1
|
Bishr A, Atwa AM, El-Mokadem BM, El-Din MN. Canagliflozin potentially promotes renal protection against glycerol-induced acute kidney injury by activating the AMPK/SIRT1/FOXO-3a/PGC-1α and Nrf2/HO-1 pathways. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04017-x. [PMID: 40257493 DOI: 10.1007/s00210-025-04017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 03/04/2025] [Indexed: 04/22/2025]
Abstract
The reno-protective potential of canagliflozin (Cana), an inhibitor of the sodium glucose-linked co-transporter-2 (SGLT-2), has been demonstrated in different models of kidney injury. However, its potential role in preventing glycerol (Gly)-induced acute kidney injury (AKI) remains to be divulged. Therefore, the aim of this study is to investigate the potential reno-protective effect of Cana and its underlying mechanism in a rat model of Gly-induced AKI. Rats were randomly allocated into five groups: normal, Gly, Gly pretreated with 10 mg/kg Cana, Gly pretreated with Cana 25 mg/kg, and normal pretreated with Cana 25 mg/kg for 14 consecutive days. Pretreatment with Cana improved renal structure and enhanced kidney functions manifested by reducing serum creatinine and blood urea nitrogen, as well as renal contents of neutrophil gelatinase-associated lipocalin and kidney injury molecule. Moreover, Cana signified its anti-inflammatory effect by reducing the Gly-induced elevation in renal contents of nuclear factor-κB and interleuκin-6. Additionally, Cana augmented the defense enzymatic antioxidants superoxide dismutase (SOD), manganese-SOD, and heme oxygenase-1, besides increasing the protein expression of the antioxidant transcription factor nuclear factor erythroid 2-related factor 2 to point for its ability to correct redox balance. Cana also upregulated the protein expression of the 5' adenosine monophosphate-activated protein kinase (AMPK), Sirtuin1 (SIRT1), Forkhead box protein O3 (FOXO-3a), and peroxisome proliferator-activated receptor-gamma coactivator 1α (PGC-1α), as well as the transcriptional activity of growth arrest and DNA damage-inducible protein alpha (GAAD45a). In conclusion, Cana demonstrated potentially novel reno-protective mechanisms and mitigated the consequences of AKI through its antioxidant and anti-inflammatory properties, partially by activating the AMPK/SIRT1/FOXO-3a/PGC-1α pathway.
Collapse
Affiliation(s)
- Abeer Bishr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt.
| | - Ahmed M Atwa
- Department of Pharmacology and Toxicology, Egyptian Russian University, Cairo, Egypt
- College of Pharmacy, Al-Ayen Iraqi University, AUIQ, An Nasiriyah, Iraq
| | - Bassant M El-Mokadem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Chinese University, Cairo, Egypt
| | - Mahmoud Nour El-Din
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Sadat City, Menoufia, Egypt
| |
Collapse
|
2
|
Simela C, Walker JM, Ghosh AK, Chen DH. SGLT2 inhibitors for prevention and management of cancer treatment-related cardiovascular toxicity: a review of potential mechanisms and clinical insights. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:15. [PMID: 39934910 DOI: 10.1186/s40959-024-00284-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/30/2024] [Indexed: 02/13/2025]
Abstract
More evidence-based strategies are needed for preventing and managing cancer treatment-related cardiovascular toxicity (CTR-CVT). Owing to the growing body of evidence supporting their cardioprotective role in several cardiac injury scenarios, sodium-glucose cotransporter 2 inhibitors (SGLT2i) may be beneficial for preventing and treating CTR-CVT. In October 2024, a search was conducted of the PubMed database to review full studies investigating the cardioprotective role of SGLT2i against CTR-CVT. We identified 44 full published/pre-print studies and 3 ongoing randomised controlled trial across eight types of cancer treatment (anthracyclines, platinum-containing therapy, immune checkpoint inhibitors, HER2-targeted therapies, kinase inhibitors, androgen deprivation therapies, multiple myeloma therapies and 5-fluorouracil). Most studies used animal models and focussed on primary prevention. 43 of the 44 studies found some cardioprotective effect of SGLT2i against CTR-CVT, which in some cases included preventing ejection fraction decline and aberrations in cardiac electrophysiological parameters. Some studies also observed beneficial effects on mortality. A central triad of anti-inflammatory, anti-oxidative and anti-apoptotic mechanisms likely underlie SGLT2i-mediated cardioprotection against CTR-CVT. Overall, this growing body of research suggests that SGLT2i may be a promising candidate for preventing CTR-CVT either as monotherapy or in combination with other cardioprotective drugs. However, the literature is limited in that no prospective randomised controlled trials investigating SGLT2i for the prevention and management of CTR-CVT exist and most existing human retrospective data is based on diabetic populations. Future work must focus on addressing these limitations of the current literature.
