1
|
Huang J, Yang J, Yang Y, Lu X, Xu J, Lu S, Pan H, Zhou W, Li W, Chen S. Mitigating Doxorubicin-Induced Cardiotoxicity and Enhancing Anti-Tumor Efficacy with a Metformin-Integrated Self-Assembled Nanomedicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415227. [PMID: 40052211 PMCID: PMC12061326 DOI: 10.1002/advs.202415227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/06/2025] [Indexed: 05/10/2025]
Abstract
Doxorubicin (Dox) is a potent chemotherapeutic agent commonly used in cancer treatment. However, cardiotoxicity severely limited its clinical application. To address this challenge, a novel self-assembled nanomedicine platform, PMDDH, is developed for the co-delivery of Dox and metformin, an antidiabetic drug with cardioprotective and anti-tumor properties. PMDDH integrates metformin into a polyethyleneimine-based bioactive excipient (PMet), with Dox intercalated into double-stranded DNA and a hyaluronic acid (HA) coating to enhance tumor targeting. The PMDDH significantly improves the pharmacokinetics and tumor-targeting capabilities of Dox, while metformin enhances the drug's anti-tumor activity by downregulating programmed cell death ligand 1 (PD-L1) and activating the AMP-activated protein kinase (AMPK) signaling pathway. Additionally, the DNA component stimulates the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway, which synergizes with Dox-induced immunogenic cell death (ICD) to promote a robust anti-tumor immune response. PMDDH markedly reduces Dox-induced cardiotoxicity by preserving mitochondrial function, reducing reactive oxygen species (ROS) production, and inducing protective autophagy in cardiomyocytes. These findings position PMDDH as a promising dual-function nanomedicine that enhances the anti-tumor efficacy of Dox while minimizing its systemic toxicity, offering a safer and more effective alternative for cancer therapy.
Collapse
Affiliation(s)
- Jiaxin Huang
- Department of PharmacySecond Xiangya HospitalCentral South UniversityChangshaHunan410011China
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Jieru Yang
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Yuanying Yang
- Department of PharmacySecond Xiangya HospitalCentral South UniversityChangshaHunan410011China
| | - Xiaofeng Lu
- Department of CardiologyShanghai General HospitalShanghai Jiao Tong University School of MedicineNo.100, Haining RdShanghai200080China
- Department of CardiologyShanghai General Hospital Jiuquan HospitalNo. 22, West StJiuquanGansu735000China
| | - Juan Xu
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
- Department of CardiologyShanghai General HospitalShanghai Jiao Tong University School of MedicineNo.100, Haining RdShanghai200080China
| | - Shan Lu
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Hong Pan
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
- Hunan Key Laboratory of The Research and Development of Novel Pharmaceutical PreparationsSchool of Pharmaceutical ScienceChangsha Medical UniversityChangshaHunan410219China
| | - Wenqun Li
- Department of PharmacySecond Xiangya HospitalCentral South UniversityChangshaHunan410011China
| | - Songwen Chen
- Department of CardiologyShanghai General HospitalShanghai Jiao Tong University School of MedicineNo.100, Haining RdShanghai200080China
| |
Collapse
|
2
|
Prabhune NM, Ameen B, Prabhu S. Therapeutic potential of synthetic and natural iron chelators against ferroptosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3527-3555. [PMID: 39601820 DOI: 10.1007/s00210-024-03640-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024]
Abstract
Ferroptosis, a regulated form of cell death, is characterized by iron accumulation that results in the production of reactive oxygen species. This further causes lipid peroxidation and damage to the cellular components, eventually culminating into oxidative stress. Recent studies have highlighted the pivotal role of ferroptosis in the pathophysiological development and progression of various diseases such as β-thalassemia, hemochromatosis, and neurodegenerative disorders like AD and PD. Extensive efforts are in progress to understand the molecular mechanisms governing the role of ferroptosis in these conditions, and chelation therapy stands out as a potential approach to mitigate ferroptosis and its related implications in their development. There are currently both synthetic and natural iron chelators that are being researched for their potential as ferroptosis inhibitors. While synthetic chelators are relatively well-established and studied, their short plasma half-life and toxic side effects necessitate the exploration and identification of natural products that can act as efficient and safe iron chelators. In this review, we comprehensively discuss both synthetic and natural iron chelators as potential therapeutic strategies against ferroptosis-induced pathologies.
