1
|
Wang C, Fan P, Wang Q. Evolving therapeutics and ensuing cardiotoxicities in triple-negative breast cancer. Cancer Treat Rev 2024; 130:102819. [PMID: 39216183 DOI: 10.1016/j.ctrv.2024.102819] [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: 05/22/2024] [Revised: 07/18/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Defined as scarce expression of hormone receptors and human epidermal growth factor receptor 2, triple-negative breast cancer (TNBC) is labeled as the most heterogeneous subtype of breast cancer with poorest prognosis. Despite rapid advancements in precise subtyping and tailored therapeutics, the ensuing cancer therapy-related cardiovascular toxicity (CTR-CVT) could exert detrimental impacts to TNBC survivors. Nowadays, this interdisciplinary issue is incrementally concerned by cardiologists, oncologists and other pertinent experts, propelling cardio-oncology as a booming field focusing on the whole-course management of cancer patients with potential cardiovascular threats. Here in this review, we initially profile the evolving molecular subtyping and therapeutic landscape of TNBC. Further, we introduce various monitoring approaches of CTR-CVT. In the main body, we elaborate on typical cardiotoxicities ensuing anti-TNBC treatments in detail, ranging from chemotherapy (especially anthracyclines), surgery, anesthetics, radiotherapy to immunotherapy, with future perspectives on promising directions in the era of artificial intelligence and traditional Chinese medicine.
Collapse
Affiliation(s)
- Chongyu Wang
- Department of Medicine, Xinglin College, Nantong University, Nantong 226007, Jiangsu, China
| | - Pinchao Fan
- The First Clinical Medical College, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Sir Run Run Hospital, Nanjing Medical University, Nanjing 211112, Jiangsu, China
| | - Qingqing Wang
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, Jiangsu, China.
| |
Collapse
|
2
|
Zhang H, Ding X, Qiu Y, Xie M, Wang H, Li T, Bao H, Huang S, Xiong Y, Tang X. Preventive effect of imperatorin against doxorubicin-induced cardiotoxicity through suppression of NLRP3 inflammasome activation. J Nat Med 2024:10.1007/s11418-024-01850-x. [PMID: 39436583 DOI: 10.1007/s11418-024-01850-x] [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: 06/24/2024] [Accepted: 09/27/2024] [Indexed: 10/23/2024]
Abstract
Cardiotoxicity is one of the major obstacles to anthracycline chemotherapy. Anthracycline cardiotoxicity is closely associated with inflammation. Imperatorin (IMP), a furocoumarin ingredient extracted from Angelica dahurica, might have potential activity in preventing anthracycline cardiotoxicity due to its anti-cancer, anti-inflammatory, anti-oxidant, cardioprotective properties. This study aims to reveal the effect of IMP on doxorubicin (DOX)-induced cardiotoxicity and its underlying mechanism. We established a rat model of DOX-induced cardiotoxicity by intraperitoneal injection with DOX (1.25 mg/kg twice weekly for 6 weeks), and found that both IMP (25 mg/kg and 12.5 mg/kg) and dexrazoxane 12.5 mg/kg relieved DOX-induced reductions in heart weight, change in cardiac histopathology, and elevated serum levels of LDH, AST and CK-MB. Moreover, DOX upregulated mRNA levels of NLRP3, CASP1, GSDMD, ASC, IL-1β and IL-18, elevated protein expressions of NLRP3, ASC, GSDMD-FL, GSDMD-N, pro‑caspase‑1, caspase‑1 p20, pro‑IL‑1β and IL‑1β in heart tissues, as well as increased serum levels of pro-inflammatory cytokines including IL-1β and IL-18, however both of IMP and dexrazoxane suppressed these alterations. In addition, we carried out neonatal rat cardiomyocytes experiments to confirm the results of the in vivo study. Consistently, pretreatment with IMP 25 µg/mL relieved DOX (1 μg/mL)-induced cardiomyocytes injury, including decreased cell viability and reduced supernatant LDH. IMP inhibited DOX-induced activation of NLRP3 inflammasome in cardiomyocytes. In conclusion, IMP had a protective effect against DOX-induced cardiotoxicity via repressing the activation of NLRP3 inflammasome. These findings suggest that IMP may be a promising alternative or adjunctive drug for the prevention of anthracycline cardiotoxicity.