Collapse
Affiliation(s)
- Carl Simela
- University College London Hospital, London, UK
| | - J Malcolm Walker
- University College London Hospital, London, UK
- Hatter Cardiovascular Institute, University College London, London, UK
| | - Arjun K Ghosh
- University College London Hospital, London, UK
- Hatter Cardiovascular Institute, University College London, London, UK
- Barts Heart Centre, London, UK
| | - Daniel H Chen
- University College London Hospital, London, UK.
- Hatter Cardiovascular Institute, University College London, London, UK.
- Barts Heart Centre, London, UK.
- Prince of Wales Hospital, Sydney, NSW, Australia.
- St George Hospital, Sydney NSW, Australia.
| |
Collapse
|
3
|
Wang J, Yang W. Advances in sodium-glucose transporter protein 2 inhibitors and tumors. Front Oncol 2025; 15:1522059. [PMID: 40007997 PMCID: PMC11850236 DOI: 10.3389/fonc.2025.1522059] [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: 11/03/2024] [Accepted: 01/15/2025] [Indexed: 02/27/2025] Open
Abstract
Tumor is a major challenge to global health and has received extensive attention worldwide due to its high degree of malignancy and poor prognosis. Although the clinical application of targeted therapy and immunotherapy has improved the status quo of tumor treatment, the development of new therapeutic tools for tumors is still necessary. Sodium-glucose transporter protein 2 (SGLT2) inhibitors are a new type of glycemic control drugs, which are widely used in clinical practice because of their effects on weight reduction and protection of cardiac and renal functions. SGLT2 has been found to be overexpressed in many tumors and involved in tumorigenesis, progression and metastasis, suggesting that SGLT2i has a wide range of applications in tumor therapy. The aim of this article is to provide a comprehensive understanding of the research progress of SGLT2i in different tumors by integrating the latest studies and to encourage further exploration of SGLT2i therapies in clinical trials. This could pave the way for more effective management strategies and improved outcomes for tumor patients.
Collapse
Affiliation(s)
- Jiaqi Wang
- Department of Oncology, Guangyuan Central Hospital, Guangyuan, Sichuan, China
| | - Wenyong Yang
- Department of Respiratory and Critical Care Medicine, Guangyuan Central Hospital, Guangyuan, Sichuan, China
| |
Collapse
|
4
|
Soares RR, Viggiani LF, Reis Filho JM, Joviano-Santos JV. Cardioprotection of Canagliflozin, Dapagliflozin, and Empagliflozin: Lessons from preclinical studies. Chem Biol Interact 2024; 403:111229. [PMID: 39244185 DOI: 10.1016/j.cbi.2024.111229] [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: 05/20/2024] [Revised: 08/12/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Clinical and preclinical studies have elucidated the favorable effects of Inhibitors of Sodium-Glucose Cotransporter-2 (iSGLT2) in patients and animal models with type 2 diabetes. Notably, these inhibitors have shown significant benefits in reducing hospitalizations and mortality among patients with heart failure. However, despite their incorporation into clinical practice for indications beyond diabetes, the decision-making process regarding their use often lacks a systematic approach. The selection of iSGLT2 remains arbitrary, with only a limited number of studies simultaneously exploring the different classes of them. Currently, no unique guideline establishes their application in both clinical and basic research. This review delves into the prevalent use of iSGLT2 in animal models previously subjected to induced cardiac stress. We have compiled key findings related to cardioprotection across various animal models, encompassing diverse dosages and routes of administration. Beyond their established role in diabetes management, iSGLT2 has demonstrated utility as agents for safeguarding heart health and cardioprotection can be class-dependent among the iSGLT2. These findings may serve as valuable references for other researchers. Preclinical studies play a pivotal role in ensuring the safety of novel compounds or treatments for potential human use. By assessing side effects, toxicity, and optimal dosages, these studies offer a robust foundation for informed decisions, identifying interventions with the highest likelihood of success and minimal risk to patients. The insights gleaned from preclinical studies, which play a crucial role in highlighting areas of knowledge deficiency, can guide the exploration of novel mechanisms and strategies involving iSGLT2.