Collapse
Affiliation(s)
- Nupura Manish Prabhune
- Department of Cellular and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Bilal Ameen
- Department of Cellular and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Sudharshan Prabhu
- Department of Cellular and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
3
|
Zhang H, Wang Y, Wang R, Yi Q, Xu H, Tan B, Zhu J. Kartogenin Improves Doxorubicin-Induced Cardiotoxicity by Alleviating Oxidative Stress and Protecting Mitochondria. Int J Mol Sci 2025; 26:2434. [PMID: 40141078 PMCID: PMC11942266 DOI: 10.3390/ijms26062434] [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: 02/03/2025] [Revised: 02/27/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Doxorubicin (DOX) is a common antitumor drug in clinical practice, but its clinical use is limited due to its cardiotoxic side effects. Oxidative stress and mitochondrial damage are involved in DOX-induced cardiotoxicity (DIC). Kartogenin (KGN) has been shown to have a potent ability to resist oxidative stress and maintain mitochondrial homeostasis. But the impact of KGN on DIC has not been reported. This study explores the potential protective effect of KGN on DIC. The effect of KGN on DIC was studied by establishing in vivo and in vitro DIC models. KGN reduced DOX-induced cardiac insufficiency, myocardial injury, oxidative stress damage, and mitochondrial dysfunction. Through network pharmacology and RNA sequencing (RNA-seq), the mechanism of KGN anti-DIC was highly correlated with oxidative stress and mitochondria. These findings suggest that KGN is a valuable and promising strategy for the prevention of doxorubicin cardiotoxicity.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Key Laboratory of Children’s Vital Organ Development and Diseases of Chongqing Health Commission, Chongqing 400014, China
| | - Yunpeng Wang
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Key Laboratory of Children’s Vital Organ Development and Diseases of Chongqing Health Commission, Chongqing 400014, China
| | - Rui Wang
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Key Laboratory of Children’s Vital Organ Development and Diseases of Chongqing Health Commission, Chongqing 400014, China
| | - Qin Yi
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Key Laboratory of Children’s Vital Organ Development and Diseases of Chongqing Health Commission, Chongqing 400014, China
| | - Hao Xu
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Key Laboratory of Children’s Vital Organ Development and Diseases of Chongqing Health Commission, Chongqing 400014, China
- Department of Clinical Laboratory, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Bin Tan
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Key Laboratory of Children’s Vital Organ Development and Diseases of Chongqing Health Commission, Chongqing 400014, China
| | - Jing Zhu
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Key Laboratory of Children’s Vital Organ Development and Diseases of Chongqing Health Commission, Chongqing 400014, 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
|
Sun ML, Dong JM, Liu C, Li P, Zhang C, Zhen J, Chen W. Metformin-mediated protection against doxorubicin-induced cardiotoxicity. Biomed Pharmacother 2024; 180:117535. [PMID: 39405911 DOI: 10.1016/j.biopha.2024.117535] [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: 03/14/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 11/14/2024] Open
Abstract
BACKGROUND A phase II clinical trial of metformin (MET) for the treatment of doxorubicin (DOX)-induced cardiotoxicity (NCT02472353) failed. OBJECTIVES The aims of this study were to confirm MET-mediated protection against DOX-induced cardiotoxicity and its mechanism using H9C2 cells, and to establish a Wistar rat model of DOX-induced cardiotoxicity. Subsequently, Wistar rats were utilized to identify clinically relevant indicators for evaluating MET-mediated protection against DOX-induced cardiotoxicity, thereby facilitating early transition towards successful clinical trials. METHODS MET-mediated protection was assessed using cell viability and cytotoxicity experiments. Additionally, intramitochondrial reactive oxygen species (ROS) levels were measured using an ROS fluorescent probe (dihydroethidium) to confirm the oxidative stress mechanism. Eighteen Wistar rats were randomly allocated to the control, DOX, and DOX+MET groups; and the body weight, adverse drug reactions (ADRs), myocardial injury, cardiac function, oxidative stress, and histopathology of heart tissues were compared between groups. RESULTS H9C2 cells treated with MET/Dexrazoxane demonstrated dose-dependent protection against DOX-induced cardiotoxicity. The fluorescence intensity of H9C2 cells suggested DOX-induced cardiomyocyte toxicity and MET-mediated protection against DOX-induced cardiotoxicity. In vivo experiments confirmed that a rat model of DOX-induced cardiotoxicity was successfully established, but MET-mediated protection against DOX-induced cardiotoxicity was not demonstrated. This was attributed to insufficient energy intake because of ADRs, such as vomiting. CONCLUSIONS We confirmed the MET-mediated protection against DOX-induced cardiomyocyte toxicity and its mechanism involving the inhibition of oxidative stress in vitro experiments. It is imperative to investigate the optimal conditions for MET-mediated protection against DOX-induced cardiotoxicity in vivo or clinical trials.
Collapse
Affiliation(s)
- Ming-Li Sun
- Phase I Clinical Trial Research Center, Beijing Shijitan Hospital Affiliated to Capital Medical University, Beijing 100038, China.
| | - Jun-Min Dong
- Phase I Clinical Trial Research Center, Beijing Shijitan Hospital Affiliated to Capital Medical University, Beijing 100038, China
| | - Chen Liu
- Phase I Clinical Trial Research Center, Beijing Shijitan Hospital Affiliated to Capital Medical University, Beijing 100038, China
| | - Pu Li
- Phase I Clinical Trial Research Center, Beijing Shijitan Hospital Affiliated to Capital Medical University, Beijing 100038, China
| | - Chao Zhang
- Department of Ultrasonography, Beijing Chaoyang Hospital affiliated to Capital Medical University, Beijing 100020, China
| | - Jie Zhen
- Department of Intensive Care Unit, Beijing Shijitan Hospital Affiliated to Capital Medical University, Beijing 100038, China
| | - Wei Chen
- Department of Intensive Care Unit, Beijing Shijitan Hospital Affiliated to Capital Medical University, Beijing 100038, China.
