ERBECP) against doxorubicin-induced myocardial infarction in wistar rats. Extraction, preliminary phytochemical analysis, acute toxicity study and body weight (b.w.) of ERBECP were achieved using standard methods. Phyto-constituents in ERBECP were indentified using Gas Chromatography-Mass Spectrometry (GC-MS) technique. Thirty (30) male albino Wistar rats of average b.w. ranging between 100 and 130 g were divided into six groups of five rats each. Groups I, II and III served as normal, doxorubicin (DOX) and standard (Vasoprin 150 mg/kg b.w) controls respectively, while groups IV, V and VI were orally pre-treated with the extract (200, 400 and 600 mg/kgb.w) for two weeks prior to intraperitoneal induction of cardiotoxicity with DOX (20 mg/kg bw) on day 14. RESULTS Disturbances in serum cardiac function bio-markers such as; Cardiac Troponin-I (CTnI), Creatine Kinase (CK), Lactate Dehydrogenase (LDH), Aspartate aminotransferase (AST), Alanine aminotransferase (ALT). Lipid profile markers such as; Total cholesterol (TC), Triacylglycerol (TAG), Low Density Lipoprotein (LDL), High Density Lipoprotein (HDL). Oxidative stress markers such as; Malondialdehyde (MDA), Superoxide Dismutase (SOD), Catalase (CAT), Glutathione (GSH) confirmed the induction of myocardial infarction. Histological assessment of heart tissues was performed to validate biochemical results. The GC-MS analysis of ERBECP identified a total of 69 compounds. Safety profile of the aqueous extract was safe for the animals up to the highest dose of 5000 mg/kg b.w. Pre-treatment of DOX group with ERBECP could significantly increase the b.w. compared to the DOX-treated group during the experimental period of 2 weeks. There were significant (p < 0.05) alterations in the levels of CTnI, CK, LDH, AST, ALT and lipid profile indices in the DOX control rats. Also, significant (p < 0.05) increase was observed in MDA and decrease in SOD, CAT and GSH in the DOX control rats. However, administration of the extract significantly (p < 0.05) normalized these alterations and reversed the architectural changes in the heart. The 69 compounds were screened against the target protein (CBR1); we identified seven hits based on the docking score and interactions with the active site residues. All the C. patens constituents had MW (g/mol) less than 500, HBA < 10 and HBD not more than 5. Apart, 9-Octadecenoic acid (Z)-, 2,3-dihydroxy propyl ester and Estra-1,3,5(10)-trien-17. beta. -ol, all the constituents had LD50 lower than 2000 mg/kg. CONCLUSIONS The findings reveals ERBECP demonstrated promising potential and can be exploited in the development novel cardiac therapeutic agents.
Collapse
Affiliation(s)
- Chidinma Pamela Ononiwu
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria
| | - Parker Elijah Joshua
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria
| | - Christian Chijioke Amah
- Department of Biochemistry, Faculty of Natural and Applied Sciences, State University of Medical and Applied Sciences Igbo-Eno, Nsukka, Enugu State, Nigeria.
| | - Rita Onyekachukwu Asomadu
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria
| | - Ekezie Matthew Okorigwe
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria
| | | | - Timothy Prince Chidike Ezeorba
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria
- Department of Molecular Biotechnology, School of Biosciences, University of Birmingham, Edgbaston, B15 2TT, UK
| | | | - Favour Chinagorom Iyidiegwu
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria
| | - Justin Onuawuchi Duru
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria
| | - Peace Nkiruka Okeke
- Department of Biotechnology, School of Medicine, 3900 University Blvd., Tyler, TX, 75799, USA
| | - Onyinyechi Becky Adiele
- Department of Science Laboratory Technology, Faculty of Physical Sciences, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria
| |
Collapse
|
7
|
Zhang Z, Du T, Wu N, Yang S, Wang J, Peng J, Jia Z, Dai J, Du X, Feng M, Chu H, Shen C. Sulfiredoxin 1 ameliorates doxorubicin-induced cardiotoxicity by suppressing oxidative stress and inflammation via the Sirt1/NLRP3 pathway. Int Immunopharmacol 2024; 141:113010. [PMID: 39182271 DOI: 10.1016/j.intimp.2024.113010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND Doxorubicin (DOX) is limited in clinical use due to its cardiotoxic side effects. Oxidative stress and inflammation are pivotal mechanisms underlying doxorubicin-induced cardiotoxicity (DIC). Sulfiredoxin 1 (Srxn1) plays a central role in antioxidant effects. However, the role of Srxn1 in DIC has not yet been fully elucidated. This study aims to explore the effects and underlying mechanisms of Srxn1 on DIC. METHODS We overexpressed Srxn1 in the myocardium using an adeno-associated virus 9 (AAV9) system, delivered through tail vein injection. C57BL/6 mice received intraperitoneal injections of DOX (4 mg/kg) weekly for four consecutive weeks to establish a mouse model of DIC. We used echocardiography, histopathological, and molecular techniques to elucidate the effects and mechanisms. RESULTS Our findings demonstrate that overexpression of Srxn1 significantly enhanced cardiac function and mitigated myocardial injury in mice exposed to DOX. Overexpressing Srxn1 attenuated oxidative stress and inflammation induced by DOX. Furthermore, Srxn1 overexpression led to upregulation of sirtuin 1 (Sirt1) expression and inhibited the activation of the NOD-like receptor protein 3 (NLRP3) inflammasome. Notably, the protective effects of Srxn1 were significantly abrogated by the Sirt1 inhibitor EX527. CONCLUSION The protective effects of Srxn1 against DOX-induced cardiac oxidative stress and inflammation operate by targeting the Sirt1/NLRP3 signaling pathway to alleviate DIC. Srxn1 could be a potential candidate for the treatment of DOX-induced myocardial injury.
Collapse
Affiliation(s)
- Zhaoxia Zhang
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, Ningbo 315000, China
| | - Tingsha Du
- Health Science Center, Ningbo University, Ningbo 315000, China
| | - Nan Wu
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, Ningbo 315000, China
| | - Shuwen Yang
- Health Science Center, Ningbo University, Ningbo 315000, China
| | - Jian Wang
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, Ningbo 315000, China
| | - Jianye Peng
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China.
| | - Zhenyu Jia
- Health Science Center, Ningbo University, Ningbo 315000, China
| | - Jiating Dai
- Health Science Center, Ningbo University, Ningbo 315000, China
| | - Xianfeng Du
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, Ningbo 315000, China
| | - Mingjun Feng
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, Ningbo 315000, China
| | - Huimin Chu
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, Ningbo 315000, China.
| | - Caijie Shen
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, Ningbo 315000, China.
| |
Collapse
|
8
|
Zhou H, Liu P, Guo X, Fang W, Wu C, Zhang M, Ji Z. Fibroblast-derived miR-425-5p alleviates cardiac remodelling in heart failure via inhibiting the TGF-β1/Smad signalling. J Cell Mol Med 2024; 28:e70199. [PMID: 39527465 PMCID: PMC11552651 DOI: 10.1111/jcmm.70199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/15/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
The pathological activation of cardiac fibroblasts (CFs) plays a crucial role in the development of pressure overload-induced cardiac remodelling and subsequent heart failure (HF). Growing evidence demonstrates that multiple microRNAs (miRNAs) are abnormally expressed in the pathophysiologic process of cardiovascular diseases, with miR-425 recently reported to be potentially involved in HF. In this study, we aimed to investigate the effects of fibroblast-derived miR-425-5p in pressure overload-induced HF and explore the underlying mechanisms. C57BL/6 mice were injected with a recombinant adeno-associated virus specifically designed to overexpress miR-425-5p in CFs, followed by transverse aortic constriction (TAC) surgery. Neonatal mouse CFs (NMCFs) were transfected with miR-425-5p mimics and subsequently stimulated with angiotensin II (Ang II). We found that miR-425-5p levels were significantly downregulated in HF mice and Ang II-treated NMCFs. Notably, fibroblast-specific overexpression of miR-425-5p markedly inhibited the proliferation and differentiation of CFs, thereby alleviating myocardial fibrosis, cardiac hypertrophy and systolic dysfunction. Mechanistically, the cardioprotective actions of miR-425-5p may be achieved by targeting the TGF-β1/Smad signalling. Interestingly, miR-425-5p mimics-treated CFs could also indirectly affect cardiomyocyte hypertrophy in this course. Together, our findings suggest that fibroblast-derived miR-425-5p mitigates TAC-induced HF, highlighting miR-425-5p as a potential diagnostic and therapeutic target for treating HF patients.
Collapse
Affiliation(s)
- Haijia Zhou
- Department of CardiologyTangdu Hospital, Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Pengyun Liu
- Department of CardiologyTangdu Hospital, Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Xuelin Guo
- Department of CardiologyTangdu Hospital, Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Wei Fang
- Department of CardiologyTangdu Hospital, Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Chan Wu
- Department of CardiologyTangdu Hospital, Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Mingming Zhang
- Department of CardiologyTangdu Hospital, Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Zhaole Ji
- Department of CardiologyTangdu Hospital, Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| |
Collapse
|
9
|
Wang R, Ma S, Yang J, Luo K, Qian Q, Pan J, Liang K, Wang Y, Gao Y, Li M. Sodium Hydrosulfide Protects Rats from Hypobaric-Hypoxia-Induced Acute Lung Injury. Int J Mol Sci 2024; 25:10734. [PMID: 39409062 PMCID: PMC11477091 DOI: 10.3390/ijms251910734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
Hydrogen sulfide (H2S), as a key gas signaling molecule, plays an important role in regulating various diseases, with appropriate concentrations providing antioxidative, anti-inflammatory, and anti-apoptotic effects. The specific role of H2S in acute hypoxic injury remains to be clarified. This study focuses on the H2S donor sodium hydrosulfide (NaHS) and explores its protective effects and mechanisms against acute hypoxic lung injury. First, various mouse hypoxia models were established to evaluate H2S's protection in hypoxia tolerance. Next, a rat model of acute lung injury (ALI) induced by hypoxia at 6500 m above sea level for 72 h was created to assess H2S's protective effects and mechanisms. Evaluation metrics included blood gas analysis, blood routine indicators, lung water content, and lung tissue pathology. Additionally, LC-MS/MS and bioinformatic analyses were combined in performing quantitative proteomics on lung tissues from the normoxic control group, the hypoxia model group, and the hypoxia model group with NaHS treatment to preliminarily explore the protective mechanisms of H2S. Further, enzyme-linked immunosorbent assays (ELISA) were used to measure oxidative stress markers and inflammatory factors in rat lung tissues. Lastly, Western blot analysis was performed to detect Nrf2, HO-1, P-NF-κB, NF-κB, HIF-1α, Bcl-2, and Bax proteins in lung tissues. Results showed that H2S exhibited significant anti-hypoxic effects in various hypoxia models, effectively modulating blood gas and blood routine indicators in ALI rats, reducing pulmonary edema, improving lung tissue pathology, and alleviating oxidative stress, inflammatory responses, and apoptosis levels.
Collapse
Affiliation(s)
- Renjie Wang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (R.W.); (S.M.); (J.Y.); (K.L.); (K.L.)
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Q.Q.); (J.P.); (Y.W.)
| | - Shuhe Ma
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (R.W.); (S.M.); (J.Y.); (K.L.); (K.L.)
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Q.Q.); (J.P.); (Y.W.)
| | - Jun Yang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (R.W.); (S.M.); (J.Y.); (K.L.); (K.L.)
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Q.Q.); (J.P.); (Y.W.)
| | - Kai Luo
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (R.W.); (S.M.); (J.Y.); (K.L.); (K.L.)
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Q.Q.); (J.P.); (Y.W.)
| | - Qingyuan Qian
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Q.Q.); (J.P.); (Y.W.)
- College of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Jinchao Pan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Q.Q.); (J.P.); (Y.W.)
- Faculty of Environment and Life, Beijing University of Technology, Beijing 100083, China
| | - Keke Liang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (R.W.); (S.M.); (J.Y.); (K.L.); (K.L.)
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Q.Q.); (J.P.); (Y.W.)
| | - Yihao Wang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Q.Q.); (J.P.); (Y.W.)
| | - Yue Gao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Q.Q.); (J.P.); (Y.W.)
- National Key Laboratory of Kidney Diseases, Beijing 100850, China
| | - Maoxing Li
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Q.Q.); (J.P.); (Y.W.)
- National Key Laboratory of Kidney Diseases, Beijing 100850, China
| |
Collapse
|
10
|
Li Y, Yan J, Yang P. The mechanism and therapeutic strategies in doxorubicin-induced cardiotoxicity: Role of programmed cell death. Cell Stress Chaperones 2024; 29:666-680. [PMID: 39343295 PMCID: PMC11490929 DOI: 10.1016/j.cstres.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/26/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024] Open
Abstract
Doxorubicin (DOX) is the most commonly used anthracycline anticancer agent, while its clinical utility is limited by harmful side effects like cardiotoxicity. Numerous studies have elucidated that programmed cell death plays a significant role in DOX-induced cardiotoxicity (DIC). This review summarizes several kinds of programmed cell death, including apoptosis, pyroptosis, necroptosis, autophagy, and ferroptosis. Furthermore, oxidative stress, inflammation, and mitochondrial dysfunction are also important factors in the molecular mechanisms of DIC. Besides, a comprehensive understanding of specific signal pathways of DIC can be helpful to its treatment. Therefore, the related signal pathways are elucidated in this review, including sirtuin deacetylase (silent information regulator 2 [Sir2]) 1 (SIRT1)/nuclear factor erythroid 2-related factor 2, SIRT1/Klotho, SIRT1/Recombinant Sestrin 2, adenosine monophosphate-activated protein kinase, AKT, and peroxisome proliferator-activated receptor. Heat shock proteins function as chaperones, which play an important role in various stressful situations, especially in the heart. Thus, some of heat shock proteins involved in DIC are also included. Hence, the last part of this review focuses on the therapeutic research based on the mechanisms above.