Collapse
Affiliation(s)
- Rayla Rodrigues Soares
- Faculdade Ciências Médicas de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Laboratório de Investigações NeuroCardíacas, Ciências Médicas de Minas Gerais (LINC CMMG), Belo Horizonte, Minas Gerais, Brazil
| | - Larissa Freitas Viggiani
- Faculdade Ciências Médicas de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Laboratório de Investigações NeuroCardíacas, Ciências Médicas de Minas Gerais (LINC CMMG), Belo Horizonte, Minas Gerais, Brazil
| | - Juliano Moreira Reis Filho
- Post-Graduate Program in Health Sciences, Faculdade Ciências Médicas de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Julliane V Joviano-Santos
- Post-Graduate Program in Health Sciences, Faculdade Ciências Médicas de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Laboratório de Investigações NeuroCardíacas, Ciências Médicas de Minas Gerais (LINC CMMG), Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
5
|
Pan R, He Y, Melisandre W, Zhang Y, Su W, Feng J, Jia C, Li S, Liu B. Bibliometric and visual analysis of SGLT2 inhibitors in cardiovascular diseases. Front Pharmacol 2024; 15:1437760. [PMID: 39539627 PMCID: PMC11557488 DOI: 10.3389/fphar.2024.1437760] [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: 05/24/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Background Cardiovascular diseases (CVD) pose a significant threat to human health due to their high mortality and morbidity rates. Despite advances in treatments, the prevalence and impact of cardiovascular disease continue to increase. Sodium-glucose transporter 2 inhibitors (SGLT2i), initially approved for the treatment of type 2 diabetes, have important research value and promising applications in reducing CVD risk, especially in heart failure (HF) and atherosclerosis patients with cardiovascular disease (ASCVD). This study aims to comprehensively review the latest progress, research trends, cutting-edge hot spots, and future development directions of SGLT2i in the field of CVD through bibliometric analysis. Methods Articles related to MSCs in cardiovascular diseases were sourced from the Web of Science. The bibliometric analysis was conducted using CiteSpace and VOSviewer, and a knowledge map was created based on the data obtained from the retrieved articles. Results In this article, we screened 3,476 relevant studies, including 2,293 articles and 1,183 reviews. The analysis found that the number of papers related to the application of SGLT2i in CVD has generally increased, peaking in 2022. The United States and China contributed the largest number of papers, with the United States accounting for 36.97% of the total and also ranking first in terms of the number of citations. However, China's high-quality papers are slightly lacking and need further improvement. Keyword analysis showed that empagliflozin, dapagliflozin, diabetes, and heart failure were the most common terms, reflecting the main research interests in currently published papers in this field. Conclusion Bibliometric analysis showed a robust and growing interest in the application of SGLT2i for treating CVD. By summarizing the latest progress of SGLT2i in the field of CVD, exploring research hotspots, and looking forward to future research development trends, this article provides valuable insights for thinking about research prospects.
Collapse
Affiliation(s)
- Runfang Pan
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuqing He
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wan Melisandre
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yunyi Zhang
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenyuan Su
- Sport Medicine and Rehabilitation Center, Shanghai University of Sport, Shanghai, China
| | - Jiaming Feng
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chengyao Jia
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Shaoling Li
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Baonian Liu
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
6
|
Faggiano A, Gherbesi E, Cardinale D, Vicenzi M, Carugo S. SGLT2-i prevent left ventricular dysfunction induced by anthracycline in mouse model: A systematic-review and meta-analysis. Vascul Pharmacol 2023; 150:107171. [PMID: 37061151 DOI: 10.1016/j.vph.2023.107171] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 04/17/2023]
Abstract
CLINICAL QUESTION Could SGLT2-i be helpful for the prevention of left ventricular dysfunction induced by anthracycline? WHAT IS THE MAIN FINDING?: SGLT2-i appear effective for the prevention of left ventricular dysfunction induced by anthracycline in mouse model.