Collapse
Affiliation(s)
- Hao Zhang
- School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Xiaoyun Ding
- School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Yumei Qiu
- School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Mengdie Xie
- School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Hu Wang
- School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Tingting Li
- School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Huiyun Bao
- School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Si Huang
- School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Yinhua Xiong
- School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Nanchang, 330013, China
| | - Xilan Tang
- School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, 330013, China.
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Nanchang, 330013, China.
| |
Collapse
|
3
|
Lin X, Ma X, Zhao S, Yao J, Han L, Jing Y, Xue X. Cardiovascular toxicity in antitumor therapy: biological and therapeutic insights. Trends Cancer 2024; 10:920-934. [PMID: 39097431 DOI: 10.1016/j.trecan.2024.07.004] [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: 04/16/2024] [Revised: 06/28/2024] [Accepted: 07/12/2024] [Indexed: 08/05/2024]
Abstract
The evolution of antitumor therapies has significantly improved cancer prognosis but has concurrently resulted in cardiovascular toxicities. Understanding the biological mechanisms behind these toxicities is crucial for effective management. Immunotherapy-related cardiovascular toxicities are primarily mediated by immune cells and secreted cytokines. Chemotherapy may cause cardiovascular damage through autophagy disruption and mitochondrial dysfunction. Targeted therapies can induce toxicity through endothelin-1 (ET-1) production and cardiac signaling disruption. Radiotherapy may lead to cardiomyopathy and myocardial fibrosis by affecting endothelial cells, triggering inflammatory responses and accelerating atherosclerosis. This review provides insights into these mechanisms and strategies, aiming to enhance the clinical prevention and treatment of cardiovascular toxicities.
Collapse
Affiliation(s)
- Xuwen Lin
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xidong Ma
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Sheng Zhao
- Department of Cardiology, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Yao
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Leng Han
- Brown Center for Immunotherapy, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Department of Biostatistics and Health Data Science, School of Medicine, Indiana University, Indianapolis, IN 46202, USA.
| | - Ying Jing
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Guangzhou, China.
| | - Xinying Xue
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Department of Respiratory and Critical Care, Shandong Second Medical University, Weifang, China.
| |
Collapse
|
4
|
Li Y, Yan J, Yang P. The mechanism and therapeutic strategies in doxorubicin-induced cardiotoxicity: Role of programmed cell death. Cell Stress Chaperones 2024; 29:666-680. [PMID: 39343295 PMCID: PMC11490929 DOI: 10.1016/j.cstres.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/26/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024] Open
Abstract
Doxorubicin (DOX) is the most commonly used anthracycline anticancer agent, while its clinical utility is limited by harmful side effects like cardiotoxicity. Numerous studies have elucidated that programmed cell death plays a significant role in DOX-induced cardiotoxicity (DIC). This review summarizes several kinds of programmed cell death, including apoptosis, pyroptosis, necroptosis, autophagy, and ferroptosis. Furthermore, oxidative stress, inflammation, and mitochondrial dysfunction are also important factors in the molecular mechanisms of DIC. Besides, a comprehensive understanding of specific signal pathways of DIC can be helpful to its treatment. Therefore, the related signal pathways are elucidated in this review, including sirtuin deacetylase (silent information regulator 2 [Sir2]) 1 (SIRT1)/nuclear factor erythroid 2-related factor 2, SIRT1/Klotho, SIRT1/Recombinant Sestrin 2, adenosine monophosphate-activated protein kinase, AKT, and peroxisome proliferator-activated receptor. Heat shock proteins function as chaperones, which play an important role in various stressful situations, especially in the heart. Thus, some of heat shock proteins involved in DIC are also included. Hence, the last part of this review focuses on the therapeutic research based on the mechanisms above.