| |
Collapse
|
6
|
Lu LQ, Li MR, Huang LL, Che YX, Qi YN, Luo XJ, Peng J. Micafungin protects mouse heart against doxorubicin-induced oxidative injury via suppressing MALT1-dependent k48-linked ubiquitination of Nrf2. Chem Biol Interact 2024; 400:111179. [PMID: 39089415 DOI: 10.1016/j.cbi.2024.111179] [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: 06/07/2024] [Revised: 07/27/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024]
Abstract
Oxidative stress contributes greatly to doxorubicin (DOX)-induced cardiotoxicity. Down-regulation of nuclear factor erythroid 2-related factor 2 (Nrf2) is a key factor in DOX-induced myocardial oxidative injury. Recently, we found that mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1)-dependent k48-linked ubiquitination was responsible for down-regulation of myocardial Nrf2 in DOX-treated mice. Micafungin, an antifungal drug, was identified as a potential MALT1 inhibitor. This study aims to explore whether micafungin can reduce DOX-induced myocardial oxidative injury and if its anti-oxidative effect involves a suppression of MALT1-dependent k48-linked ubiquitination of Nrf2. To establish the cardiotoxicity models in vivo and in vitro, mice were treated with a single dose of DOX (15 mg/kg, i.p.) and cardiomyocytes were incubated with DOX (1 μM) for 24 h, respectively. Using mouse model of DOX-induced cardiotoxicity, micafungin (10 or 20 mg/kg) was shown to improve cardiac function, concomitant with suppression of oxidative stress, mitochondrial dysfunction, and cell death in a dose-dependent manner. Similar protective roles of micafungin (1 or 5 μM) were observed in DOX-treated cardiomyocytes. Mechanistically, micafungin weakened the interaction between MALT1 and Nrf2, decreased the k48-linked ubiquitination of Nrf2 while elevated the protein levels of Nrf2 in both DOX-treated mice and cardiomyocytes. Furthermore, MALT1 overexpression counteracted the cardioprotective effects of micafungin. In conclusion, micafungin reduces DOX-induced myocardial oxidative injury via suppression of MALT1, which decreases the k48-linked ubiquitination of Nrf2 and elevates Nrf2 protein levels. Thus, micafungin may be repurposed for treating DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Li-Qun Lu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Ming-Rui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Lin-Lu Huang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Yan-Xi Che
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Ya-Nan Qi
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China; Hunan Provincial Key Laboratory of Cardiovascular Research, School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China.
| |
Collapse
|
7
|
Sun ML, Chen W, Wang XH. Reliability of Metformin's protective effects against doxorubicin-induced cardiotoxicity: a meta-analysis of animal studies. Front Pharmacol 2024; 15:1435866. [PMID: 39175538 PMCID: PMC11338926 DOI: 10.3389/fphar.2024.1435866] [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: 05/21/2024] [Accepted: 07/24/2024] [Indexed: 08/24/2024] Open
Abstract
Background The protective effects of metformin (Met) against doxorubicin (Dox)-induced cardiotoxicity via potential hypotheses of mechanisms of action with unknown reliability and credibility. Objectives This study aimed to investigate the protective effects of Met against Dox-induced cardiotoxicity and the underlying mechanisms of action, as well as examine their reliability and credibility. Methods A comprehensive search was conducted within the PubMed, Embase, Web of Science, Science Direct, Scopus, and CNKI databases from inception to 31 December 2023. Animal experiments evaluating the efficacy of Met against Dox-induced cardiotoxicity were included in this study. The primary efficacy outcomes were markers of myocardial injury. Effect size was measured using the standardized mean difference for continuous variables. Data were pooled using a random-effects model in the Stata 18 statistical software package. Results Twenty-one studies involving 203-208 animals treated with Dox and 271-276 animals treated with Dox and Met were included in this analysis. Quality assessment revealed high-quality scores. Pooled results favored Met treatment based on the serum lactate dehydrogenase (LDH), creatine kinase-myocardial band (CK-MB), cardiac troponin I (cTnI), and aspartate aminotransferase levels. Sensitivity analysis using the leave-one-out method demonstrated stable results. Funnel plots, Egger's test, and Begg's test confirmed potential publication bias. The oxidative stress hypothesis has been investigated extensively based on abundant evidence. Conclusion Met is effective and safe for protecting against Dox-induced cardiotoxicity, thus making it an appropriate drug for clinical investigation. The oxidative stress hypothesis of mechanism of action is well established with highest reliability and credibility.
Collapse
Affiliation(s)
- Ming-Li Sun
- Phase I Clinical Trial Research Center, Beijing Shijitan Hospital Affiliated to Capital Medical University, Beijing, China
| | - Wei Chen
- Department of Intensive Care Unit, Beijing Shijitan Hospital Affiliated to Capital Medical University, Beijing, China
| | - Xing-He Wang
- Phase I Clinical Trial Research Center, Beijing Shijitan Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Wang F, Yan X, Yue A, Zhang K, Li P, Xu J, Sun K, Zhang Q, Li Y. Apigenin alleviates doxorubicin-induced myocardial pyroptosis by inhibiting glycogen synthase kinase-3β in vitro and in vivo. Drug Dev Res 2024; 85:e22196. [PMID: 38812449 DOI: 10.1002/ddr.22196] [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: 11/29/2023] [Revised: 03/01/2024] [Accepted: 05/03/2024] [Indexed: 05/31/2024]
Abstract