Collapse
Affiliation(s)
- Andrea Faggiano
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy.
| | - Elisa Gherbesi
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Daniela Cardinale
- Cardioncology Unit, European Institute of Oncology, IRCCS, Milan, Italy
| | - Marco Vicenzi
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Stefano Carugo
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| |
Collapse
|
7
|
Schisandrin B Diet Inhibits Oxidative Stress to Reduce Ferroptosis and Lipid Peroxidation to Prevent Pirarubicin-Induced Hepatotoxicity. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5623555. [PMID: 36060128 PMCID: PMC9433297 DOI: 10.1155/2022/5623555] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/18/2022] [Accepted: 05/03/2022] [Indexed: 12/06/2022]
Abstract
Objective Pirarubicin (THP) is one of anthracycline anticancer drugs. It is widely used in the treatment of various cancers, but its hepatotoxicity cannot be ignored. Schisandrin B (SchB) is a traditional liver-protecting drug, which has the ability to promote mitochondrial function and upregulate cellular antioxidant defense mechanism. However, whether it can resist THP-induced hepatotoxicity has not been reported. The purpose of this study was to observe and explore the effect of SchB on THP-induced hepatotoxicity and its potential mechanism by adding SchB to the diet of rats with THP-induced hepatotoxicity. Methods The rat model of THP-induced hepatotoxicity was established and partly treated with SchB diet. The changes of serum liver function indexes ALT and AST were observed. The histomorphological changes of liver were observed by HE staining. The biomarker levels of oxidative stress in rat serum and liver were measured to observe oxidative stress state. The expressions of ferroptosis-related protein GPX4 and oxidative stress-related protein were detected by Western blot. Primary hepatocytes were prepared and cocultured with THP, SchB, and Fer-1 to detect the production of reactive oxygen species (ROS) and verify the above signal pathways. Results THP rats showed a series of THP-induced hepatotoxicity changes, such as liver function damage, oxidative stress, and ferroptosis. SchB diet effectively alleviated these adverse reactions. Further studies showed that SchB had strong antioxidant and antiferroptosis abilities in THP-induced hepatotoxicity. Conclusion SchB has obvious protective effect on THP-induced hepatotoxicity. The mechanism may be closely related to inhibiting oxidative stress and ferroptosis in the liver.
Collapse
|
8
|
Wei Y, Zhao J, Xiong J, Chai J, Yang X, Wang J, Chen J, Wang J. Wogonin reduces cardiomyocyte apoptosis from mitochondrial release of cytochrome c to improve doxorubicin‑induced cardiotoxicity. Exp Ther Med 2022; 23:205. [PMID: 35126708 PMCID: PMC8796616 DOI: 10.3892/etm.2022.11128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/29/2021] [Indexed: 11/05/2022] Open
Affiliation(s)
- Yunjie Wei
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Junhao Zhao
- The First Clinical College, Jinyun Mountain Campus of Chongqing Medical University, Chongqing 401331, P.R. China
| | - Jian Xiong
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Jingjing Chai
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Xi Yang
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Junfeng Wang
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Jiajuan Chen
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Jing Wang
- Department of Neurology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| |
Collapse
|
9
|
Du S, Shi H, Xiong L, Wang P, Shi Y. Canagliflozin mitigates ferroptosis and improves myocardial oxidative stress in mice with diabetic cardiomyopathy. Front Endocrinol (Lausanne) 2022; 13:1011669. [PMID: 36313744 PMCID: PMC9616119 DOI: 10.3389/fendo.2022.1011669] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/27/2022] [Indexed: 11/13/2022] Open
Abstract
Canagliflozin (Cana), an anti-diabetes drug belongs to sodium-glucose cotransporter 2 inhibitor, is gaining interest because of its extra cardiovascular benefits. Ferroptosis is a new mode of cell death, which can promote the occurrence of diabetic cardiomyopathy (DCM). Whether Cana can alleviate DCM by inhibiting ferroptosis is the focus of this study. Here, we induced DCM models in diabetic C57BL6 mice and treated with Cana. Meanwhile, in order to exclude its hypoglycemic effect, the high glucose model in H9C2 cells were established. In the in vivo study, we observed that Cana could effectively alleviate the damage of cardiac function in DCM mice, including the increasing of lactate dehydrogenase (LDH) and cardiac troponin I (cTnI), the alleviating of myocardial fiber breakage, inflammation, collagen fiber deposition and mitochondrial structural disorder. We evaluated reactive oxygen species (ROS) levels by DCFH-DA and BODIPY 581/591 C11, in vitro Cana reduced ROS and lipid ROS in H9C2 cells induced by high glucose. Meanwhile, JC-1 fluorochrome assay showed that the decreased mitochondrial membrane potential (MMP) was increased by Cana. Furthermore, the inhibitory effects of Cana on myocardial oxidative stress and ferroptosis were verified in vivo and in vitro by protein carbonyl (PCO), malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), glutathione (GSH). As a key inducer of ferroptosis, the deposition of total iron and Fe2+ can be inhibited by Cana both in vivo and in vitro. In addition, western blot results indicated that the expression of ferritin heavy-chain (FTN-H) was down-regulated, and cystine-glutamate antiporter (xCT) was up-regulated by Cana in DCM mice and cells, suggesting that Cana inhibit ferroptosis by balancing cardiac iron homeostasis and promoting the system Xc-/GSH/GPX4 axis in DCM. These findings underscore the fact that ferroptosis plays an important role in the development and progression of DCM and targeting ferroptosis may be a novel strategy for prevention and treatment. In conclusion, Cana may exert some of its cardiovascular benefits by attenuating ferroptosis.