Collapse
Affiliation(s)
- Yanzhao Li
- Department of Second Clinical Medical College, Southern Medical University, Guangzhou, China.
| | - Jing Yan
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Pingzhen Yang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Zhang M, Wu X, Wen Y, Li Z, Chen F, Zou Y, Dong X, Liu X, Wang J. Epirubicin induces cardiotoxicity through disrupting ATP6V0A2-dependent lysosomal acidification and triggering ferroptosis in cardiomyocytes. Cell Death Discov 2024; 10:337. [PMID: 39048556 PMCID: PMC11269639 DOI: 10.1038/s41420-024-02095-z] [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: 01/28/2024] [Revised: 07/03/2024] [Accepted: 07/09/2024] [Indexed: 07/27/2024] Open
Abstract
Epirubicin (EPI) is effective in the treatment of malignant cancers, but its application is limited by life-threatening cardiotoxicity. Iron homeostasis disturbance has been implicated in anthracycline induced cardiotoxicity (AIC), and ferroptosis is involved in AIC which dependent upon intracellular iron. However, the role and exact mechanisms of ferroptosis in the pathogenesis of epirubicin-induced cardiotoxicity (EIC) remain elusive. In this study, we aimed to investigate mechanisms underlying ferroptosis-driven EIC. Epirubicin triggered ferroptosis both in vivo and in cultured cardiomyocytes, and pretreatment with ferroptosis inhibitor, Ferrostatin-1(Fer-1) alleviates EIC. Microarray analysis was performed to screen for potential molecules involved in EIC in neonatal primary mouse ventricular cardiomyocytes (NMVMs). We found that the transcript level of ATP6V0A2, a subunit of vacuolar ATPase (V-ATPase), was significantly downregulated when NMVMs were subjected to EPI, which was verified in vivo and in vitro as measured by real time quantitative reverse transcription PCR (qRT-PCR) and immunoblotting. Intriguingly, overexpression of ATP6V0A2 effectively decreased excessive oxidative stress and lipid-peroxidation accumulation, thereby inhibiting ferroptosis and protecting cardiomyocytes against EIC, as evidenced by functional, enzymatic, and morphological changes. Mechanistically, forced expression of ATP6V0A2 restored lysosomal acidification in EPI-treated cardiomyocytes and protected cardiomyocytes and mice hearts from ferroptosis-driven EIC. In this study, our data elucidate that ferroptosis is involved in EIC, which is ignited by ATP6V0A2-dependent lysosomal acidification dysfunction. Our study provides a new potential therapeutic target for ameliorating EIC.
Collapse
Affiliation(s)
- Mingming Zhang
- Department of Cardiology, the first affiliated hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, China
| | - Xin Wu
- Department of Obstetrics, the first affiliated hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, China
| | - Yuting Wen
- Department of Pathogen Biology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, China
| | - Zhiquan Li
- Department of Cardiology, the first affiliated hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, China
| | - Fuzhong Chen
- Department of Cardiology, the first affiliated hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, China
| | - Yu Zou
- Department of Cardiology, the first affiliated hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, China
| | - Xiaoyu Dong
- Department of Cardiology, the first affiliated hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, China
| | - Xinjian Liu
- Department of Pathogen Biology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, China.
- Key Laboratory of Antibody Technique of National Health Commission of China, Nanjing Medical University, 101 Longmian Avenue, Nanjing, China.
| | - Junhong Wang
- Department of Cardiology, the first affiliated hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, China.
| |
Collapse
|
6
|
Wang B, Wang J, Liu C, Li C, Meng T, Chen J, Liu Q, He W, Liu Z, Zhou Y. Ferroptosis: Latest evidence and perspectives on plant-derived natural active compounds mitigating doxorubicin-induced cardiotoxicity. J Appl Toxicol 2024. [PMID: 39030835 DOI: 10.1002/jat.4670] [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: 06/08/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/22/2024]
Abstract
Doxorubicin (DOX) is a chemotherapy drug widely used in clinical settings, acting as a first-line treatment for various malignant tumors. However, its use is greatly limited by the cardiotoxicity it induces, including doxorubicin-induced cardiomyopathy (DIC). The mechanisms behind DIC are not fully understood, but its potential biological mechanisms are thought to include oxidative stress, inflammation, energy metabolism disorders, mitochondrial damage, autophagy, apoptosis, and ferroptosis. Recent studies have shown that cardiac injury induced by DOX is closely related to ferroptosis. Due to their high efficacy, availability, and low side effects, natural medicine treatments hold strong clinical potential. Currently, natural medicines have been shown to mitigate DOX-induced ferroptosis and ease DIC through various functions such as antioxidation, iron ion homeostasis correction, lipid metabolism regulation, and mitochondrial function improvement. Therefore, this review summarizes the mechanisms of ferroptosis in DIC and the regulation by natural plant products, with the expectation of providing a reference for future research and development of inhibitors targeting ferroptosis in DIC. This review explores the mechanisms of ferroptosis in doxorubicin-induced cardiomyopathy (DIC) and summarizes how natural plant products can alleviate DIC by inhibiting ferroptosis through reducing oxidative stress, correcting iron ion homeostasis, regulating lipid metabolism, and improving mitochondrial function.