Apigenin, a natural flavonoid compound found in chamomile (Matricaia chamomilla L.) from the Asteraceae family, has been shown in our previous study to possess antimyocardial hypertrophy and anti-cardiac fibrosis effects. However, its effects and mechanisms on the pyroptosis of cardiomyocytes induced by doxorubicin (DOX) are poorly understood. The objective of this study was to investigate the role of GSK-3β and the effects of apigenin in DOX-induced cardiotoxicity. H9c2 cells stimulated with DOX were treated with SB216763 and apigenin. Additionally, a mouse model of DOX-induced cardiotoxicity was prepared and further treated with apigenin and SB216763 for 30 days. The findings revealed that treatment with SB216763 or apigenin resulted in a significant reduction in the levels of pyroptosis-related factors. Furthermore, the phosphorylation of GSK-3β was enhanced while the phosphorylation of nuclear factor-kB (NF-κB) p65 was reduced following treatment with either SB216763 or apigenin. Conversely, the effects of apigenin treatment were nullified in siRNA-GSK-3β-transfected cells. Results from computer simulation and molecular docking analysis supported that apigenin could directly target the regulation of GSK-3β. Therefore, our study confirmed that the inhibition of GSK-3β and treatment with apigenin effectively suppressed the pyroptosis of cardiomyocytes in both DOX-stimulated H9c2 cells and mice. These benefits may be attributed in part to the decrease in GSK-3β expression and subsequent reduction in NF-κB p65 activation. Overall, our findings revealed that the pharmacological targeting of GSK-3β may offer a promising therapeutic approach for alleviating DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Feng Wang
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Xinxin Yan
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Anna Yue
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Kaiyu Zhang
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Ping Li
- Department of Central Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Jingyi Xu
- Department of Central Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Kangyun Sun
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Qian Zhang
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Yuan Li
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| |
Collapse
|
9
|
Zhang B, Li Y, Liu N, Liu B. AP39, a novel mitochondria-targeted hydrogen sulfide donor ameliorates doxorubicin-induced cardiotoxicity by regulating the AMPK/UCP2 pathway. PLoS One 2024; 19:e0300261. [PMID: 38568919 PMCID: PMC10990198 DOI: 10.1371/journal.pone.0300261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 02/25/2024] [Indexed: 04/05/2024] Open
Abstract
Doxorubicin (DOX) is a broad-spectrum, highly effective antitumor agent; however, its cardiotoxicity has greatly limited its use. Hydrogen sulfide (H2S) is an endogenous gaseous transmitter that exerts cardioprotective effects via the regulation of oxidative stress and apoptosis and maintenance of mitochondrial function, among other mechanisms. AP39 is a novel mitochondria-targeted H2S donor that, at appropriate concentrations, attenuates intracellular oxidative stress damage, maintains mitochondrial function, and ameliorates cardiomyocyte injury. In this study, DOX-induced cardiotoxicity models were established using H9c2 cells and Sprague-Dawley rats to evaluate the protective effect of AP39 and its mechanisms of action. Both in vivo and in vitro experiments showed that DOX induces oxidative stress injury, apoptosis, and mitochondrial damage in cardiomyocytes and decreases the expression of p-AMPK/AMPK and UCP2. All DOX-induced changes were attenuated by AP39 treatment. Furthermore, the protective effect of AP39 was significantly attenuated by the inhibition of AMPK and UCP2. The results suggest that AP39 ameliorates DOX-induced cardiotoxicity by regulating the expression of AMPK/UCP2.
Collapse
Affiliation(s)
- Bin Zhang
- The Second Hospital of Jilin University, Nanguan District, Changchun City, Jilin Province, China
| | - Yangxue Li
- The Second Hospital of Jilin University, Nanguan District, Changchun City, Jilin Province, China
| | - Ning Liu
- The Second Hospital of Jilin University, Nanguan District, Changchun City, Jilin Province, China
| | - Bin Liu
- The Second Hospital of Jilin University, Nanguan District, Changchun City, Jilin Province, China
| |
Collapse
|
10
|
Huang C, Guo Y, Li T, Sun G, Yang J, Wang Y, Xiang Y, Wang L, Jin M, Li J, Zhou Y, Han B, Huang R, Qiu J, Tan Y, Hu J, Wei Y, Wu B, Mao Y, Lei L, Song X, Li S, Wang Y, Zhang T. Pharmacological activation of GPX4 ameliorates doxorubicin-induced cardiomyopathy. Redox Biol 2024; 70:103024. [PMID: 38232458 PMCID: PMC10827549 DOI: 10.1016/j.redox.2023.103024] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 12/31/2023] [Accepted: 12/31/2023] [Indexed: 01/19/2024] Open
Abstract
Due to the cardiotoxicity of doxorubicin (DOX), its clinical application is limited. Lipid peroxidation caused by excessive ferrous iron is believed to be a key molecular mechanism of DOX-induced cardiomyopathy (DIC). Dexrazoxane (DXZ), an iron chelator, is the only drug approved by the FDA for reducing DIC, but it has many side effects and cannot be used as a preventive drug in clinical practice. Single-nucleus RNA sequencing (snRNA-seq) analysis identified myocardial and epithelial cells that are susceptible to DOX-induced ferroptosis. The glutathione peroxidase 4 (GPX4) activator selenomethione (SeMet) significantly reduced polyunsaturated fatty acids (PUFAs) and oxidized lipid levels in vitro. Consistently, SeMet significantly decreased DOX-induced lipid peroxidation in H9C2 cells and mortality in C57BL/6 mice compared to DXZ, ferrostatin-1, and normal saline. SeMet can effectively reduce serum markers of cardiac injury in C57BL/6 mice and breast cancer patients. Depletion of the GPX4 gene in C57BL/6 mice resulted in an increase in polyunsaturated fatty acid (PUFA) levels and eliminated the protective effect of SeMet against DIC. Notably, SeMet exerted antitumor effects on breast cancer models with DOX while providing cardiac protection for the same animal without detectable toxicities. These findings suggest that pharmacological activation of GPX4 is a valuable and promising strategy for preventing the cardiotoxicity of doxorubicin.