Collapse
Affiliation(s)
- Yanzhao Li
- Department of Second Clinical Medical College, Southern Medical University, Guangzhou, China.
| | - Jing Yan
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Pingzhen Yang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
11
|
Yeh KL, Wu SW, Chiang CY, Chen CJ, Chen WY, Tseng CC, Kuan YH, Chou CC. Enhancing ocular protection against UVB: The role of irigenin in modulating oxidative stress and apoptotic pathways In Vivo. Biomed Pharmacother 2024; 179:117346. [PMID: 39232385 DOI: 10.1016/j.biopha.2024.117346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/07/2024] [Accepted: 08/22/2024] [Indexed: 09/06/2024] Open
Abstract
Oxidative damage contributes to age-related macular degeneration. Irigenin possesses diverse pharmacologic properties, including antioxidative and antiapoptotic effects. Our in vivo experiments indicated that irigenin mitigates UVB-induced histopathologic changes and oxidative DNA damage. Histologic analyses and TUNEL staining revealed that this compound dose-dependently ameliorated UVB-induced retinal damage and apoptosis. Furthermore, irigenin substantially reduced the level of 8-hydroxyguanosine, a biomarker of UVB-induced oxidative DNA damage. We further explored the molecular mechanisms that mediate the protective effects of irigenin. Our findings suggested that UVB-induced generation of ROS disrupts the stability of the mitochondrial membrane, activating intrinsic apoptotic pathways; the underlying mechanisms include the release of cytochrome c, activation of caspase-9 and caspase-3, and subsequent degradation of PARP-1. Notably, irigenin reversed mitochondrial disruption and apoptosis. It also modulated the Bax and Bcl-2 expression but influenced the mitochondrial apoptotic pathways. Our study highlights the role of the Nrf2 pathway in mitigating the effects of oxidative stress. We found that UVB exposure downregulated, but irigenin treatment upregulated the expression of Nrf2 and antioxidant enzymes. Therefore, irigenin activates the Nrf2 pathway to address oxidative stress. In conclusion, irigenin exhibits protective effects against UVB-induced ocular damage, evidenced by the diminution of histological alterations. It mitigates oxidative DNA damage and apoptosis in the retinal tissues by modulating the intrinsic apoptotic pathways and the AIF mechanisms. Furthermore, irigenin effectively reduces lipid peroxidation, enhancing the activity of antioxidant enzymes by stimulating the Nrf2 pathway. This protective mechanism underscores the potential benefit of irigenin in combating UVB-mediated ocular damage.
Collapse
Affiliation(s)
- Kun-Lin Yeh
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Sheng-Wen Wu
- Division of Nephrology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan; Department of Internal Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chen-Yu Chiang
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Chun-Jung Chen
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Wen-Ying Chen
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Ching-Chi Tseng
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China; Department of Dermatology, Shiso Municipal Hospital, Hyogo, Japan
| | - Yu-Hsiang Kuan
- Department of Pharmacology, School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Pharmacy, Chung Shan Medical University Hospital, Taichung, Taiwan.
| | - Chi-Chung Chou
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan.
| |
Collapse
|
12
|
Aghaei SM, Hosseini SM. Inflammation-related miRNAs in obesity, CVD, and NAFLD. Cytokine 2024; 182:156724. [PMID: 39106574 DOI: 10.1016/j.cyto.2024.156724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/08/2024] [Accepted: 08/01/2024] [Indexed: 08/09/2024]
Abstract
Obesity, cardiovascular diseases (CVD), and nonalcoholic fatty liver disease (NAFLD) pose significant worldwide health challenges, characterized by complex interplay among inflammatory pathways that underlie their development. In this review, we examine the contribution of inflammation and associated signaling molecules to the pathogenesis of these conditions, while also emphasizing the significant participation of non-coding RNAs (ncRNAs) in modulating inflammatory pathways. In the context of obesity, aberrant expression patterns of inflammatory-associated miRNAs play a contributory role in adipose tissue inflammation and insulin resistance, thereby exacerbating disturbances in metabolic homeostasis. Similarly, in CVD, dysregulated miRNA expression alters inflammatory reactions, disrupts endothelial function, and induces cardiac remodeling, thereby impacting the advancement of the disease. Moreover, in the context of NAFLD, inflammatory-associated miRNAs are implicated in mediating hepatic inflammation, lipid deposition, and fibrosis, underscoring their candidacy as promising therapeutic targets. Additionally, the competing endogenous RNA (ceRNA) network has emerged as a novel regulatory mechanism in the etiology of CVD, obesity, and NAFLD, wherein ncRNAs assume pivotal roles in facilitating communication across diverse molecular pathways. Moreover, in the concluding section, we underscored the potential efficacy of directing interventions towards inflammatory-related miRNAs utilizing herbal remedies and therapies based on exosome delivery systems as a promising strategy for ameliorating pathologies associated with inflammation in obesity, CVD, and NAFLD.
Collapse
Affiliation(s)
- Sayed Mohsen Aghaei
- Student Research Committee, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Sayed Mostafa Hosseini
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Fang X, Zhao H, Xu T, Wu H, Sheng G. Anti-Inflammatory and Antioxidant Effects of Irigenen Alleviate Osteoarthritis Progression through Nrf2/HO-1 Pathway. Pharmaceuticals (Basel) 2024; 17:1268. [PMID: 39458910 PMCID: PMC11510601 DOI: 10.3390/ph17101268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/07/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Osteoarthritis (OA) is a prevalent degenerative disease globally, characterized by cartilage degradation and joint dysfunction. Current treatments are insufficient for halting OA progression. Irigenin (IRI), a flavonoid extracted from natural plants with anti-inflammatory and antioxidant properties, has demonstrated potential in mitigating inflammation and oxidative stress in various diseases; however, its effects on OA remain unexplored. This study aims to evaluate the therapeutic effects of IRI on OA through in vivo and in vitro experiments and to elucidate the underlying molecular mechanisms. METHODS In vitro, chondrocytes were exposed to hydrogen peroxide (H2O2) to induce an oxidative stress environment and were then treated with IRI. Western blotting, RT-qPCR, immunofluorescence staining assays, flow cytometry, and apoptosis assays were employed to assess the effects of IRI on chondrocyte matrix homeostasis, inflammatory response, and apoptosis. In vivo, an OA rat model was treated with regular IRI injections, and therapeutic effects were evaluated using micro-CT, histological staining, and immunohistochemistry assays. RESULTS IRI treatment restored matrix homeostasis in chondrocytes and effectively suppressed H2O2-induced inflammation and apoptosis. Subsequent studies further revealed that IRI exerts its therapeutic effects by activating the Nrf2/HO-1 pathway. Inhibition of Nrf2 expression in chondrocytes partially blocked the anti-inflammatory and antioxidant effects of IRI. In the OA rat model, regular IRI injections effectively ameliorated cartilage degeneration. CONCLUSIONS This study identifies IRI as a promising strategy for OA treatment by modulating inflammation and apoptosis through the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
| | | | | | | | - Gaohong Sheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (X.F.); (H.Z.); (T.X.); (H.W.)
| |
Collapse
|
14
|
Bayer AL, Zambrano MA, Smolgovsky S, Robbe ZL, Ariza A, Kaur K, Sawden M, Avery A, London C, Asnani A, Alcaide P. Cytotoxic T cells drive doxorubicin-induced cardiac fibrosis and systolic dysfunction. NATURE CARDIOVASCULAR RESEARCH 2024; 3:970-986. [PMID: 39196030 DOI: 10.1038/s44161-024-00507-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 06/13/2024] [Indexed: 08/29/2024]
Abstract
Doxorubicin, the most prescribed chemotherapeutic drug, causes dose-dependent cardiotoxicity and heart failure. However, our understanding of the immune response elicited by doxorubicin is limited. Here we show that an aberrant CD8+ T cell immune response following doxorubicin-induced cardiac injury drives adverse remodeling and cardiomyopathy. Doxorubicin treatment in non-tumor-bearing mice increased circulating and cardiac IFNγ+CD8+ T cells and activated effector CD8+ T cells in lymphoid tissues. Moreover, doxorubicin promoted cardiac CD8+ T cell infiltration and depletion of CD8+ T cells in doxorubicin-treated mice decreased cardiac fibrosis and improved systolic function. Doxorubicin treatment induced ICAM-1 expression by cardiac fibroblasts resulting in enhanced CD8+ T cell adhesion and transformation, contact-dependent CD8+ degranulation and release of granzyme B. Canine lymphoma patients and human patients with hematopoietic malignancies showed increased circulating CD8+ T cells after doxorubicin treatment. In human cancer patients, T cells expressed IFNγ and CXCR3, and plasma levels of the CXCR3 ligands CXCL9 and CXCL10 correlated with decreased systolic function.
Collapse
Grants
- NIH K08 HL145019 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL162200 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL159907A U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- NIH R01 HL163172 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- Springboard Tier 1 Tufts University
- HL144477 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 906361 American Heart Association (American Heart Association, Inc.)
- 3R01HL144477-04S1 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL144477 NHLBI NIH HHS
- 906561 American Heart Association (American Heart Association, Inc.)
- HL165725 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- NIH U01CA272268 U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
Collapse
Affiliation(s)
| | | | | | | | - Abul Ariza
- CardioVascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Kuljeet Kaur
- Department of Immunology, Tufts University, Boston, MA, USA
| | - Machlan Sawden
- Department of Immunology, Tufts University, Boston, MA, USA
| | - Anne Avery
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Science, Colorado State University, Fort Collins, CO, USA
| | - Cheryl London
- Department of Immunology, Tufts University, Boston, MA, USA
- Cummings School of Veterinary Medicine, Tufts University, Boston, MA, USA
| | - Aarti Asnani
- CardioVascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Pilar Alcaide
- Department of Immunology, Tufts University, Boston, MA, USA.
| |
Collapse
|
15
|
AlAsmari AF, Al-Shehri MM, Algarini N, Alasmari NA, Alhazmi A, AlSwayyed M, Alharbi M, Alasmari F, Ali N. Role of diosmin in preventing doxorubicin-induced cardiac oxidative stress, inflammation, and hypertrophy: A mechanistic approach. Saudi Pharm J 2024; 32:102103. [PMID: 38799001 PMCID: PMC11127263 DOI: 10.1016/j.jsps.2024.102103] [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: 03/25/2024] [Accepted: 05/14/2024] [Indexed: 05/29/2024] Open
Abstract
Chemotherapeutic drugs, such as doxorubicin (Dox), are commonly used to treat a variety of malignancies. However, Dox-induced cardiotoxicity limits the drug's clinical applications. Hence, this study intended to investigate whether diosmin could prevent or limit Dox-induced cardiotoxicity in an animal setting. Thirty-two rats were separated into four distinct groups of controls, those treated with Dox (20 mg/kg, intraperitoneal, i.p.), those treated with diosmin 100 mg plus Dox, and those treated with diosmin 200 mg plus Dox. At the end of the experiment, rats were anesthetized and sacrificed and their blood and hearts were collected. Cardiac toxicity markers were analyzed in the blood, and the heart tissue was analyzed by the biochemical assays MDA, GSH, and CAT, western blot analysis (NF-kB, IL-6, TLR-4, TNF-α, iNOS, and COX-2), and gene expression analysis (β-MHC, BNP). Formalin-fixed tissue was used for histopathological studies. We demonstrated that a Dox insult resulted in increased oxidative stress, inflammation, and hypertrophy as shown by increased MDA levels and reduced GSH content and CAT activity. Furthermore, Dox treatment induced cardiac hypertrophy and damage, as evidenced by the biochemical analysis, ELISA, western blot analysis, and gene expression analysis. However, co-administration of diosmin at both doses, 100 mg and 200 mg, mitigated these alterations. Data derived from the current research revealed that the cardioprotective effect of diosmin was likely due to its ability to mitigate oxidative stress and inflammation. However, further study is required to investigate the protective effects of diosmin against Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Abdullah F. AlAsmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Mohammed M. Al-Shehri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Nasser Algarini
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Nada A. Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Alabid Alhazmi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Mohammed AlSwayyed
- Department of Pathology, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| |
Collapse
|
16
|
Szabados T, Molnár A, Kenyeres É, Gömöri K, Pipis J, Pósa B, Makkos A, Ágg B, Giricz Z, Ferdinandy P, Görbe A, Bencsik P. Identification of New, Translatable ProtectomiRs against Myocardial Ischemia/Reperfusion Injury and Oxidative Stress: The Role of MMP/Biglycan Signaling Pathways. Antioxidants (Basel) 2024; 13:674. [PMID: 38929113 PMCID: PMC11201193 DOI: 10.3390/antiox13060674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/22/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
INTRODUCTION Ischemic conditionings (ICon) were intensively investigated and several protective signaling pathways were identified. Previously, we have shown the role of matrix metalloproteinases (MMP) in myocardial ischemia/reperfusion injury (MIRI) and the cardioprotective role of biglycan (BGN), a small leucine-rich proteoglycan in vitro. Here, we hypothesized that cardiac MMP and BGN signaling are involved in the protective effects of ICon. METHODS A reverse target-microRNA prediction was performed by using the miRNAtarget™ 2.0 software to identify human microRNAs with a possible regulatory effect on MMP and BGN, such as on related genes. To validate the identified 1289 miRNAs in the predicted network, we compared them to two cardioprotective miRNA omics datasets derived from pig and rat models of MIRI in the presence of ICons. RESULTS Among the experimentally measured miRNAs, we found 100% sequence identity to human predicted regulatory miRNAs in the case of 37 porcine and 24 rat miRNAs. Upon further analysis, 42 miRNAs were identified as MIRI-associated miRNAs, from which 24 miRNAs were counter-regulated due to ICons. CONCLUSIONS Our findings highlight 24 miRNAs that potentially regulate cardioprotective therapeutic targets associated with MMPs and BGN in a highly translatable porcine model of acute myocardial infarction.