Collapse
Affiliation(s)
- Shuqin Du
- Central Laboratory of Molecular Medicine Research Center, Jiaxing Traditional Chinese Medicine (TCM) Hospital Affiliated to Zhejiang University of Traditional Chinese Medicine, Jiaxing, China
- Jiaxing Key Laboratory of Diabetic Angiopathy Research, Jiaxing, China
- School of Pharmacy, Zhejiang University of Technology, Hangzhou, China
- School of Medicine, Jiaxing University, Jiaxing, China
| | - Hanqiang Shi
- Central Laboratory of Molecular Medicine Research Center, Jiaxing Traditional Chinese Medicine (TCM) Hospital Affiliated to Zhejiang University of Traditional Chinese Medicine, Jiaxing, China
- Jiaxing Key Laboratory of Diabetic Angiopathy Research, Jiaxing, China
| | - Lie Xiong
- Central Laboratory of Molecular Medicine Research Center, Jiaxing Traditional Chinese Medicine (TCM) Hospital Affiliated to Zhejiang University of Traditional Chinese Medicine, Jiaxing, China
- Jiaxing Key Laboratory of Diabetic Angiopathy Research, Jiaxing, China
| | - Ping Wang
- School of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Yanbo Shi
- Central Laboratory of Molecular Medicine Research Center, Jiaxing Traditional Chinese Medicine (TCM) Hospital Affiliated to Zhejiang University of Traditional Chinese Medicine, Jiaxing, China
- Jiaxing Key Laboratory of Diabetic Angiopathy Research, Jiaxing, China
- *Correspondence: Yanbo Shi,
| |
Collapse
|
10
|
Tang H, Zhao J, Feng R, Pu P, Wen L. Reducing oxidative stress may be important for treating pirarubicin-induced cardiotoxicity with schisandrin B. Exp Ther Med 2021; 23:68. [PMID: 34934439 PMCID: PMC8649856 DOI: 10.3892/etm.2021.10991] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/08/2021] [Indexed: 12/22/2022] Open
Abstract
The cardiotoxicity of pirarubicin (THP) seriously affects its clinical application, which cannot be ignored. The antioxidant effect of schisandrin B (SchB) has been extensively reported in the context of dietotherapy. However, whether this antioxidant effect can protect the heart from THP damage remains unknown. The aim of the present study was to investigate whether the antioxidant effect of SchB can antagonize the cardiotoxicity of THP. Changes in electrocardiogram (ECG), echocardiography and serum lactate dehydrogenase, brain natriuretic peptide, creatine kinase MB and cardiac troponin T levels were used to detect the degree of cardiac damage. The levels of superoxide dismutase (SOD), malondialdehyde, catalase and total antioxidant capacity in the serum and heart were measured to observe the oxidative stress state of rats. Primary cardiomyocytes were cultured, and cell viability and reactive oxygen species (ROS) production were detected. Western blotting was used to detect the expression levels of SOD2, NOX2, pro/cleaved-caspase3 and Bcl-2/Bax in heart tissue and primary cardiomyocytes to verify the related signaling pathways. THP-treated rats showed a range of cardiac damage, including an abnormal ECG, echocardiography and myocardial enzymes. In the cellular experiments, cell viability decreased and ROS increased. However, this damage was alleviated after SchB treatment. Further studies demonstrated that SchB antagonized THP cardiotoxicity via its antioxidant effect. In conclusion, SchB protects the heart from THP damage in rats, and the mechanism may be closely associated with its antioxidant effect.
Collapse
Affiliation(s)
- Heng Tang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, P.R. China
| | - Junhao Zhao
- The First Clinical College, Jinyun Mountain Campus of Chongqing Medical University, Chongqing 401331, P.R. China
| | - Rui Feng
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, P.R. China
| | - Peng Pu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, P.R. China
| | - Li Wen
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, P.R. China
| |
Collapse
|