Collapse
Affiliation(s)
- Chuying Huang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, 445000, China; Hubei Provincial Key Lab of Selenium Resources and Bioapplications, Enshi, 445000, China.
| | - Yishan Guo
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Department of Cardiology, Binzhou Medical University Hospital, Binzhou, 256600, China
| | - Tuo Li
- Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, 445000, China; Hubei Provincial Key Lab of Selenium Resources and Bioapplications, Enshi, 445000, China
| | - Guogen Sun
- Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, 445000, China; Hubei Provincial Key Lab of Selenium Resources and Bioapplications, Enshi, 445000, China
| | - Jinru Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yuqi Wang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Ying Xiang
- Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, 445000, China; Hubei Provincial Key Lab of Selenium Resources and Bioapplications, Enshi, 445000, China
| | - Li Wang
- Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, 445000, China; Hubei Provincial Key Lab of Selenium Resources and Bioapplications, Enshi, 445000, China
| | - Min Jin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiao Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yong Zhou
- Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, 445000, China
| | - Bing Han
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Rui Huang
- Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, 445000, China; Hubei Provincial Key Lab of Selenium Resources and Bioapplications, Enshi, 445000, China
| | - Jiao Qiu
- Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, 445000, China; Hubei Provincial Key Lab of Selenium Resources and Bioapplications, Enshi, 445000, China
| | - Yong Tan
- Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, 445000, China; Hubei Provincial Key Lab of Selenium Resources and Bioapplications, Enshi, 445000, China
| | - Jiaxing Hu
- Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, 445000, China; Hubei Provincial Key Lab of Selenium Resources and Bioapplications, Enshi, 445000, China
| | - Yumiao Wei
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bo Wu
- Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, 445000, China; Hubei Provincial Key Lab of Selenium Resources and Bioapplications, Enshi, 445000, China
| | - Yong Mao
- Wuhan Frasergen Bioinformatics Co. Ltd., Wuhan, 430070, China
| | - Lingshan Lei
- Wuhan Frasergen Bioinformatics Co. Ltd., Wuhan, 430070, China
| | - Xiusheng Song
- Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, 445000, China; Hubei Provincial Key Lab of Selenium Resources and Bioapplications, Enshi, 445000, China
| | - Shuijie Li
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
| | - Yongsheng Wang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, Sichuan University, West China Hospital, Chengdu, 610041, China.
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
11
|
Li K, Chen W, Ma L, Yan L, Wang B. Approaches for reducing chemo/radiation-induced cardiotoxicity by nanoparticles. ENVIRONMENTAL RESEARCH 2024; 244:117264. [PMID: 37776941 DOI: 10.1016/j.envres.2023.117264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 09/13/2023] [Accepted: 09/27/2023] [Indexed: 10/02/2023]
Abstract
Nanoparticles are fascinating and encouraging carriers for cancer treatment due to their extraordinary properties and potential applications in targeted drug delivery, treatment, and diagnosis. Experimental studies including in vitro and in vivo examinations show that nanoparticles can cause a revolution in different aspects of cancer therapy. Normal tissue toxicity and early and late consequences are the major limitations of cancer therapy by radiotherapy and chemotherapy. However, the delivery of drugs into tumors or reducing the accumulation of drugs in normal tissues can permit a more satisfactory response of malignancies to therapy with more inferior side effects. Cardiac toxicity is one of the major problems for chemotherapy and radiotherapy. Therefore, several experimental studies have been performed to minimize the degenerative impacts of cancer treatment on the heart and also enhance the influences of radiotherapy and chemotherapy agents in cancers. This review article emphasizes the benefits of nanoparticle-based drug delivery techniques, including minimizing the exposure of the heart to anticancer drugs, enhancing the accumulation of drugs in cancers, and expanding the effectiveness of radiotherapy. The article also discusses the challenges and problems accompanied with nanoparticle-based drug delivery techniques such as toxicity, which need to be addressed through further research. Moreover, the article emphasizes the importance of developing safe and effective nanoparticle-based therapies that can be translated into clinical practice.
Collapse
Affiliation(s)
- Ketao Li
- Department of Cardiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang, 310022, China
| | - Wan Chen
- Department of Cardiology, Jiulongpo First People's Hospital, Chongqing, 400051, China
| | - Liping Ma
- Department of Cardiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang, 310022, China
| | - Laixing Yan
- Department of Cardiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang, 310022, China
| | - Bing Wang
- Department of Cardiology, Zouping People's Hospital, Zouping, shandong, 256299, China.
| |
Collapse
|
12
|
Chen L, Sun X, Wang Z, Chen M, He Y, Zhang H, Han D, Zheng L. Resveratrol protects against doxorubicin-induced cardiotoxicity by attenuating ferroptosis through modulating the MAPK signaling pathway. Toxicol Appl Pharmacol 2024; 482:116794. [PMID: 38142782 DOI: 10.1016/j.taap.2023.116794] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
Doxorubicin (Dox) is a widely used antitumor agent with dose-dependent and cumulative cardiotoxic effects. Resveratrol (Res) is a natural non-flavonoid polyphenol that can potentially provide cardiovascular benefits. We aimed to estimate the protective effect of Res on Dox-induced cardiotoxicity (DIC) and explore whether it was related to attenuating ferroptosis. We established DIC models in C57BL/6 J mice, H9C2 cardiomyoblasts, and neonatal rat cardiomyocytes (NRCMs). We further treated H9C2 cells with RSL3, a ferroptosis agonist, to investigate whether Res exerted protective effects through inhibiting ferroptosis. Ferrostatin-1 (Fer-1) was applied to suppress ferroptosis. Dox treatment caused cardiac dysfunction and resulted in apparent ferroptotic damage in cardiac tissue, involving increased iron accumulation, glutathione depletion, increased expression of ferroptosis-related proteins, and decreased expression of glutathione peroxidase 4, which were alleviated by Fer-1 and Res administration. These findings were also confirmed in Dox-treated H9C2 cells and NRCMs, with Fer-1 and Res effectively attenuating Dox-induced cytotoxicity and ferroptosis. Furthermore, Res protected H9C2 cells from RSL3-induced ferroptotic cell death, and the protective effect was similar to that of Fer-1. Both Dox and RSL3 treatment increased the phosphorylation levels of mitogen-activated protein kinases (MAPKs), including extracellular signal-regulated kinase, p38, and c-Jun N-terminal kinases; however, these changes were hindered by Res. This study demonstrates that Res effectively alleviates DIC by suppressing ferroptosis possibly through modulating the MAPK signaling pathway. Our results highlight that targeting ferroptosis can be a potential cardioprotective strategy for DIC.