Collapse
Affiliation(s)
- Tamara Szabados
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, H-6720 Szeged, Hungary; (T.S.); (A.M.); (É.K.); (K.G.); (J.P.); (B.P.); (A.G.)
| | - Arnold Molnár
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, H-6720 Szeged, Hungary; (T.S.); (A.M.); (É.K.); (K.G.); (J.P.); (B.P.); (A.G.)
- Pharmahungary Group, Hajnóczy u. 6, H-6722 Szeged, Hungary; (B.Á.); (Z.G.); (P.F.)
| | - Éva Kenyeres
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, H-6720 Szeged, Hungary; (T.S.); (A.M.); (É.K.); (K.G.); (J.P.); (B.P.); (A.G.)
| | - Kamilla Gömöri
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, H-6720 Szeged, Hungary; (T.S.); (A.M.); (É.K.); (K.G.); (J.P.); (B.P.); (A.G.)
| | - Judit Pipis
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, H-6720 Szeged, Hungary; (T.S.); (A.M.); (É.K.); (K.G.); (J.P.); (B.P.); (A.G.)
- Pharmahungary Group, Hajnóczy u. 6, H-6722 Szeged, Hungary; (B.Á.); (Z.G.); (P.F.)
| | - Bence Pósa
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, H-6720 Szeged, Hungary; (T.S.); (A.M.); (É.K.); (K.G.); (J.P.); (B.P.); (A.G.)
| | - András Makkos
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary;
| | - Bence Ágg
- Pharmahungary Group, Hajnóczy u. 6, H-6722 Szeged, Hungary; (B.Á.); (Z.G.); (P.F.)
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary;
| | - Zoltán Giricz
- Pharmahungary Group, Hajnóczy u. 6, H-6722 Szeged, Hungary; (B.Á.); (Z.G.); (P.F.)
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary;
| | - Péter Ferdinandy
- Pharmahungary Group, Hajnóczy u. 6, H-6722 Szeged, Hungary; (B.Á.); (Z.G.); (P.F.)
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary;
| | - Anikó Görbe
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, H-6720 Szeged, Hungary; (T.S.); (A.M.); (É.K.); (K.G.); (J.P.); (B.P.); (A.G.)
- Pharmahungary Group, Hajnóczy u. 6, H-6722 Szeged, Hungary; (B.Á.); (Z.G.); (P.F.)
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary;
| | - Péter Bencsik
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, H-6720 Szeged, Hungary; (T.S.); (A.M.); (É.K.); (K.G.); (J.P.); (B.P.); (A.G.)
- Pharmahungary Group, Hajnóczy u. 6, H-6722 Szeged, Hungary; (B.Á.); (Z.G.); (P.F.)
| |
Collapse
|
17
|
Wang S, Zhang X, Hou Y, Zhang Y, Chen J, Gao S, Duan H, Gu S, Yu S, Cai Y. SIRT6 activates PPARα to improve doxorubicin-induced myocardial cell aging and damage. Chem Biol Interact 2024; 392:110920. [PMID: 38395252 DOI: 10.1016/j.cbi.2024.110920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 01/26/2024] [Accepted: 02/18/2024] [Indexed: 02/25/2024]
Abstract
The Sirtuins family, formally known as the Silent Information Regulator Factors, constitutes a highly conserved group of histone deacetylases. Recent studies have illuminated SIRT6's role in doxorubicin (DOX)-induced oxidative stress and apoptosis within myocardial cells. Nevertheless, the extent of SIRT6's impact on DOX-triggered myocardial cell aging and damage remains uncertain, with the associated mechanisms yet to be fully understood. In our research, we examined the influence of SIRT6 on DOX-induced cardiomyocyte senescence using β-galactosidase and γ-H2AX staining. Additionally, we gauged the mRNA expression of senescence-associated genes, namely p16, p21, and p53, through Real-time PCR. Employing ELISA assay kits, MDA, and total SOD activity assay kits, we measured inflammatory factors TNF-α, IL-6, and IL-1β, alongside oxidative stress-related indicators. The results unequivocally indicated that SIRT6 overexpression robustly inhibited DOX-induced cardiomyocyte senescence. Furthermore, we established that SIRT6 overexpression suppressed the inflammatory response and oxidative stress induced by DOX in cardiomyocytes. Conversely, silencing SIRT6 exacerbated DOX-induced cardiomyocyte injury. Our investigations further unveiled that SIRT6 upregulated the expression of genes CD36, CPT1, LCAD, MCAD associated with fatty acid oxidation through its interaction with PPARα, thereby exerting anti-aging effects. In vivo, the overexpression of SIRT6 was observed to restore DOX-induced declines in EF and FS to normal levels in mice. Echocardiography and HE staining revealed the restoration of cardiomyocyte alignment, affording protection against DOX-induced myocardial senescence and injury. The findings from this study suggest that SIRT6 holds significant promise as a therapeutic target for mitigating DOX-induced cardiomyopathy.
Collapse
Affiliation(s)
- Shulin Wang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xuan Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Afffliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yanhong Hou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Afffliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yuliang Zhang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiamin Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Afffliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shuhan Gao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Afffliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Huiying Duan
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Afffliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shaoju Gu
- Laboratory Animal Centre, Guangzhou Medical University, Guangzhou, China.
| | - Shanshan Yu
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| | - Yi Cai
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, China; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Afffliated Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
18
|
Gocer Z, Elek A, Caska H, Bozgeyik I. MicroRNAs and cardiac fibrosis: A comprehensive update on mechanisms and consequences. Pathol Res Pract 2023; 251:154853. [PMID: 37857035 DOI: 10.1016/j.prp.2023.154853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/30/2023] [Accepted: 10/02/2023] [Indexed: 10/21/2023]
Abstract
Fibrosis is a pathological wound-healing mechanism that results by the overactivation of fibroblasts. Fibrosis can become obstructive and deleterious during regeneration of various body tissues including cardiac muscle. This ultimately results in the development of cardiac fibrosis, characterized by an excessive buildup of extracellular matrix proteins. Thus, it could lead to arrhythmias and heart failure which creates a leading public health burden worldwide. MiRNAs are small non-coding RNAs with great potential for diagnostic and therapeutic purposes. Mounting evidence indicates that miRNAs are involved in the deregulation of tissue homeostasis during myocardial fibrosis. For instance, miRNAs that are implicated in the regulation of TGF-beta signaling pathway have been reported to be significantly altered in myocardial fibrosis. Accordingly, in this comprehensive review, we discuss and highlight recent available data on the role of miRNAs during myocardial fibrosis, providing valuable insights into the miRNA modulation of cardiac fibrosis and miRNAs targets that can be used in the future therapeutic interventions to cardiac fibrosis.
Collapse
Affiliation(s)
- Zekihan Gocer
- Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Alperen Elek
- Faculty of Medicine, Ege University, Izmir, Turkey
| | - Halil Caska
- Department of Medical Biology and Genetics, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Ibrahim Bozgeyik
- Department of Medical Biology, Faculty of Medicine, Adiyaman University, Adiyaman, Turkey.
| |
Collapse
|
19
|
Sangweni NF, Gabuza K, van Aarde R, Mabasa L, van Vuuren D, Huisamen B, Barry R, Johnson R. Doxorubicin-Induced Cardiomyopathy: A Preliminary Study on the Cardioprotective Benefits of 7-Hydroxyflavanone. Int J Mol Sci 2023; 24:15395. [PMID: 37895075 PMCID: PMC10607478 DOI: 10.3390/ijms242015395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/14/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
The therapeutic properties of flavonoids are reported to offer cardioprotective benefits against doxorubicin (Dox)-induced cardiotoxicity (DIC). In the current study, we aimed to investigate the prophylactic properties of 7-hydroxyflavanone (7H), a flavonoid with antioxidative properties, against DIC. An in vitro model of DIC was established by exposing H9c2 cardiomyoblasts to Dox for 6 days. Similarly, cells were also co-treated with 7H to assess its ability to mitigate DIC. The data obtained indicate that 7H, as a co-treatment, alleviates Dox-induced oxidative stress by enhancing total glutathione content (p ≤ 0.001) and superoxide dismutase activity (p ≤ 0.001) whilst decreasing ROS (p ≤ 0.001), malondialdehyde production (p ≤ 0.001) and the secretion of interleukin-6 (p ≤ 0.001). The data also showed an improvement in mitochondrial function as shown via enhanced bioenergetics, mitochondrial membrane potential, and PGC1-alpha (p ≤ 0.05) and pAMPK (p ≤ 0.001) expression. The cardioprotective potential of 7H was further highlighted by its ability attenuate Dox-induced caspase 3/7 activity (p ≤ 0.001), apoptosis (p ≤ 0.001) and necrosis (p ≤ 0.05). In conclusion, our findings demonstrated the cardioprotective benefits of 7H and thus suggests that it could be a suitable candidate cardioprotective agent against DIC.
Collapse
Affiliation(s)
- Nonhlakanipho F. Sangweni
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, Tygerberg, Cape Town 7505, South Africa; (K.G.); (R.v.A.); (L.M.); (R.J.)
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa; (D.v.V.); (B.H.)
| | - Kwazi Gabuza
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, Tygerberg, Cape Town 7505, South Africa; (K.G.); (R.v.A.); (L.M.); (R.J.)
| | - Ruzayda van Aarde
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, Tygerberg, Cape Town 7505, South Africa; (K.G.); (R.v.A.); (L.M.); (R.J.)
| | - Lawrence Mabasa
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, Tygerberg, Cape Town 7505, South Africa; (K.G.); (R.v.A.); (L.M.); (R.J.)
| | - Derick van Vuuren
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa; (D.v.V.); (B.H.)
| | - Barbara Huisamen
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa; (D.v.V.); (B.H.)
| | | | - Rabia Johnson
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, Tygerberg, Cape Town 7505, South Africa; (K.G.); (R.v.A.); (L.M.); (R.J.)
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa; (D.v.V.); (B.H.)
| |
Collapse
|
20
|
Chen W, Ruan M, Zou M, Liu F, Liu H. Clinical Significance of Non-Coding RNA Regulation of Programmed Cell Death in Hepatocellular Carcinoma. Cancers (Basel) 2023; 15:4187. [PMID: 37627215 PMCID: PMC10452865 DOI: 10.3390/cancers15164187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/02/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a widely prevalent and malignantly progressive tumor. Most patients are typically diagnosed with HCC at an advanced stage, posing significant challenges in the execution of curative surgical interventions. Non-coding RNAs (ncRNAs) represent a distinct category of RNA molecules not directly involved in protein synthesis. However, they possess the remarkable ability to regulate gene expression, thereby exerting significant regulatory control over cellular processes. Notably, ncRNAs have been implicated in the modulation of programmed cell death (PCD), a crucial mechanism that various therapeutic agents target in the fight against HCC. This review summarizes the clinical significance of ncRNA regulation of PCD in HCC, including patient diagnosis, prognosis, drug resistance, and side effects. The aim of this study is to provide new insights and directions for the diagnosis and drug treatment strategies of HCC.
Collapse
Affiliation(s)
| | | | | | - Fuchen Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai 200438, China; (W.C.); (M.R.)
| | - Hui Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai 200438, China; (W.C.); (M.R.)
| |
Collapse
|
21
|
Prasad Panda S, Kesharwani A, Prasanna Mallick S, Prasanth D, Kumar Pasala P, Bharadwaj Tatipamula V. Viral-induced neuronal necroptosis: Detrimental to brain function and regulation by necroptosis inhibitors. Biochem Pharmacol 2023; 213:115591. [PMID: 37196683 DOI: 10.1016/j.bcp.2023.115591] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 05/19/2023]
Abstract
Neuronal necroptosis (programmed necrosis) in the CNS naturally occurs through a caspase-independent way and, especially in neurodegenerative diseases (NDDs) such as Alzheimer's disease (AD), Parknson's disease (PD), Amyotrophic Lateral Sclerosis (ALS) and viral infections. Understanding necroptosis pathways (death receptor-dependent and independent), and its connections with other cell death pathways could lead to new insights into treatment. Receptor-interacting protein kinase (RIPK) mediates necroptosis via mixed-lineage kinase-like (MLKL) proteins. RIPK/MLKL necrosome contains FADD, procaspase-8-cellular FLICE-inhibitory proteins (cFLIPs), RIPK1/RIPK3, and MLKL. The necrotic stimuli cause phosphorylation of MLKL and translocate to the plasma membrane, causing an influx of Ca2+ and Na+ ions and, the immediate opening of mitochondrial permeability transition pore (mPTP) with the release of inflammatory cell damage-associated molecular patterns (DAMPs) like mitochondrial DNA (mtDNA), high-mobility group box1 (HMGB1), and interleukin1 (IL-1). The MLKL translocates to the nucleus to induce transcription of the NLRP3 inflammasome complex elements. MLKL-induced NLRP3 activity causes caspase-1 cleavage and, IL-1 activation which promotes neuroinflammation. RIPK1-dependent transcription increases illness-associated microglial and lysosomal abnormalities to facilitate amyloid plaque (Aβ) aggregation in AD. Recent research has linked neuroinflammation and mitochondrial fission with necroptosis. MicroRNAs (miRs) such as miR512-3p, miR874, miR499, miR155, and miR128a regulate neuronal necroptosis by targeting key components of necroptotic pathways. Necroptosis inhibitors act by inhibiting the membrane translocation of MLKL and RIPK1 activity. This review insights into the RIPK/MLKL necrosome-NLRP3 inflammasome interactions during death receptor-dependent and independent neuronal necroptosis, and clinical intervention by miRs to protect the brain from NDDs.