Collapse
Affiliation(s)
- Boyu Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jiameng Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Changxing Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chengjia Li
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Tianwei Meng
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jia Chen
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qingnan Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wang He
- First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhiping Liu
- First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yabin Zhou
- First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
7
|
Vitale R, Marzocco S, Popolo A. Role of Oxidative Stress and Inflammation in Doxorubicin-Induced Cardiotoxicity: A Brief Account. Int J Mol Sci 2024; 25:7477. [PMID: 39000584 PMCID: PMC11242665 DOI: 10.3390/ijms25137477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/01/2024] [Accepted: 07/06/2024] [Indexed: 07/16/2024] Open
Abstract
Cardiotoxicity is the main side effect of several chemotherapeutic drugs. Doxorubicin (Doxo) is one of the most used anthracyclines in the treatment of many tumors, but the development of acute and chronic cardiotoxicity limits its clinical usefulness. Different studies focused only on the effects of long-term Doxo administration, but recent data show that cardiomyocyte damage is an early event induced by Doxo after a single administration that can be followed by progressive functional decline, leading to overt heart failure. The knowledge of molecular mechanisms involved in the early stage of Doxo-induced cardiotoxicity is of paramount importance to treating and/or preventing it. This review aims to illustrate several mechanisms thought to underlie Doxo-induced cardiotoxicity, such as oxidative and nitrosative stress, inflammation, and mitochondrial dysfunction. Moreover, here we report data from both in vitro and in vivo studies indicating new therapeutic strategies to prevent Doxo-induced cardiotoxicity.
Collapse
Affiliation(s)
| | | | - Ada Popolo
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (R.V.); (S.M.)
| |
Collapse
|
8
|
Fan X, Han J, Zhong L, Zheng W, Shao R, Zhang Y, Shi S, Lin S, Huang Z, Huang W, Cai X, Ye B. Macrophage-Derived GSDMD Plays an Essential Role in Atherosclerosis and Cross Talk Between Macrophages via the Mitochondria-STING-IRF3/NF-κB Axis. Arterioscler Thromb Vasc Biol 2024; 44:1365-1378. [PMID: 38695170 DOI: 10.1161/atvbaha.123.320612] [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: 12/19/2023] [Accepted: 04/16/2024] [Indexed: 05/24/2024]
Abstract
BACKGROUND Macrophages play a crucial role in atherosclerotic plaque formation, and the death of macrophages is a vital factor in determining the fate of atherosclerosis. GSDMD (gasdermin D)-mediated pyroptosis is a programmed cell death, characterized by membrane pore formation and inflammatory factor release. METHODS ApoE-/- and Gsdmd-/- ApoE-/- mice, bone marrow transplantation, and AAV (adeno-associated virus serotype 9)-F4/80-shGSDMD (shRNA-GSDMD) were used to examine the effect of macrophage-derived GSDMD on atherosclerosis. Single-cell RNA sequencing was used to investigate the changing profile of different cellular components and the cellular localization of GSDMD during atherosclerosis. RESULTS First, we found that GSDMD is activated in human and mouse atherosclerotic plaques and Gsdmd-/- attenuates the atherosclerotic lesion area in high-fat diet-fed ApoE-/- mice. We performed single-cell RNA sequencing of ApoE-/- and Gsdmd-/- ApoE-/- mouse aortas and showed that GSDMD is principally expressed in atherosclerotic macrophages. Using bone marrow transplantation and AAV-F4/80-shGSDMD, we identified the potential role of macrophage-derived GSDMD in aortic pyroptosis and atherosclerotic injuries in vivo. Mechanistically, GSDMD contributes to mitochondrial perforation and mitochondrial DNA leakage and subsequently activates the STING (stimulator of interferon gene)-IRF3 (interferon regulatory factor 3)/NF-κB (nuclear factor kappa B) axis. Meanwhile, GSDMD regulates the STING pathway activation and macrophage migration via cytokine secretion. Inhibition of GSDMD with GSDMD-specific inhibitor GI-Y1 (GSDMD inhibitor Y1) can effectively alleviate the progression of atherosclerosis. CONCLUSIONS Our study has provided a novel macrophage-derived GSDMD mechanism in the promotion of atherosclerosis and demonstrated that GSDMD can be a potential therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Xiaoxi Fan
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
| | - Jibo Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Zhejiang, China (J.H.)