Collapse
Affiliation(s)
- Lu Chen
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Xingang Sun
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Zhen Wang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Miao Chen
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Yuxian He
- Cardiovascular Medicine Department, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, 158 Shangtang Road, Hangzhou 310014, Zhejiang, China
| | - Han Zhang
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Deheng Han
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Liangrong Zheng
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China.
| |
Collapse
|
13
|
Stansfeld A, Radia U, Goggin C, Mahalingam P, Benson C, Napolitano A, Jones RL, Rosen SD, Karavasilis V. Pharmacological strategies to reduce anthracycline-associated cardiotoxicity in cancer patients. Expert Opin Pharmacother 2022; 23:1641-1650. [DOI: 10.1080/14656566.2022.2124107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Anna Stansfeld
- Medical Oncology, The Royal Marsden Hospital NHS Foundation Trust and Institute of Cancer Research, UK
| | - Utsav Radia
- Medical Oncology, The Royal Marsden Hospital NHS Foundation Trust and Institute of Cancer Research, UK
| | - Caitriona Goggin
- Medical Oncology, The Royal Marsden Hospital NHS Foundation Trust and Institute of Cancer Research, UK
| | - Preethika Mahalingam
- Medical Oncology, The Royal Marsden Hospital NHS Foundation Trust and Institute of Cancer Research, UK
| | - Charlotte Benson
- Medical Oncology, The Royal Marsden Hospital NHS Foundation Trust and Institute of Cancer Research, UK
| | - Andrea Napolitano
- Medical Oncology, The Royal Marsden Hospital NHS Foundation Trust and Institute of Cancer Research, UK
| | - Robin L Jones
- Medical Oncology, The Royal Marsden Hospital NHS Foundation Trust and Institute of Cancer Research, UK
| | - Stuart D Rosen
- Cardiology, London North West University Healthcare NHS Trust and Royal Brompton Hospitals, UK
| | | |
Collapse
|
14
|
Jones IC, Dass CR. Doxorubicin-induced cardiotoxicity: causative factors and possible interventions. J Pharm Pharmacol 2022; 74:1677-1688. [PMID: 35994421 DOI: 10.1093/jpp/rgac063] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 08/03/2022] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Doxorubicin (Dox) belongs to the anthracycline drug classification and is a widely administered chemotherapeutic. However, Dox use in therapy is limited by its cardiotoxicity, representing a significant drawback of Dox treatment applicability. A large amount of current research is on reducing Dox-induced cardiotoxicity by developing targeted delivery systems and investigating cardiotoxicity mechanisms. Recently, discrepancies have challenged the traditional understanding of Dox metabolism, mechanisms of action and cardiotoxicity drivers. This review summarises the current knowledge around Dox's metabolism, mechanisms of anticancer activity, and delivery systems and offers a unique perspective on the relationships between several proposed mechanisms of Dox-induced cardiotoxicity. KEY FINDINGS While there is a strong understanding of Dox's pharmacokinetic properties, it is unclear which enzymes contribute to Dox metabolism and how Dox induces its cytotoxic effect in neoplastic and non-neoplastic cells. Evidence suggests that there are several potentially synergistic mechanisms involved in Dox-induced cardiotoxicity. SUMMARY It has become clear that Dox operates in a multifactorial fashion dependent on cellular context. Accumulation of oxidative stress appears to be a common factor in cardiotoxicity mechanisms, highlighting the importance of novel delivery systems and antioxidant therapies.
Collapse
Affiliation(s)
- Isobel C Jones
- Curtin Medical School, Bentley 6102, Australia.,Curtin Health Innovation Research Institute, Bentley 6102, Australia
| | - Crispin R Dass
- Curtin Medical School, Bentley 6102, Australia.,Curtin Health Innovation Research Institute, Bentley 6102, Australia
| |
Collapse
|
15
|
Li W, Qu X, Kang X, Zhang H, Zhang X, Hu H, Yao L, Zhang L, Zheng J, Zheng Y, Zhang J, Xu Y. Silibinin eliminates mitochondrial ROS and restores autophagy through IL6ST/JAK2/STAT3 signaling pathway to protect cardiomyocytes from doxorubicin-induced injury. Eur J Pharmacol 2022; 929:175153. [PMID: 35839932 DOI: 10.1016/j.ejphar.2022.175153] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/26/2022] [Accepted: 07/08/2022] [Indexed: 11/26/2022]
Abstract
Growing evidence indicates that silibinin (SLB), a main component extracted from Chinese herb Silybum marianum, can effectively antagonize doxorubicin (DOX) induced myocardial injury (DIMI), but the specific molecular mechanism is still unelucidated. Herein, DOX induced human AC16 cardiomyocyte injury model and Network Pharmacology are used to predict and verify the potential mechanism. The analysis results of the core PPI network of SLB against DIMI show that JAK/STAT signaling pathway and autophagy are significantly enriched. Molecular docking results indicate that SLB has stronger binding ability to signaling key proteins IL6ST, JAK2 and STAT3 (affinity ≤ -7.0 kcal/mol). The detection results of pathway activation and autophagy level demonstrate that SLB significantly alleviates DOX induced IL6ST/JAK2/STAT3 signaling pathway inhibition and autophagy inhibition, reduces the death rate of cardiomyocytes. This protective effect of SLB is eliminated when key pathway proteins (IL6ST, JAK2, STAT3) are knocked down or autophagy is inhibited (3-MA or Beclin1 knockdown). These results suggest that the regulation of IL6ST/JAK2/STAT3 signaling pathway and autophagy may be important mechanism for SLB's protective effect on DOX injured cardiomyocytes. Further experimental results prove that knockdown of IL6ST, JAK2 and STAT3 eliminate the mitochondrial ROS scavenging effect and autophagy promoting effect of SLB. In sum, SLB can decrease the mitochondrial ROS and restore autophagy to antagonize DOX-induced cardiomyocyte injury by activating IL6ST/JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Wenbiao Li
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xinni Qu
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiangping Kang
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Haiyin Zhang
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xueli Zhang
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Haiyan Hu
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lingai Yao
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lina Zhang
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jing Zheng
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yuejuan Zheng
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Jianghong Zhang
- Department of Radiation Biology, Institute of Radiation Medicine, Fudan University, Shanghai, 200032, China.