Collapse
Affiliation(s)
- Siva Prasad Panda
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India.
| | - Adarsh Kesharwani
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Sarada Prasanna Mallick
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation, Guntur, Andhrapradesh, India
| | - Dsnbk Prasanth
- Department of Pharmacognosy, KVSR Siddhartha College of Pharmaceutical Sciences, Vijayawada, AP, India
| | | | - Vinay Bharadwaj Tatipamula
- Center for Molecular Biology, College of Medicine and Pharmacy, Duy Tan University, Danang 550000, Viet Nam
| |
Collapse
|
22
|
Bradic J, Andjic M, Novakovic J, Kocovic A, Tomovic M, Petrovic A, Nikolic M, Mitrovic S, Jakovljevic V, Pecarski D. Lady's Bedstraw as a Powerful Antioxidant for Attenuation of Doxorubicin-Induced Cardiotoxicity. Antioxidants (Basel) 2023; 12:1277. [PMID: 37372007 DOI: 10.3390/antiox12061277] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/02/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
This study aimed to examine the effects of a 14-day treatment with lady's bedstraw methanol extract on doxorubicin-induced cardiotoxicity through functional, biochemical and histological examinations. We used 24 male Wistar albino rats divided into the following groups: control (CTRL), doxorubicin (DOX), and DOX + GVE (Galium verum extract). GVE was administered orally at a dose of 50 mg/kg per day for 14 days, while a single dose of doxorubicin was injected into the DOX groups. After accomplishing treatment with GVE, cardiac function was assessed, which determined the redox state. During the autoregulation protocol on the Langendorff apparatus, ex vivo cardiodynamic parameters were measured. Our results demonstrated that the consumption of GVE effectively suppressed the disturbed response of the heart to changes in perfusion pressures caused by administration of DOX. Intake of GVE was associated with a reduction in most of the measured prooxidants in comparison to the DOX group. Moreover, this extract was capable of increasing the activity of the antioxidant defense system. Morphometric analyses showed that rat hearts treated with DOX showed more pronounced degenerative changes and necrosis compared to the CTRL group. However, GVE pretreatment seems to be able to prevent the pathological injuries caused by DOX injection via decrease in oxidative stress and apoptosis.
Collapse
Affiliation(s)
- Jovana Bradic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, Svetozara Makovica 69, 34000 Kragujevac, Serbia
| | - Marijana Andjic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, Svetozara Makovica 69, 34000 Kragujevac, Serbia
| | - Jovana Novakovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, Svetozara Makovica 69, 34000 Kragujevac, Serbia
| | - Aleksandar Kocovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, Svetozara Makovica 69, 34000 Kragujevac, Serbia
| | - Marina Tomovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Anica Petrovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, Svetozara Makovica 69, 34000 Kragujevac, Serbia
| | - Marina Nikolic
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, Svetozara Makovica 69, 34000 Kragujevac, Serbia
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Slobodanka Mitrovic
- Department of Pathology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Vladimir Jakovljevic
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, Svetozara Makovica 69, 34000 Kragujevac, Serbia
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Department of Human Pathology, 1st Moscow State Medical, University IM Sechenov, Moscow 119991, Russia
| | - Danijela Pecarski
- The College of Health Science, Academy of Applied Studies Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
23
|
Li C, Guo Z, Liu F, An P, Wang M, Yang D, Tang Q. PCSK6 attenuates cardiac dysfunction in doxorubicin-induced cardiotoxicity by regulating autophagy. Free Radic Biol Med 2023; 203:114-128. [PMID: 37061139 DOI: 10.1016/j.freeradbiomed.2023.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 04/07/2023] [Accepted: 04/08/2023] [Indexed: 04/17/2023]
Abstract
Doxorubicin (DOX) is a chemotherapeutic drug widely used in the field of cancer, but its side effects on the heart hinder its clinical application. In cardiac injury caused by DOX, apoptosis and oxidative stress are both involved in cardiac damage, and autophagy is also one of the key responses. Both apoptosis and oxidative stress interact with autophagy. Proper promotion of autophagy effectively protects the myocardium and blocks cardiac injury. DOX mainly acts downstream of the autophagic flow and hinders the degradation process of autophagolysosomes, resulting in abnormal accumulation of autophagolysosomes in cells, which can prevent the timely removal of harmful substances and disrupt the normal function of cells. Proprotein convertase subtilisin/kexin type 6 (PCSK6) is involved in the occurrence and development of various cardiovascular diseases, blood pressure regulation and the inflammatory response, but its role in DOX is still unclear. Here, we constructed cardiac PCSK6-overexpressing mice by injecting AAV9-PCSK6. Both in vivo and in vitro experiments confirmed that overexpression of PCSK6 effectively protected cardiac function, inhibited apoptosis and oxidative stress. We focused on the effect of PCSK6 overexpression on autophagy. We have detected an increase in autophagosomes production and a decrease in autophagolysosomes accumulation. This suggests that PCSK6 promotes the level of autophagy, while possibly acting on the sites where DOX inhibits degradation, so that the autophagic flux inhibited by DOX is restored and the degradation process of autophagolysosomes is restored. The effect of PCSK6 was dependent on FOXO3a, which promoted the nuclear translocation of Forkhead box O3 (FOXO3a), and Sirtuin 1 (SIRT1) regulated the expression of FOXO3a. When SIRT1 was inhibited, the protective effect of PCSK6 was diminished. In conclusion, overexpression of PCSK6 exerts a protective effect through SIRT1/FOXO3a in cardiac injury induced by DOX, suggesting that PCSK6 may be a therapeutic target for DOX cardiomyopathy.
Collapse
Affiliation(s)
- Chenfei Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China.
| | - Zhen Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China.
| | - Fangyuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Peng An
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Mingyu Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Dan Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China.
| |
Collapse
|
24
|
Kuang Z, Wu J, Tan Y, Zhu G, Li J, Wu M. MicroRNA in the Diagnosis and Treatment of Doxorubicin-Induced Cardiotoxicity. Biomolecules 2023; 13:biom13030568. [PMID: 36979503 PMCID: PMC10046787 DOI: 10.3390/biom13030568] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/12/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Doxorubicin (DOX), a broad-spectrum chemotherapy drug, is widely applied to the treatment of cancer; however, DOX-induced cardiotoxicity (DIC) limits its clinical therapeutic utility. However, it is difficult to monitor and detect DIC at an early stage using conventional detection methods. Thus, sensitive, accurate, and specific methods of diagnosis and treatment are important in clinical practice. MicroRNAs (miRNAs) belong to non-coding RNAs (ncRNAs) and are stable and easy to detect. Moreover, miRNAs are expected to become biomarkers and therapeutic targets for DIC; thus, there are currently many studies focusing on the role of miRNAs in DIC. In this review, we list the prominent studies on the diagnosis and treatment of miRNAs in DIC, explore the feasibility and difficulties of using miRNAs as diagnostic biomarkers and therapeutic targets, and provide recommendations for future research.
Collapse
Affiliation(s)
- Ziyu Kuang
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jingyuan Wu
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Ying Tan
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Guanghui Zhu
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jie Li
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Min Wu
- Cardiovascular Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| |
Collapse
|
25
|
Liu D, Wang Q, Yuan W, Wang Q. Irigenin attenuates lipopolysaccharide-induced acute lung injury by inactivating the mitogen-activated protein kinase (MAPK) signaling pathway. Hum Exp Toxicol 2023; 42:9603271231155098. [PMID: 36738242 DOI: 10.1177/09603271231155098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Acute lung injury (ALI) is a serious pulmonary inflammation disease with high mortality. Irigenin, an isoflavone from rhizomes of the Belamcanda chinensis, has been reported to exert anti-inflammatory, anti-oxidative, and anti-apoptotic activities in several diseases. However, it is still unclear whether irigenin can exert a beneficial effect in ALI. A network pharmacology method was utilized to predict the hub targets and potential therapeutic mechanisms of irigenin against ALI. Lipopolysaccharide (LPS) was used to establish the mice model of ALI for evaluating the effects of irigenin. According to the protein-protein interaction (PPI) network, we identified EGFR, HRAS, AKT1, SRC, and HSP90AA1 as the top five significant genes. Gene Ontology (GO) and Kyoto Encyclopaedia of Genes and Genomes (KEGG) enrichment assays showed that irigenin might affect inflammatory response, cytokine production, and cell death by the mitogen-activated protein kinase (MAPK) signaling pathway. In vivo experiment results manifested that irigenin decreased pathological changes, lung Wet/Dry weight ratio, and total protein content in bronchoalveolar lavage fluid (BALF). Irigenin also reduced the production of inflammatory cytokines, including tumor necrosis factor-a (TNF-a), interleukin-1β (IL-1β), interleukin-6 (IL-6), and interleukin-18 (IL-18), and neutrophil infiltration. Additionally, irigenin inhibited pulmonary apoptosis in LPS-treated ALI mice. Moreover, LPS-induced phosphorylation of p38, JNK, and ERK was significantly abated due to the treatment of irigenin. In summary, irigenin ameliorates LPS-induced ALI by suppressing pulmonary inflammation and apoptosis via inactivation of the MAPK signaling pathway. These findings indicated the therapeutic potential of irigenin in ALI.
Collapse
Affiliation(s)
- Dan Liu
- Department of Pharmacy, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Qing Wang
- Department of Pharmacy, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Wen Yuan
- Department of Pharmacy, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Qiang Wang
- Department of Pharmacy, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| |
Collapse
|
26
|
Saetang J, Tedasen A, Sangkhathat S, Sangkaew N, Dokduang S, Prompat N, Taraporn S, Graidist P. The attenuation effect of low piperine Piper nigrum extract on doxorubicin-induced toxicity of blood chemical and immunological properties in mammary tumour rats. PHARMACEUTICAL BIOLOGY 2022; 60:96-107. [PMID: 34962450 PMCID: PMC8735876 DOI: 10.1080/13880209.2021.2018470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 10/26/2021] [Accepted: 12/10/2021] [Indexed: 06/01/2023]
Abstract
CONTEXT Many natural extracts have been shown to minimize the toxicity of doxorubicin (Dox). Low piperine Piper nigrum L. (Piperaceae) extract (PFPE) is a natural extract containing many types of antioxidants that may reduce Dox toxicities. OBJECTIVE To evaluate the effect of PFPE in attenuating the side effects of Dox. MATERIALS AND METHODS Tumour-bearing Sprague Dawley rats were divided into five groups including normal, vehicle, 100 mg/kg BW of PFPE plus 2 mg/kg BW of Dox (P100 + Dox), 100 mg/kg BW of PFPE plus 2 mg/kg BW of Dox (P200 + Dox) and Dox. Rats were treated with Dox and/or PFPE three times/week for 4 weeks. Tumour burden, blood parameters, weight of internal organs and immunological data were investigated. RESULTS The addition of 200 mg/kg PFPE significantly restored the levels of AST from 174.60 ± 45.67 U/L in the Dox group near to normal levels at 109.80 ± 4.99 U/L. The combination of PFPE and Dox also decreased the levels of CXCL7, TIMP-1, sICAM-1 and l-selectin about 1.4-1.6-fold compared to Dox group. Feeding rats with 200 mg/kg BW of PFPE combination with Dox slightly increased Th1 from 161.67 ± 14.28 cells in Dox group to 200.75 ± 5.8 cells meanwhile suppressed Treg from 3088 ± 78 cells in Dox to 2561 ± 71 cells. DISCUSSION AND CONCLUSIONS This study showed that PFPE ameliorated Dox toxicity in many aspects indicating the role of antioxidant and other substances in the extract on toxicity attenuation. This suggested the using of PFPE may be valuable for Dox treated patients.
Collapse
Affiliation(s)
- Jirakrit Saetang
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
- Department of Surgery, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
- EZ-Mol-Design Laboratory, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| | - Aman Tedasen
- Department of Medical Technology, School of Allied Health Sciences and Public Health, Walailak University, Thai Buri, Thailand
| | - Surasak Sangkhathat
- Department of Surgery, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| | - Natnaree Sangkaew
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| | - Sirinapa Dokduang
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| | - Napat Prompat
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| | - Siriporn Taraporn
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| | - Potchanapond Graidist
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| |
Collapse
|
27
|
Xu J, Sun S, Zhang W, Dong J, Huang C, Wang X, Jia M, Yang H, Wang Y, Jiang Y, Cao L, Huang Z. Irigenin inhibits glioblastoma progression through suppressing YAP/β-catenin signaling. Front Pharmacol 2022; 13:1027577. [PMID: 36532767 PMCID: PMC9748621 DOI: 10.3389/fphar.2022.1027577] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/16/2022] [Indexed: 09/14/2024] Open
Abstract
Glioblastoma (GBM) is the most malignant glioma in brain tumors with low survival and high recurrence rate. Irigenin, as an isoflavone compound extracted from Shegan, has shown many pharmacological functions such as antioxidant, anti-inflammatory and anti-tumor. However, the effects of irigenin on GBM cells and the related molecular mechanisms remain unexplored. In this study, we found that irigenin inhibited the proliferation of GBM cells in a dose-dependent manner by several assays in vitro. Subsequently, we found that irigenin arrested cell cycle at G2/M phase and induced apoptosis of GBM cells in vitro. In addition, irigenin inhibited the migration of GBM cells. Mechanically, we found that irigenin treatment decreased the expression of YAP (yes-associated protein), suppressed β-catenin signaling. Furthermore, overexpression of YAP partially restored the anti-tumor effects of irigenin on GBM cells in vitro. Finally, we found that irigenin inhibited the growth of tumor in GBM xenograft mice model through inactivation of YAP. Taken together, these results suggest that irigenin exerts its anticancer effects on GBM via inhibiting YAP/β-catenin signaling, which may provide a new strategy for the treatment of GBM.
Collapse
Affiliation(s)
- Jiayun Xu
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Shanshan Sun
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Wei Zhang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jianhong Dong
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Changgang Huang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xin Wang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Mengxian Jia
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Hao Yang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yongjie Wang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yuanyuan Jiang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Liying Cao
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Laboratory of Aging and Cancer Biology of Zhejiang Province, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Zhihui Huang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
28
|
Al-Kuraishy HM, Al-Gareeb AI, Alkhuriji AF, Al-Megrin WAI, Elekhnawy E, Negm WA, De Waard M, Batiha GES. Investigation of the impact of rosuvastatin and telmisartan in doxorubicin-induced acute cardiotoxicity. Biomed Pharmacother 2022; 154:113673. [PMID: 36942604 DOI: 10.1016/j.biopha.2022.113673] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/22/2022] [Accepted: 09/05/2022] [Indexed: 12/06/2022] Open
Abstract
Cardiac injury is the main dose-limiting factor for doxorubicin (Dox) use as an anticancer agent. The cardiotoxicity of Dox is linked to a number of complex mechanisms, including oxidative stress, mitochondrial damage, intracellular calcium dysregulation, and apoptosis/necrosis. This study investigates several aspects of Dox-induced cardiotoxicity. We investigated the effects of pre-treatment with rosuvastatin and telmisartan, which were used in different doses alone or combination, on the acute cardiotoxicity induced by Dox. The results of this study showed that Dox induced significant pathological changes in the cardiomyocytes. Adverse effects were observed on several biomarkers related to cardiac damage like cardiac troponin I (cTnI) and lactate dehydrogenase (LDH), oxidative stress like malondialdehyde (MDA), an inflammatory process like interleukin-17 (IL-17) with important histopathological changes. We illusterate the cardio-protective contribution of the two pharmacological agents against the acute cardiotoxic effects of Dox. This is manifested by the significant improvement in the biomarker levels and the associated histological damage. This study points out the beneficial use of both rosuvastatin and telmisartan alone or in combination as a clinical option for decreasing the acute toxicity of Dox on cardiomyocytes.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-mustansiriyiah University, Iraq.
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-mustansiriyiah University, Iraq.
| | - Afrah Fahad Alkhuriji
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia.
| | - Wafa Abdullah I Al-Megrin
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia.
| | - Engy Elekhnawy
- Pharmaceutical Microbiology Department, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Walaa A Negm
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Michel De Waard
- Smartox Biotechnology, 6 rue des Platanes, 38120 Saint-Egrève, France; L'institut du Thorax, INSERM, CNRS, UNIV NANTES, F-44007 Nantes, France; LabEx Ion Channels, Science & Therapeutics, Université de Nice Sophia-Antipolis, F-06560 Valbonne, France.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AL Beheira, Egypt.
| |
Collapse
|
29
|
Feng P, Yang Y, Liu N, Wang S. Baicalin regulates TLR4/IκBα/NFκB signaling pathway to alleviate inflammation in Doxorubicin related cardiotoxicity. Biochem Biophys Res Commun 2022; 637:1-8. [DOI: 10.1016/j.bbrc.2022.10.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/05/2022] [Accepted: 10/17/2022] [Indexed: 11/28/2022]
|
30
|
Oo TT, Pratchayasakul W, Chattipakorn N, Chattipakorn SC. Emerging roles of toll-like receptor 4 in chemotherapy-induced neurotoxicity. Neurotoxicology 2022; 93:112-127. [PMID: 36152729 DOI: 10.1016/j.neuro.2022.09.006] [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: 07/08/2022] [Revised: 09/12/2022] [Accepted: 09/19/2022] [Indexed: 10/14/2022]
Abstract
Chemotherapy-induced neurotoxicity is one of the most prevalent side effects in cancer patients and survivors. Cognitive decline and peripheral neuropathy are the most common chemotherapy-induced neurotoxic symptoms. These symptoms lead not only to the limiting of the dose of chemotherapy given to cancer patients, but also have an impact on the quality of life of cancer survivors. Although the exact mechanisms involved in chemotherapy-induced neurotoxicity are still unclear, neuroinflammation is widely regarded as being one of the major causes involved in chemotherapy-induced neurotoxicity. It is known that Toll-like receptor 4 (TLR4) plays a critical role in the inflammatory process, and it has been recently reported that it is associated with chemotherapy-induced neurotoxicity. In this review, we summarize and discuss all available evidence regarding the activation of the TLR4 signaling pathway in various models of chemotherapy-induced neurotoxicity. This review also emphasizes the evidence pertinent to TLR4 inhibition on chemotherapy-induced neurotoxicity in rodent studies. Understanding the role of the TLR4 signaling pathway behind chemotherapy-induced neurotoxicity is crucial for improving treatments and ensuring the long-term survival of cancer patients.
Collapse
Affiliation(s)
- Thura Tun Oo
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand
| | - Wasana Pratchayasakul
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
31
|
Sun W, Xu J, Wang L, Jiang Y, Cui J, Su X, Yang F, Tian L, Si Z, Xing Y. Non-coding RNAs in cancer therapy-induced cardiotoxicity: Mechanisms, biomarkers, and treatments. Front Cardiovasc Med 2022; 9:946137. [PMID: 36082126 PMCID: PMC9445363 DOI: 10.3389/fcvm.2022.946137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/28/2022] [Indexed: 02/06/2023] Open
Abstract
As a result of ongoing breakthroughs in cancer therapy, cancer patients' survival rates have grown considerably. However, cardiotoxicity has emerged as the most dangerous toxic side effect of cancer treatment, negatively impacting cancer patients' prognosis. In recent years, the link between non-coding RNAs (ncRNAs) and cancer therapy-induced cardiotoxicity has received much attention and investigation. NcRNAs are non-protein-coding RNAs that impact gene expression post-transcriptionally. They include microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs). In several cancer treatments, such as chemotherapy, radiotherapy, and targeted therapy-induced cardiotoxicity, ncRNAs play a significant role in the onset and progression of cardiotoxicity. This review focuses on the mechanisms of ncRNAs in cancer therapy-induced cardiotoxicity, including apoptosis, mitochondrial damage, oxidative stress, DNA damage, inflammation, autophagy, aging, calcium homeostasis, vascular homeostasis, and fibrosis. In addition, this review explores potential ncRNAs-based biomarkers and therapeutic strategies, which may help to convert ncRNAs research into clinical practice in the future for early detection and improvement of cancer therapy-induced cardiotoxicity.
Collapse
Affiliation(s)
- Wanli Sun
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Juping Xu
- The Second People's Hospital of Jiaozuo, Jiaozuo, China
| | - Li Wang
- Department of Breast Surgery, Xingtai People's Hospital, Xingtai, China
| | - Yuchen Jiang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jingrun Cui
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xin Su
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fan Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Tian
- Beijing University of Chinese Medicine, Beijing, China
| | - Zeyu Si
- The First Clinical Medical College of Shaanxi University of Chinese Medicine, Taiyuan, China
- Zeyu Si
| | - Yanwei Xing
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yanwei Xing
| |
Collapse
|
32
|
Sangweni NF, van Vuuren D, Mabasa L, Gabuza K, Huisamen B, Naidoo S, Barry R, Johnson R. Prevention of Anthracycline-Induced Cardiotoxicity: The Good and Bad of Current and Alternative Therapies. Front Cardiovasc Med 2022; 9:907266. [PMID: 35811736 PMCID: PMC9257015 DOI: 10.3389/fcvm.2022.907266] [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: 03/29/2022] [Accepted: 05/26/2022] [Indexed: 12/20/2022] Open
Abstract
Doxorubicin (Dox)-induced cardiotoxicity (DIC) remains a serious health burden, especially in developing countries. Unfortunately, the high cost of current preventative strategies has marginalized numerous cancer patients because of socio-economic factors. In addition, the efficacy of these strategies, without reducing the chemotherapeutic properties of Dox, is frequently questioned. These limitations have widened the gap and necessity for alternative medicines, like flavonoids, to be investigated. However, new therapeutics may also present their own shortcomings, ruling out the idea of “natural is safe”. The U.S. Food and Drug Administration (FDA) has stipulated that the concept of drug-safety be considered in all pre-clinical and clinical studies, to explore the pharmacokinetics and potential interactions of the drugs being investigated. As such our studies on flavonoids, as cardio-protectants against DIC, have been centered around cardiac and cancer models, to ensure that the efficacy of Dox is preserved. Our findings thus far suggest that flavonoids of Galenia africana could be suitable candidates for the prevention of DIC. However, this still requires further investigation, which would focus on drug-interactions as well as in vivo experimental models to determine the extent of cardioprotection.
Collapse
Affiliation(s)
- Nonhlakanipho F Sangweni
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa.,Division of Medical Physiology, Faculty of Medicine and Health Sciences, Centre for Cardio-metabolic Research in Africa, Stellenbosch University, Stellenbosch, South Africa
| | - Derick van Vuuren
- Division of Medical Physiology, Faculty of Medicine and Health Sciences, Centre for Cardio-metabolic Research in Africa, Stellenbosch University, Stellenbosch, South Africa
| | - Lawrence Mabasa
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa
| | - Kwazi Gabuza
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa
| | - Barbara Huisamen
- Division of Medical Physiology, Faculty of Medicine and Health Sciences, Centre for Cardio-metabolic Research in Africa, Stellenbosch University, Stellenbosch, South Africa
| | - Sharnay Naidoo
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa.,Division of Medical Physiology, Faculty of Medicine and Health Sciences, Centre for Cardio-metabolic Research in Africa, Stellenbosch University, Stellenbosch, South Africa
| | - Reenen Barry
- Research and Development Department, BioPharm, Hamilton, New Zealand
| | - Rabia Johnson
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa.,Division of Medical Physiology, Faculty of Medicine and Health Sciences, Centre for Cardio-metabolic Research in Africa, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
33
|
Abdel-Baki PM, El-Sherei MM, Khaleel AE, Abdel-Aziz MM, Okba MM. Irigenin, a novel lead from Iris confusa for management of Helicobacter pylori infection with selective COX-2 and HpIMPDH inhibitory potential. Sci Rep 2022; 12:11457. [PMID: 35794127 PMCID: PMC9259591 DOI: 10.1038/s41598-022-15361-w] [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: 02/13/2022] [Accepted: 06/20/2022] [Indexed: 12/17/2022] Open
Abstract
The development of new natural drugs for Helicobacter pylori (H. pylori) management has recently received significant attention. Iris confusa (I. confusa) was long used for the treatment of bacterial infections and gastritis. This study aimed at evaluating its effect on management of H. pylori infection and exploring its bioactive metabolites. The inhibitory potential of the polar (PF), non-polar (NPF) fractions and the isolated compounds against H. pylori using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay in addition to their cyclooxygenases (COX-1 and COX-2), and nitric oxide (NO) inhibitory activities were assessed. The most biologically active compound was tested for its selective H. pylori inosine-5'-monophosphate dehydrogenase (HpIMPDH) inhibitory potential. Chromatographic purification of PF and NPF allowed isolation of tectoridin, orientin, irigenin, tectorigenin, isoarborinol and stigmasterol. The PF exhibited significant anti-H. pylori (MIC 62.50 µg/mL), COX-1, COX-2 (IC50 of 112.08 ± 0.60 and 47.90 ± 1.50 µg/mL respectively, selectivity index SI of 2.34), and NO (IC50 47.80 ± 0.89 µg/mL) inhibitory activities, while irigenin was the most potent isolated compound. Irigenin was found to have a promising activity against HpIMPDH enzyme (IC50 of 2.07 ± 1.90 μM) with low activity against human hIMPDH2 (IC50 > 10 μM) than clarithromycin, assuring its selectivity. Overall, I. confusa and its isolated compounds may serve as a potential source of plant-based drugs for H. pylori control. This study scientifically validated the claimed anti-bacterial activity of I. confusa and revealed irigenin potential as a novel lead exhibiting anti H. pylori activity in a first record.