| | - Yanwu Xu
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
16
|
Li Z, Liu J, Sun Z, Li Y, Yu B, Zhao F, Wang H, Xu H. Nanomicelles co-loaded with doxorubicin and salvianolic acid A for breast cancer chemotherapy. Cancer Nanotechnol 2022. [DOI: 10.1186/s12645-022-00127-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Abstract
Background
Multi-drug delivery system based on polymer carrier is emerging for alleviating dose-limiting toxicities of first-line cytotoxic anticancer drugs, such as doxorubicin (DOX) for breast cancer chemotherapy. By co-loading the premium natural antioxidant salvianolic acid A (SAA) through colloidal self-assembly of amphiphilic copolymer, we herein developed CPMSD, a complex polymeric micellar system to overcome cardiotoxicity associated with DOX.
Results
Optimal formulation was obtained by DOE study and CPMSD micelles were well constructed by using mPEG-PCL for entrapment at a drug–carrier mass ratio of 1:5 and DOX–SAA mass ratio of 1:4. Molecular dynamics simulation revealed the ratiometrical co-encapsulation of SAA into the hydrophobic cavity but DOX to ball-shaped surface of micelles due to hydrophilicity. Characterization study manifested favorable biopharmaceutical properties, such as small and uniform particle size, fairly high drug loading capacity, as well as good colloidal stability and controlled drug release. CPMSD maintained anticancer efficacy of DOX and the action mechanism, which did not be affected by co-administering SAA. More to the point, it was of great benefit to systemic safety and cardioprotective effect against oxidative stress injuries associated with DOX in tumor-bearing mice.
Conclusions
All the findings substantiated that CPMSD would be a promising multifunctional nanosystem of DOX for breast cancer chemotherapy.
Collapse
|
17
|
Arifi S, Constantinidou A, Jones R. Managing the risk of toxicity in the treatment of elderly patients with soft tissue sarcomas. Expert Opin Drug Saf 2021; 20:903-913. [PMID: 33956569 DOI: 10.1080/14740338.2021.1915985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Nearly half of soft tissue sarcomas (STS) occur after the age of 65 years. Treating these patients is a complex issue in the absence of specific guidelines. AREAS COVERED This is a narrative review that summarizes current data on the efficacy and the safety of different treatment strategies in this subpopulation. EXPERT OPINION Age per se should not be a limiting factor to treatment. Surgery remains the treatment of choice offering the only chance of cure. The potential for benefit from adjuvant therapies must be discussed in the context of expected treatment-related toxicities and impairment of quality of life. Efficacy of systemic treatment in advanced disease did not differ from that in younger patients. However, safety must be considered when selecting treatments. Managing the risk of toxicity requires an assessment of vulnerabilities with validated tools. The Comprehensive geriatric assessment has become increasingly accepted but need to be validated in STS patients. Frailty should not exclude patients from potentially life-saving therapy. The correction of reversible conditions and active supportive care may make the treatment safer. Future studies are warranted to define better the patterns, benefits, risks of existing treatments. New options remain to be identified to reduce toxicity.
Collapse
Affiliation(s)
- Samia Arifi
- Medical Oncology Department, Hassan II University hospital/Faculty of Medicine and Pharmacy. University of Sidi Mohamed Ben Abdellah. Fez, Morocco
| | - Anastasia Constantinidou
- Medical School, University of Cyprus, Nicosia, Cyprus.,Bank of Cyprus Oncology Centre, Nicosia, Cyprus
| | - Robin Jones
- Sarcoma Unit, Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, UK
| |
Collapse
|
18
|
Spalato M, Italiano A. The safety of current pharmacotherapeutic strategies for osteosarcoma. Expert Opin Drug Saf 2021; 20:427-438. [PMID: 33478264 DOI: 10.1080/14740338.2021.1881060] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Introduction: Peri-operative chemotherapy is the backbone of treatment for patients with osteosarcoma. Methotrexate, cisplatinum, doxorubicin and ifosfamide are the main drugs used in chemotherapy regimens used for osteosarcoma.Areas covered: We have reviewed here the relevant literature related to the incidence and management of acute and late toxicities of systemic treatment used for the management of patients with osteosarcoma.Expert opinion: Early diagnosis and appropriate management of acute and late toxicities of chemotherapy is crucial for an efficient care of osteosarcoma patients. Although the incidence and management of chemotherapy-related acute toxicities are well known by most oncologists, the use of high doses of methotrexate have the potential to cause fatal toxicities and, therefore, needs careful monitoring. Moreover, the diagnosis of late toxicities is more challenging and requires long-term follow-up for an appropriate management.