Collapse
Affiliation(s)
- Passent M Abdel-Baki
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Moshera M El-Sherei
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Amal E Khaleel
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Marwa M Abdel-Aziz
- Regional Center for Mycology and Biotechnology (RCMB), Al-Azhar University, Cairo, 11651, Egypt
| | - Mona M Okba
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| |
Collapse
|
34
|
Li J, Xu Z, Gu J. UGT1A1 and UGT1A9 Are Responsible for Phase II Metabolism of Tectorigenin and Irigenin In Vitro. Molecules 2022; 27:molecules27134104. [PMID: 35807350 PMCID: PMC9268515 DOI: 10.3390/molecules27134104] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 06/07/2022] [Accepted: 06/17/2022] [Indexed: 12/04/2022] Open
Abstract
Tectorigenin and irigenin are biologically active isoflavones of Belamcanda chinensis (L.) DC. Previous studies indicated that both compounds could be metabolized in vivo; however, the kinetic parameters of enzymes involved in the metabolization of tectorigenin and irigenin have not been identified. The aim of this study was to investigate UGTs involved in the glucuronidation of tectorigenin and irigenin and determine enzyme kinetic parameters using pooled human liver microsomes (HLMs) and recombinant UGTs. Glucuronides of tectorigenin and irigenin were identified using high-performance liquid chromatography (HPLC) coupled with mass spectrometry and quantified by HPLC using a response factor method. The results showed that tectorigenin and irigenin were modified by glucuronidation in HLMs. One metabolite of tectorigenin (M) and two metabolites of irigenin (M1 and M2) were detected. Chemical inhibition and recombinant enzyme experiments revealed that several enzymes could catalyze tectorigenin and irigenin glucuronidation. Among them, UGT1A1 and UGT1A9 were the primary enzymes for both tectorigenin and irigenin; however, the former mostly produced irigenin glucuronide M1, while the latter mostly produced irigenin glucuronide M2. These findings suggest that UGT1A1 and UGT1A9 were the primary isoforms metabolizing tectorigenin and irigenin in HLMs, which could be involved in drug–drug interactions and, therefore, should be monitored in clinical practice.
Collapse
Affiliation(s)
- Ji Li
- Department of Radiation Oncology, Eye and ENT Hospital, Fudan University, Shanghai 200031, China;
| | - Zhangyao Xu
- Department of Pharmacy, Eye and ENT Hospital, Fudan University, Shanghai 200031, China;
| | - Jifeng Gu
- Department of Pharmacy, Eye and ENT Hospital, Fudan University, Shanghai 200031, China;
- Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, Fudan University, Shanghai 200031, China
- Correspondence:
| |
Collapse
|
35
|
Zhang R, Jiang Y, Hao L, Yang Y, Gao Y, Zhang N, Zhang X, Song Y. CD44/Folate Dual Targeting Receptor Reductive Response PLGA-Based Micelles for Cancer Therapy. Front Pharmacol 2022; 13:829590. [PMID: 35359873 PMCID: PMC8960309 DOI: 10.3389/fphar.2022.829590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/10/2022] [Indexed: 11/13/2022] Open
Abstract
In this study, a novel poly (lactic-co-glycolic acid) (PLGA)-based micelle was synthesized, which could improve the therapeutic effect of the antitumor drug doxorubicin hydrochloride (DOX) and reduce its toxic and side effects. The efficient delivery of DOX was achieved by active targeting mediated by double receptors and stimulating the reduction potential in tumor cells. FA-HA-SS-PLGA polymer was synthesized by amidation reaction, and then DOX-loaded micelles were prepared by dialysis method. The corresponding surface method was used to optimize the experimental design. DOX/FA-HA-SS-PLGA micelles with high drug loading rate and encapsulation efficiency were prepared. The results of hydrophilic experiment, critical micelle concentration determination, and hemolysis test all showed that DOX/FA-HA-SS-PLGA micelles had good physicochemical properties and biocompatibility. In addition, both in vitro reduction stimulus response experiment and in vitro release experiment showed that DOX/FA-HA-SS-PLGA micelles had reduction sensitivity. Molecular docking experiments showed that it can bind to the target protein. More importantly, in vitro cytology studies, human breast cancer cells (MCF-7), human non-small cell lung cancer cells (A549), and mouse colon cancer cells (CT26) were used to demonstrate that the dual receptor-mediated endocytosis pathway resulted in stronger cytotoxicity to tumor cells and more significant apoptosis. In and in vivo antitumor experiment, tumor-bearing nude mice were used to further confirm that the micelles with double targeting ligands had better antitumor effect and lower toxicity. These experimental results showed that DOX/FA-HA-SS-PLGA micelles have the potential to be used as chemotherapeutic drugs for precise tumor treatment.
Collapse
Affiliation(s)
- Ru Zhang
- Pharmaceutical Engineering Laboratory, Department of Pharmaceutical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Yunying Jiang
- Pharmaceutical Engineering Laboratory, Department of Pharmaceutical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Linkun Hao
- Pharmaceutical Engineering Laboratory, Department of Pharmaceutical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Yang Yang
- Pharmaceutical Engineering Laboratory, Department of Pharmaceutical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Ying Gao
- Pharmaceutical Engineering Laboratory, Department of Pharmaceutical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Ningning Zhang
- Pharmaceutical Engineering Laboratory, Department of Pharmaceutical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Xuecheng Zhang
- Pharmaceutical Engineering Laboratory, Colloge of Marines Life Science, Ocean University of China, Qingdao, China
| | - Yimin Song
- Pharmaceutical Engineering Laboratory, Department of Pharmaceutical Engineering, Qingdao University of Science and Technology, Qingdao, China
| |
Collapse
|
36
|
Huang Y, Wang WF, Huang CX, Li XH, Liu H, Wang HL. miR-731 modulates the zebrafish heart morphogenesis via targeting Calcineurin/Nfatc3a pathway. Biochim Biophys Acta Gen Subj 2022; 1866:130133. [PMID: 35346765 DOI: 10.1016/j.bbagen.2022.130133] [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: 10/08/2021] [Revised: 03/18/2022] [Accepted: 03/23/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Zebrafish miR-731 is orthologous of human miR-425, which has been demonstrated to have cardio-protective roles by a variety of mechanisms. The miR-731 morphants show pericardium enlargement, and many DEGs (differentially expressed genes) are enriched in 'Cardiac muscle contraction' and 'Calcium signaling pathway', implying that miR-731 plays a potential role in heart function and development. However,the in vivo physiological role of miR-731 in the heart needs to be fully defined. METHODS Zebrafish miR-731 morphants were generated by morpholino knockdown, and miR-731 knockout zebrafish was generated by CRISRP/Cas9. We observed cardiac morphogenesis based on whole-mount in situ hybridization. Furthermore, RNA-seq and qRT-PCR were used to elucidate the molecular mechanism and analyze the gene expression. Double luciferase verification and Western blot were used to verify the target gene. RESULTS The depletion of miR-731 in zebrafish embryos caused the deficiency of cardiac development and function, which was associated with reduced heart rate, ventricular enlargement and heart looping disorder. In addition, mechanistic study demonstrated that Calcineurin/Nfatc3a signaling involved in miR-731 depletion induced abnormal cardiac function and developmental defects. CONCLUSION MiR-731 regulates cardiac function and morphogenesis through Calcineurin/Nfatc3a signaling. GENERAL SIGNIFICANCE Our studies highlight the potential importance of miR-731 in cardiac development.
Collapse
Affiliation(s)
- Yan Huang
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Wei-Feng Wang
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Chun-Xiao Huang
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Xuan-Hui Li
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Hong Liu
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Huan-Ling Wang
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, Wuhan, Hubei, PR China.
| |
Collapse
|
37
|
Li D, Liu X, Pi W, Zhang Y, Yu L, Xu C, Sun Z, Jiang J. Fisetin Attenuates Doxorubicin-Induced Cardiomyopathy In Vivo and In Vitro by Inhibiting Ferroptosis Through SIRT1/Nrf2 Signaling Pathway Activation. Front Pharmacol 2022; 12:808480. [PMID: 35273493 PMCID: PMC8902236 DOI: 10.3389/fphar.2021.808480] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/31/2021] [Indexed: 12/12/2022] Open
Abstract
Doxorubicin (DOX) is an anthracycline antibiotic that is used extensively for the management of carcinoma; however, its clinical application is limited due to its serious cardiotoxic side effects. Ferroptosis represents iron-dependent and reactive oxygen species (ROS)-related cell death and has been proven to contribute to the progression of DOX-induced cardiomyopathy. Fisetin is a natural flavonoid that is abundantly present in fruits and vegetables. It has been reported to exert cardioprotective effects against DOX-induced cardiotoxicity in experimental rats. However, the underlying mechanisms remain unknown. The present study investigated the cardioprotective role of fisetin and the underlying molecular mechanism through experiments in the DOX-induced cardiomyopathy rat and H9c2 cell models. The results revealed that fisetin treatment could markedly abate DOX-induced cardiotoxicity by alleviating cardiac dysfunction, ameliorating myocardial fibrosis, mitigating cardiac hypertrophy in rats, and attenuating ferroptosis of cardiomyocytes by reversing the decline in the GPX4 level. Mechanistically, fisetin exerted its antioxidant effect by reducing the MDA and lipid ROS levels and increasing the glutathione (GSH) level. Moreover, fisetin exerted its protective effect by increasing the SIRT1 expression and the Nrf2 mRNA and protein levels and its nuclear translocation, which resulted in the activation of its downstream genes such as HO-1 and FTH1. Selective inhibition of SIRT1 attenuated the protective effects of fisetin in the H9c2 cells, which in turn decreased the GSH and GPX4 levels, as well as Nrf2, HO-1, and FTH1 expressions. In conclusion, fisetin exerts its therapeutic effects against DOX-induced cardiomyopathy by inhibiting ferroptosis via SIRT1/Nrf2 signaling pathway activation.
Collapse
Affiliation(s)
- Danlei Li
- Department of Cardiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Xiaoman Liu
- Department of Cardiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Wenhu Pi
- Key Laboratory of Radiation Oncology of Taizhou, Department of Radiation Oncology, Radiation Oncology Institute of Enze Medical Health Academy, Affiliated Taizhou Hospital of Wenzhou Medical University, Linhai, China
| | - Yang Zhang
- Department of Cardiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Lei Yu
- Department of Cardiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Cheng Xu
- Department of Cardiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Zhenzhu Sun
- Department of Cardiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Jianjun Jiang
- Department of Cardiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| |
Collapse
|
38
|
Nauclea orientalis (L.) Bark Extract Protects Rat Cardiomyocytes from Doxorubicin-Induced Oxidative Stress, Inflammation, Apoptosis, and DNA Fragmentation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1714841. [PMID: 35198093 PMCID: PMC8860544 DOI: 10.1155/2022/1714841] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/22/2022] [Accepted: 01/27/2022] [Indexed: 12/12/2022]
Abstract
The therapeutic efficacy of anthracycline antibiotic, doxorubicin (Dox), is hampered due to the dose-dependent cardiotoxicity. The objective of the study was to explore the counteraction of aqueous bark extract of Nauclea orientalis in Dox-induced cardiotoxicity in Wistar rats. The acute and subchronic toxicity study performed with 2.0 g/kg of the plant extract revealed biochemical and haematological parameters to be within the physiological range, and no histological alterations were observed in any organs isolated. Screening of plant extract for the protection of the myocardium from Dox-induced oxidative stress, inflammation, and apoptosis was performed on five groups of rats: control, plant extract control, Dox control (distilled water (D.H2O) 2 weeks + on the 11th day single injection of Dox, 18 mg/kg), plant + Dox (2.0 g/kg plant extract 2 weeks + on the 11th day Dox, 18 mg/kg), and positive control, dexrazoxane. A significant increase in cardiac biomarkers and lipid peroxidation (p < 0.001) and a significant decrease in antioxidant parameters (p < 0.001) were observed in the Dox control group. All these parameters were reversed significantly (p < 0.05) in the plant-pretreated group. The histopathological assessment of myocardial damage provided supportive evidence for the biochemical results obtained. Inflammatory markers, myeloperoxidase, expression of TNFα and caspase-3, and DNA fragmentation (TUNEL positive nuclei) were significantly elevated (p < 0.05), and expression of Bcl-2 was significantly decreased (p < 0.05) in the Dox control; however, all these parameters were significantly reversed in the plant extract-treated group. In conclusion, the aqueous bark extract of Nauclea orientalis (2.0 g/kg) has the ability to attenuate the Dox-induced oxidative stress, inflammation, apoptosis, and DNA fragmentation in Wistar rats.
Collapse
|
39
|
Liao ZQ, Jiang YN, Su ZL, Bi HL, Li JT, Li CL, Yang XL, Zhang Y, Xie X. Rutaecarpine Inhibits Doxorubicin-Induced Oxidative Stress and Apoptosis by Activating AKT Signaling Pathway. Front Cardiovasc Med 2022; 8:809689. [PMID: 35071368 PMCID: PMC8766983 DOI: 10.3389/fcvm.2021.809689] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 11/23/2021] [Indexed: 11/13/2022] Open
Abstract
Patients with cancer who receive doxorubicin (DOX) treatment can experience cardiac dysfunction, which can finally develop into heart failure. Oxidative stress is considered the most important mechanism for DOX-mediated cardiotoxicity. Rutaecarpine (Rut), a quinazolinocarboline alkaloid extracted from Evodia rutaecarpa was shown to have a protective effect on cardiac disease. The purpose of this study is to investigate the role of Rut in DOX-induced cardiotoxicity and explore the underlying mechanism. Intravenous injection of DOX (5 mg/kg, once a week) in mice for 4 weeks was used to establish the cardiotoxic model. Echocardiography and pathological staining analysis were used to detect the changes in structure and function in the heart. Western blot and real-time PCR analysis were used to detect the molecular changes. In this study, we found that DOX time-dependently decreased cardiac function with few systemic side effects. Rut inhibited DOX-induced cardiac fibrosis, reduction in heart size, and decrease in heart function. DOX-induced reduction in superoxide dismutase (SOD) and glutathione (GSH), enhancement of malondialdehyde (MDA) was inhibited by Rut administration. Meanwhile, Rut inhibited DOX-induced apoptosis in the heart. Importantly, we further found that Rut activated AKT or nuclear factor erythroid 2-related factor 2 (Nrf-2) which further upregulated the antioxidant enzymes such as heme oxygenase-1 (HO-1) and GSH cysteine ligase modulatory subunit (GCLM) expression. AKT inhibitor (AKTi) partially inhibited Nrf-2, HO-1, and GCLM expression and abolished the protective role of Rut in DOX-induced cardiotoxicity. In conclusion, this study identified Rut as a potential therapeutic agent for treating DOX-induced cardiotoxicity by activating AKT.
Collapse
Affiliation(s)
- Zi-Qi Liao
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yi-Nong Jiang
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhuo-Lin Su
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hai-Lian Bi
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jia-Tian Li
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Cheng-Lin Li
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiao-Lei Yang
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ying Zhang
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xin Xie
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
40
|
Li C, Gou X, Gao H. Doxorubicin nanomedicine based on ginsenoside Rg1 with alleviated cardiotoxicity and enhanced antitumor activity. NANOMEDICINE (LONDON, ENGLAND) 2021; 16:2587-2604. [PMID: 34719938 DOI: 10.2217/nnm-2021-0329] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aim: The authors aimed to develop Dox@Rg1 nanoparticles with decreased cardiotoxicity to expand their application in cancer. Materials & methods: Dox@Rg1 nanoparticles were developed by encapsulating doxorubicin (Dox) in a self-assembled Rg1. The antitumor effect of the nanoparticles was estimated using 4T1 tumor-bearing mice and the protective effect on the heart was investigated in vitro and in vivo. Results: Different from Dox, the Dox@Rg1 nanoparticles induced increased cytotoxicity to tumor cells, which was decreased in cardiomyocytes by the inhibition of apoptosis. The study in vivo revealed that the Dox@Rg1 nanoparticles presented a perfect tumor-targeting ability and improved antitumor effects. Conclusion: Dox@Rg1 nanoparticles could enhance the antitumor effects and decrease the cardiotoxicity of Dox.
Collapse
Affiliation(s)
- Chaoqi Li
- Tianjin Key Laboratory of Drug Targeting & Bioimaging, Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, School of Chemistry & Chemical Engineering, Tianjin University of Technology, Tianjin, China
| | - Xiangbo Gou
- Tianjin Key Laboratory of Drug Targeting & Bioimaging, Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, School of Chemistry & Chemical Engineering, Tianjin University of Technology, Tianjin, China
| | - Hui Gao
- Tianjin Key Laboratory of Drug Targeting & Bioimaging, Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, School of Chemistry & Chemical Engineering, Tianjin University of Technology, Tianjin, China.,State Key Laboratory of Separation Membranes & Membrane Processes, School of Materials Science & Engineering, Tiangong University, Tianjin, 300384, China
| |
Collapse
|
41
|
Ding L, Li D, Li M, Zhao D, Govindhan A, Santhoshkumar M, Xiang H. An in vivo and in vitro model on the protective effect of corilagin on doxorubicin-induced cardiotoxicity via regulation of apoptosis and PI3-K/AKT signaling pathways. J Biochem Mol Toxicol 2021; 35:e22926. [PMID: 34605098 DOI: 10.1002/jbt.22926] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 08/29/2021] [Accepted: 09/20/2021] [Indexed: 11/08/2022]
Abstract
Globally, doxorubicin (DOX)-induced cardio dysfunction is a serious cause of morbidity and mortality in cancerous patients. An adverse event of cardiotoxicity is the main deem to restrict in the clinical application by oncologists. Corilagin (CN) is well known for its antioxidative, anti-fibrosis, and anticancer effects. Herein, we aimed to evaluate the action of CN on DOX-induced experimental animals and H9c2 cells. The myocardium-specific marker, CK-MB, and the influx of mitochondrial calcium levels were measured by using commercial kits. Biochemical indices reflecting oxidative stress and antioxidant attributes such as malondialdehyde, glutathione peroxidase, reduced glutathione, superoxide dismutase, and catalase were also analyzed in DOX-induced cardiotoxic animals. In addition, mitochondrial ROS were measured by DCFH-DA in H9c2 cells under fluorescence microscopy. DOX induction significantly increased oxidative stress levels and also modulated apoptosis/survival protein expressions in myocardial tissues. Western blots were used to measure the expressional levels of Bax/Bcl-2, caspase-3, PI3-K/AKT, and PPARγ signaling pathways. Histological studies were executed to observe morphological changes in myocardial tissues. All of these DOX-induced effects were attenuated by CN (100 mg/kg bw). These in vitro and in vivo results point towards the fact that CN might be a novel cardioprotective agent against DOX-induced cardiotoxicity through modulating cardio apoptosis and oxidative stress.
Collapse
Affiliation(s)
- Lianqin Ding
- Department of Cardiology, Shenzhen Samii Medical Center (The Fourth People's Hospital of Shenzhen), Shenzhen, China
| | - Di Li
- Department of Cardiology, Daqing Oil Field General Hospital, Daqing, China
| | - Ming Li
- Department of Reproductive Medicine Center, Huizhou Second Maternal and Child Health Care Hospital, Huizhou, China
| | - Dexia Zhao
- Department of Echocardiographic, Daqing Oil Field General Hospital, Daqing, China
| | - Annamalai Govindhan
- Department of Biotechnology, Joseph Arts and Science Collage, Thirunavallur, India
| | - Mani Santhoshkumar
- Department of Botany, Joseph Arts and Science Collage, Thirunavallur, India
| | - Hongli Xiang
- Department of General Medicine, Chongqing University Three Gorges Hospital, Chongqing, China
| |
Collapse
|
42
|
Wu C, Zhao L, Li X, Xu Y, Guo H, Huang Z, Wang Q, Liu H, Chen D, Zhu M. Integrated Bioinformatics Analysis of Potential mRNA and miRNA Regulatory Networks in Mice With Ischemic Stroke Treated by Electroacupuncture. Front Neurol 2021; 12:719354. [PMID: 34566862 PMCID: PMC8461332 DOI: 10.3389/fneur.2021.719354] [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: 06/10/2021] [Accepted: 07/22/2021] [Indexed: 12/02/2022] Open
Abstract
Background: The complicated molecular mechanisms underlying the therapeutic effect of electroacupuncture (EA) on ischemic stroke are still unclear. Recently, more evidence has revealed the essential role of the microRNA (miRNA)–mRNA networks in ischemic stroke. However, a systematic analysis of novel key genes, miRNAs, and miRNA–mRNA networks regulated by EA in ischemic stroke is still absent. Methods: We established a middle cerebral artery occlusion (MCAO) mouse model and performed EA therapy on ischemic stroke mice. Behavior tests and measurement of infarction area were applied to measure the effect of EA treatment. Then, we performed RNA sequencing to analyze differentially expressed genes (DEGs) and functional enrichment between the EA and control groups. In addition, a protein–protein interaction (PPI) network was built, and hub genes were screened by Cytoscape. Upstream miRNAs were predicted by miRTarBase. Then hub genes and predicted miRNAs were verified as key biomarkers by RT-qPCR. Finally, miRNA–mRNA networks were constructed to explore the potential mechanisms of EA in ischemic stroke. Results: Our analysis revealed that EA treatment could significantly alleviate neurological deficits in the affected limbs and reduce infarct area of the MCAO model mice. A total of 174 significant DEGs, including 53 upregulated genes and 121 downregulated genes, were identified between the EA and control groups. Functional enrichment analysis showed that these DEGs were associated with the FOXO signaling pathway, NF-kappa B signaling pathway, T-cell receptor signaling pathway, and other vital pathways. The top 10 genes with the highest degree scores were identified as hub genes based on the degree method, but only seven genes were verified as key genes according to RT-qPCR. Twelve upstream miRNAs were predicted to target the seven key genes. However, only four miRNAs were significantly upregulated and indicated favorable effects of EA treatment. Finally, comprehensive analysis of the results identified the miR-425-5p-Cdk1, mmu-miR-1186b-Prc1, mmu-miR-434-3p-Prc1, and mmu-miR-453-Prc1 miRNA–mRNA networks as key networks that are regulated by EA and linked to ischemic stroke. These networks might mainly take place in neuronal cells regulated by EA in ischemic stroke. Conclusion: In summary, our study identified key DEGs, miRNAs, and miRNA–mRNA regulatory networks that may help to facilitate the understanding of the molecular mechanism underlying the effect of EA treatment on ischemic stroke.
Collapse
Affiliation(s)
- Chunxiao Wu
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Guangdong, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lijun Zhao
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Xinrong Li
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Yingshan Xu
- Clinical Medical of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongji Guo
- Clinical Medical of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zifeng Huang
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Qizhang Wang
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Helu Liu
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Dongfeng Chen
- The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Meiling Zhu
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Guangdong, China
| |
Collapse
|
43
|
Yarmohammadi F, Hayes AW, Karimi G. The cardioprotective effects of hydrogen sulfide by targeting endoplasmic reticulum stress and the Nrf2 signaling pathway: A review. Biofactors 2021; 47:701-712. [PMID: 34161646 DOI: 10.1002/biof.1763] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 12/20/2022]
Abstract
Cardiac diseases are emerging due to lifestyle, urbanization, and the accelerated aging process. Oxidative stress has been associated with cardiac injury progression through interference with antioxidant strategies and endoplasmic reticulum (ER) function. Hydrogen sulfide (H2 S) is generated endogenously from l-cysteine in various tissues including heart tissue. Pharmacological evaluation of H2 S has suggested a potential role for H2 S against diabetic cardiomyopathy, ischemia/reperfusion injury, myocardial infarction, and cardiotoxicity. Nuclear factor E2-related factor 2 (Nrf2) activity is crucial for cell survival in response to oxidative stress. H2 S up-regulates Nrf2 expression and its related signaling pathway in myocytes. H2 S also suppresses the expression and activity of ER stress-related proteins. H2 S has been reported to improve various cardiac conditions through antioxidant and anti-ER stress-related activities.
Collapse
Affiliation(s)
- Fatemeh Yarmohammadi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Hayes
- Center for Environmental Occupational Risk Analysis and Management, College of Public Health, University of South Florida, Tampa, Florida, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
44
|
Yarmohammadi F, Karbasforooshan H, Hayes AW, Karimi G. Inflammation suppression in doxorubicin-induced cardiotoxicity: natural compounds as therapeutic options. Naunyn Schmiedebergs Arch Pharmacol 2021; 394:2003-2011. [PMID: 34350498 DOI: 10.1007/s00210-021-02132-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 07/28/2021] [Indexed: 12/20/2022]
Abstract
Doxorubicin (DOX) is a potent chemotherapeutic agent; however, the accompanying cardiotoxicity is a significant complication of the usefulness of treatment with DOX. Multiple mechanisms have been suggested for this often fatal side effect, one of which is inflammation. Several pathways with different targets have been reported to result in DOX-induced heart inflammation. Some natural occurring compounds (NCs) have been reported to interact with the DOX-induced cardiotoxicity through targeting one or more of several pathways, including the Nrf2/NF-kB, TLR-4/NF-kB, MAPK/NF-kB, and NLRP3 inflammasome pathways. This article reviews several of these pathways and the potential protective effect of some NCs against the cardiac inflammation induced by DOX.
Collapse
Affiliation(s)
- Fatemeh Yarmohammadi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hedyieh Karbasforooshan
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Hayes
- Center for Environmental Occupational Risk Analysis and Management, College of Public Health, University of South Florida, Tampa, FL, USA.,Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran. .,Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
45
|
Awad HH, El-Derany MO, Mantawy EM, Michel HE, El-Naa MM, Salah El-Din RA, El-Brairy AI, El-Demerdash E. Comparative study on beneficial effects of vitamins B and D in attenuating doxorubicin induced cardiotoxicity in rats: Emphasis on calcium homeostasis. Biomed Pharmacother 2021; 140:111679. [PMID: 34029952 DOI: 10.1016/j.biopha.2021.111679] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/25/2021] [Accepted: 04/27/2021] [Indexed: 12/13/2022] Open
Abstract
The use of doxorubicin (DOX) to treat various tumors is limited by its cardiotoxicity. This study aimed to investigate and compare the cardioprotective effects of nicotinamide (NAM) and alfacalcidol (1α(OH)D3), against DOX-induced cardiotoxicity. Sprague Dawley male rats received DOX (5 mg/kg, i.p.) once/week for four consecutive weeks. Treated groups received either NAM (600 mg/kg, p.o.) for 28 consecutive days or 1α(OH)D3 (0.5 ug/kg, i.p.) once/week for four consecutive weeks. DOX elicited marked cardiac tissue injury manifested by elevated serum cardiotoxicity indices, conduction and histopathological abnormalities. Both NAM and 1α(OH)D3 successfully reversed all these changes. From the mechanistic point of view, DOX provoked intense cytosolic and mitochondrial calcium (Ca2+) overload hence switching on calpain1 (CPN1) and mitochondrial-mediated apoptotic cascades as confirmed by upregulating Bax and caspase-3 while downregulating Bcl-2 expression. DOX also disrupted cardiac bioenergetics as evidenced by adenosine triphosphate (ATP) depletion and a declined ATP/ADP ratio. Moreover, DOX upregulated the Ca2+ sensor; calmodulin kinase II gamma (CaMKII-δ) which further contributed to cardiac damage. Interestingly, co-treatment with either NAM or 1α(OH)D3 reversed all DOX associated abnormalities by preserving Ca2+ homeostasis, replenishing ATP stores and obstructing apoptotic events. Additionally, DOX prompted nuclear factor kappa B (NF-κB) dependent inflammatory responses and subsequently upregulated interleukin-6 (IL-6) expression. Co-treatment with NAM or 1α(OH)D3 effectively obstructed these inflammatory signals. Remarkably, NAM showed superior beneficial cardioprotective properties over 1α(OH)D3. Both NAM and 1α(OH)D3 efficiently attenuated DOX-cardiomyopathy mainly via preserving Ca2+ homeostasis and diminishing apoptotic and inflammatory pathways. NAM definitely exhibited effective cardioprotective capabilities over 1α(OH)D3.