Collapse
Affiliation(s)
| | - Antoine Italiano
- Sarcoma Unit, Institut Bergonié, Bordeaux, France.,University of Bordeaux, Faculty of Medicine, Bordeaux, France
| |
Collapse
|
19
|
Lódi M, Bánhegyi V, Bódi B, Gyöngyösi A, Kovács Á, Árokszállási A, Hamdani N, Fagyas M, Édes I, Csanádi Z, Czuriga I, Kisvárday Z, Lekli I, Bai P, Tóth A, Papp Z, Czuriga D. Prophylactic, single-drug cardioprotection in a comparative, experimental study of doxorubicin-induced cardiomyopathy. J Transl Med 2020; 18:470. [PMID: 33298102 PMCID: PMC7725221 DOI: 10.1186/s12967-020-02564-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 10/10/2020] [Indexed: 12/28/2022] Open
Abstract
Background Cardiomyopathy is a common side effect of doxorubicin (DOX) chemotherapy. Despite intensive research efforts in the field, there is still no evidence available for routine cardioprotective prophylaxis to prevent cardiotoxicity in the majority of oncological patients at low risk of cardiovascular disease. We have recently demonstrated the advantages of a prophylactic, combined heart failure therapy in an experimental model of DOX-induced cardiomyopathy. In the current work, we focus on individually applied prophylactic medications studied in the same translational environment to clarify their distinct roles in the prevention of DOX cardiotoxicity. Methods Twelve-week-old male Wistar rats were divided into 5 subgroups. Prophylactic β-blocker (BB, bisoprolol), angiotensin-converting enzyme inhibitor (ACEI, perindopril) or aldosterone antagonist (AA, eplerenone) treatments were applied 1 week before DOX administration, then 6 cycles of intravenous DOX chemotherapy were administered. Rats receiving only intravenous DOX or saline served as positive and negative controls. Blood pressure, heart rate, body weight, and echocardiographic parameters were monitored in vivo. Two months after the last DOX administration, the animals were sacrificed, and their heart and serum samples were frozen in liquid nitrogen for histological, mechanical, and biochemical measurements. Results All prophylactic treatments increased the survival of DOX-receiving animals. The lowest mortality rates were seen in the BB and ACEI groups. The left ventricular ejection fraction was only preserved in the BB group. The DOX-induced increase in the isovolumetric relaxation time could not be prevented by any prophylactic treatment. A decreased number of apoptotic nuclei and a preserved myocardial ultrastructure were found in all groups receiving prophylactic cardioprotection, while the DOX-induced fibrotic remodelling and the increase in caspase-3 levels could only be substantially prevented by the BB and ACEI treatments. Conclusion Primary prophylaxis with cardioprotective agents like BB or ACEI has a key role in the prevention of DOX-induced cardiotoxicity in healthy rats. Future human studies are necessary to implement this finding in the clinical management of oncological patients free of cardiovascular risk factors.
Collapse
Affiliation(s)
- Mária Lódi
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Viktor Bánhegyi
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Beáta Bódi
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Alexandra Gyöngyösi
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Árpád Kovács
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Anita Árokszállási
- Department of Oncology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Nazha Hamdani
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Miklós Fagyas
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - István Édes
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Csanádi
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - István Czuriga
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Kisvárday
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - István Lekli
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Péter Bai
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary
| | - Attila Tóth
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dániel Czuriga
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
20
|
Todorova VK, Siegel ER, Kaufmann Y, Kumarapeli A, Owen A, Wei JY, Makhoul I, Klimberg VS. Dantrolene Attenuates Cardiotoxicity of Doxorubicin Without Reducing its Antitumor Efficacy in a Breast Cancer Model. Transl Oncol 2020; 13:471-480. [PMID: 31918212 PMCID: PMC7031101 DOI: 10.1016/j.tranon.2019.12.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/02/2019] [Accepted: 12/03/2019] [Indexed: 02/07/2023] Open
Abstract
Dysregulation of calcium homeostasis is a major mechanism of doxorubicin (DOX)-induced cardiotoxicity. Treatment with DOX causes activation of sarcoplasmic reticulum (SR) ryanodine receptor (RYR) and rapid release of Ca2+ in the cytoplasm resulting in depression of myocardial function. The aim of this study was to examine the effect of dantrolene (DNT) a RYR blocker on both the cardiotoxicity and antitumor activity of DOX in a rat model of breast cancer. Female F344 rats with implanted MAT B III breast cancer cells were randomized to receive intraperitoneal DOX twice per week (12 mg/kg total dose), 5 mg/kg/day oral DNT or a combination of DOX + DNT for 3 weeks. Echocardiography and blood troponin I levels were used to measure myocardial injury. Hearts and tumors were evaluated for histopathological alterations. Blood glutathione was assessed as a measure of oxidative stress. The results showed that DNT improved DOX-induced alterations in the echocardiographic parameters by 50%. Histopathologic analysis of hearts showed reduced DOX induced cardiotoxicity in the group treated with DOX + DNT as shown by reduced interstitial edema, cytoplasmic vacuolization, and myofibrillar disruption, compared with DOX-only–treated hearts. Rats treated with DNT lost less body weight, had higher blood GSH levels and lower troponin I levels than DOX-treated rats. These data indicate that DNT is able to provide protection against DOX cardiotoxicity without reducing its antitumor activity. Further studies are needed to determine the optimal dosing of DNT and DOX in a tumor-bearing host.
Collapse
Affiliation(s)
- Valentina K Todorova
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, USA.
| | - Eric R Siegel
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Yihong Kaufmann
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Asangi Kumarapeli
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Aaron Owen
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Jeanne Y Wei
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Issam Makhoul
- Division of Medical Oncology, University of Arkansas for Medical Sciences, Little Rock, USA
| | - V Suzanne Klimberg
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, USA
| |
Collapse
|