Collapse
Affiliation(s)
- Heba H Awad
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, October University for Modern Sciences & Arts (MSA University), Cairo, Egypt
| | - Marwa O El-Derany
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Eman M Mantawy
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Haidy E Michel
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mona M El-Naa
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Sadat City, Sadat City, Egypt
| | | | - Amany I El-Brairy
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, October University for Modern Sciences & Arts (MSA University), Cairo, Egypt
| | - Ebtehal El-Demerdash
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
46
|
Tang S, Kan J, Sun R, Cai H, Hong J, Jin C, Zong S. Anthocyanins from purple sweet potato alleviate doxorubicin-induced cardiotoxicity in vitro and in vivo. J Food Biochem 2021; 45:e13869. [PMID: 34287964 DOI: 10.1111/jfbc.13869] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/16/2021] [Accepted: 07/03/2021] [Indexed: 12/21/2022]
Abstract
In this study, anthocyanins were extracted and purified from purple sweet potato anthocyanins (PSPA) and the alleviative effect of PSPA on doxorubicin (DOX)-induced cardiotoxicity was investigated. High Performance Liquid Chromatography-Mass Spectrometry (HPLC-MS) results showed that 10 kinds of substances were identified in PSPA and the PSPA was mainly composed of cyanidin (62.9%) and peonidin (21.46%). In in vitro experiments, PSPA reduced the excessive release of inflammatory factors (NO and TNF-α) induced by DOX and decreased the secretion of trimethylamine oxide (TMAO), lactic dehydrogenase (LDH), and creatine kinase (CK) caused by myocardial injury. In in vivo experiments, PSPA inhibited the release of NO and MDA levels in heart tissue. Meanwhile, mice treated with PSPA decreased the levels of LDH, CK, TNF-α, and TMAO in serum and heart tissue when compared with the DOX group. In addition, the histopathological results of the heart tissue also showed a protective effect of PSPA on the pathological changes in heart. These results provide a reference for the application of PSPA as a functional food to intervene in DOX-induced cardiotoxicity. PRACTICAL APPLICATIONS: The effects of anthocyanins from purple sweet potato anthocyanins (PSPA) on doxorubicin (DOX)-induced cardiotoxicity were investigated in vitro and in vivo. The results indicated that PSPA could significantly ameliorate DOX-induced heart failure. The obtained results could provide the potential application of PSPA as an alternative therapy for cardiotoxicity caused by DOX in the functional food industry.
Collapse
Affiliation(s)
- Sixue Tang
- College of Food Science and Engineering, Yangzhou University, Yangzhou, PR China
| | - Juan Kan
- College of Food Science and Engineering, Yangzhou University, Yangzhou, PR China
| | - Rui Sun
- College of Food Science and Engineering, Yangzhou University, Yangzhou, PR China
| | - Huahao Cai
- College of Food Science and Engineering, Yangzhou University, Yangzhou, PR China
| | - Jinhai Hong
- College of Food Science and Engineering, Yangzhou University, Yangzhou, PR China
| | - Changhai Jin
- College of Food Science and Engineering, Yangzhou University, Yangzhou, PR China
| | - Shuai Zong
- College of Food Science and Engineering, Yangzhou University, Yangzhou, PR China
| |
Collapse
|
47
|
Chen Y, Xu Y, Deng Z, Wang Y, Zheng Y, Jiang W, Jiang L. MicroRNA expression profiling involved in doxorubicin-induced cardiotoxicity using high-throughput deep-sequencing analysis. Oncol Lett 2021; 22:560. [PMID: 34093775 PMCID: PMC8170198 DOI: 10.3892/ol.2021.12821] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 03/09/2021] [Indexed: 12/31/2022] Open
Abstract
MicroRNAs (miRNAs/miRs) are sensitive biomarkers and endogenous repressors of gene expression by decreasing mRNA stability and interfering with mRNA translation. Despite a number of investigations revealing the dysregulation of miRNA expression associated with cardiotoxicity induced by doxorubicin (Dox), perturbation of miRNAs directly resulting from Dox at early stage in cardiomyocytes and the target gene interaction remain largely unknown. In the present study, high-throughput deep-sequencing was used to analyze changes in global miRNA expression in H9c2 cardiomyocytes exposed to 5 µg/ml Dox for 0, 12 or 24 h. Compared with the 0-h time point, the expression levels of 386 unique miRNAs were altered. Based on miRNA expression and fold-change, the target genes of 76 selected miRNAs were further analyzed using gene interaction networks and pathway enrichment analysis. These miRNAs were involved in the regulation of different pathways, whose functions included apoptosis, cell proliferation, extracellular matrix remodeling, oxidative stress and lipid metabolism. These differentially expressed miRNAs included let-7 family, miR-29b-3p, miR-378-3/5p, miR-351-3p, miR-664-3p, miR-455-3p, miR-298-3p, miR-702-5p, miR-128-1-5p, miR-671 and miR-421-5p. The present data indicated that global wide miRNA profiling in Dox-induced cardiomyocytes may provide a novel mechanistic insight into understanding Dox-induced heart failure and cardiotoxicity, as well as novel biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Ying Chen
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Yingjie Xu
- Department of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Zhoufeng Deng
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Yin Wang
- Department of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Ying Zheng
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200336, P.R. China
| | - Weihua Jiang
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Li Jiang
- Department of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| |
Collapse
|
48
|
Li J, Tu J, Gao H, Tang L. MicroRNA-425-3p inhibits myocardial inflammation and cardiomyocyte apoptosis in mice with viral myocarditis through targeting TGF-β1. Immun Inflamm Dis 2021; 9:288-298. [PMID: 33332750 PMCID: PMC7860592 DOI: 10.1002/iid3.392] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/22/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Emerging articles have profiled the relations between microRNAs and viral myocarditis. This research was unearthed to explore the capacity of miR-425-3p on cardiomyocyte apoptosis in mice with viral myocarditis and its mechanism. METHODS A total of 120 mice were classified into 4 groups in a random fashion (n = 30). The mice were intraperitoneally injected with coxsackievirus type B3 (CVB3) to induce myocarditis. On the 7th day after CVB3 infection, 10 mice in each group were euthanized to assess the heart function indices of mice, observe the pathological conditions, detect myocardial tissue apoptosis, and measure the inflammatory factor levels in myocardial tissues. Expression of miR-425-3p, transforming growth factor (TGF-β1), and apoptosis-associated proteins in myocardial tissues was determined. The remaining 20 mice in each group were used for survival observation. The luciferase activity assay was implemented to validate the relationship between miR-425-3p and TGF-β1. miR-425-3p mimic was transfected into mouse cardiomyocytes HL-1 and then infected with CVB3 to further verify the regulatory effect of miR-425-3p on the cardiomyocyte apoptosis in viral myocarditis. RESULTS miR-425-3p was lowly expressed in myocardial tissues of mice with viral myocarditis. Overexpressed miR-425-3p improved the cardiac function, alleviated pathological conditions, reduced cardiomyocyte apoptosis, decreased Bax and cleaved Caspase-3 expression, elevated Bcl-2 expression, decreased levels of inflammatory factors and improved survival rate of mice with viral myocarditis. Luciferase activity assay verified that miR-425-3p could bind to TGF-β1, and overexpressed miR-425-3p suppressed TGF-β1, p-smad2/smad2 and p-smad3/smad3 expression. In vitro experiments further verified that overexpression of miR-425-3p inhibited the apoptosis of CVB3-HL-1 cells, and the addition of TGF-β1 would reverse this effect. CONCLUSION Our research indicates that miR-425-3p is poorly expressed in myocardial tissues of mice with viral myocarditis. Overexpressed miR-425-3p inhibits cardiomyocyte apoptosis and myocardial inflammation in mice with viral myocarditis as well as improves their survival rates through suppressing the TGF-β1/smad axis.
Collapse
Affiliation(s)
- Junhua Li
- Department of CardiologyThe Third Affiliated Hospital of Nanchang University (The First Hospital of Nanchang)NanchangJiangxiChina
| | - Jiehong Tu
- Department of CardiologyThe Third Affiliated Hospital of Nanchang University (The First Hospital of Nanchang)NanchangJiangxiChina
| | - Hong Gao
- Department of CardiologyThe Third Affiliated Hospital of Nanchang University (The First Hospital of Nanchang)NanchangJiangxiChina
| | - Lu Tang
- Department of PediatricsXD Group HospitalXi'anShaanxiChina
| |
Collapse
|
49
|
Zhang Q, Xue T, Guan J, Wang W, Shi J, Lu J, Jiang X. Irigenin alleviates angiotensin II-induced oxidative stress and apoptosis in HUVEC cells by activating Nrf2 pathway. Drug Dev Res 2021; 82:999-1007. [PMID: 33634899 DOI: 10.1002/ddr.21802] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/25/2021] [Accepted: 01/29/2021] [Indexed: 11/12/2022]
Abstract
Endothelial dysfunction is closely related to various cardiovascular diseases. Oxidative stress and apoptosis are involved in the progress of endothelial dysfunction. Irigenin (IR) has antioxidative properties. We investigated IR as a novel therapy for angiotensin II (Ang II)-induced endothelial dysfunction and explored the potential mechanisms of IR. After human umbilical vein endothelial cell lines (HUVECs) were treated with Ang II (100, 200, 300 and 400 nmol/L) alone, IR (2.5, 5, 10, 20 and 40 μmol/L) alone or Ang II plus IR for 24 h, HUVECs viability, lactate dehydrogenase (LDH), apoptosis, oxidative stress, apoptosis-related protein and nuclear factor E2-related factor 2 (Nrf2) levels were detected by Cell Counting Kit (CCK)-8 assay, enzyme-linked immunosorbent assay, flow cytometry and western blot. Transfection rate of Nrf2 was detected by western blot. In the next rescue experiment, we used silent Nrf2 (siNrf2) to verify the previous experimental results. Different concentrations' Ang II repressed HUVECs viability and increased LDH release, and different concentrations' IR did not affect HUVECs viability or LDH release. Furthermore, IR elevated cell viability and Nrf2 level, inhibited LDH release, apoptosis, oxidative stress and apoptosis-related protein levels in Ang II-induced HUVECs. More important, siNrf2 suppressed the expression of Nrf2, and siNrf2 abrogated the protective effect of IR on Ang II-induced Nrf2 expression, cell viability, LDH activity, oxidative stress generation and apoptosis-related protein in HUVECs. IR protected HUVECs from Ang II-induced oxidative stress and apoptosis injury by activating Nrf2 pathway.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Cardiology, First People's Hospital Affiliated to Huzhou University, Huzhou City, Zhejiang Province, China
| | - Tao Xue
- Department of Medical Therapeutics, First People's Hospital Affiliated to Huzhou University, Huzhou City, Zhejiang Province, China
| | - Jianming Guan
- Department of Ultrasound, First People's Hospital Affiliated to Huzhou University, Huzhou City, Zhejiang Province, China
| | - Wei Wang
- Department of Cardiology, First People's Hospital Affiliated to Huzhou University, Huzhou City, Zhejiang Province, China
| | - Ji Shi
- Department of Cardiology, First People's Hospital Affiliated to Huzhou University, Huzhou City, Zhejiang Province, China
| | - Jianzhong Lu
- Department of Cardiology, First People's Hospital Affiliated to Huzhou University, Huzhou City, Zhejiang Province, China
| | - Xiping Jiang
- Department of Cardiology, The First People's Hospital Of Jiande, Jiande City, Zhejiang Province, China
| |
Collapse
|
50
|
Bone marrow mesenchymal stem cell-derived exosomes alleviate hyperoxia-induced lung injury via the manipulation of microRNA-425. Arch Biochem Biophys 2020; 697:108712. [PMID: 33264631 DOI: 10.1016/j.abb.2020.108712] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 11/22/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Hyperoxia-induced lung injury (HILI) is an acute lung injury (LI) induced by extended periods of exposure to hyperoxia. Alleviating LI by bone marrow mesenchymal stem cell-derived exosomes (BMSCs-Exos) and microRNAs (miRs) has been previously reported. This study is devised to probe the interaction between BMSCs-Exos and miR-425 in HILI. METHODS Firstly, BMSCs-Exos were isolated and identified. Then, HILI rat models and RLE-6TN cell models were successfully established and treated by BMSCs-Exos. Afterwards, functional assays were conducted to explore cell biological behaviors in models, with miR-425 expression detected. Then, the target relation between miR-425 and PTEN was clarified by luciferase reporter assay. Eventually, expression of PTEN and the PI3K/Akt axis was assessed by Western blotting and qRT-PCR. RESULTS BMSCs-Exos promoted miR-425 expression and attenuated HILI and H2O2 induced RLE-6TN cell injury as evidence by alleviated lung cell injury, decreased TUNEL-positive cells, induced cell viability and declined apoptosis (all p < 0.05). Besides, when miR-425 was knocked-down, the protective role of BMSCs-Exos in HILI was also reduced (all p < 0.05). miR-425 targeted PTEN mRNA, whose upregulation reversed the protective role of BMSCs-Exos in HILI (all p < 0.05). BMSCs-Exos improved the quenched levels of the PI3K/AKT axis in HILI (all p < 0.05). CONCLUSION Our data supported that miR-425 in BMSCs-Exos inhibits HILI by targeting PTEN and upregulating the PI3K/AKT axis. This study may provide personalized interventions for HILI remedy.
Collapse
|