| | - Lingfeng Zhong
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
| | - Wenyuan Zheng
- Key Laboratory of Precision Medicine for Atherosclerosis Disease of Zhejiang Province, Department of Cardiology, Affiliated First Hospital of Ningbo University, Zhejiang, China (W.Z., B.Y.)
| | - Ruiyin Shao
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
| | - Yucong Zhang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
| | - Si Shi
- First School of Medicine, Wenzhou Medical University, Zhejiang, China (S.S.)
| | - Shuang Lin
- Department of Cardiology, Ningbo Medical Center Li Huili Hospital, Zhejiang, China (S.L.)
| | - Zhouqing Huang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
| | - Weijian Huang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
| | - Xueli Cai
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
| | - Bozhi Ye
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
- Key Laboratory of Precision Medicine for Atherosclerosis Disease of Zhejiang Province, Department of Cardiology, Affiliated First Hospital of Ningbo University, Zhejiang, China (W.Z., B.Y.)
| |
Collapse
|
9
|
Zhao W, Wang C, Zhu Y, Wang Q, Xu X, Shao Z, Chen M, Feng Y, Meng X. Visualized Tracking and Multidimensional Assessing of Mitochondria-Associated Pyroptosis in Cancer Cells by a Small-Molecule Fluorescent Probe. Anal Chem 2024; 96:6381-6389. [PMID: 38593059 DOI: 10.1021/acs.analchem.4c00318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Pyroptosis is closely related to the development and treatment of various cancers; thus, comprehensive studies of the correlations between pyroptosis and its inductive or inhibitive factors can provide new ideas for the intervention and diagnosis of tumors. The dysfunction of mitochondria may induce pyroptosis in cancer cells, which can be reflected by the fluctuations of the microenvironmental parameters in mitochondria as well as the changes of mitochondrial DNA level and morphology, etc. To precisely track and assess the mitochondria-associated pyroptosis process, simultaneous visualization of changes in multiphysiological parameters in mitochondria is highly desirable. In this work, we reported a nonreaction-based, multifunctional small-molecule fluorescent probe Mito-DK with the capability of crosstalk-free response to polarity and mtDNA as well as mitochondrial morphology. Accurate assessment of mitochondria-associated pyroptosis induced by palmitic acid/H2O2 was achieved through monitoring changes in mitochondrial multiple parameters with the help of Mito-DK. In particular, the pyroptosis-inducing ability of an antibiotic doxorubicin and the pyroptosis-inhibiting capacity of an anticancer agent puerarin were evaluated by Mito-DK. These results provide new perspectives for visualizing mitochondria-associated pyroptosis and offer new approaches for screening pyroptosis-related anticancer agents.
Collapse
Affiliation(s)
- Wenhao Zhao
- School of Chemistry and Chemical Engineering & Institutes of Physical Science and Information Technology, Anhui Province Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials & Key Laboratory of Structure and Functional Regulation of Hybrid Materials of Ministry of Education, Anhui University, Hefei 230601, P. R. China
| | - Chengyuan Wang
- School of Chemistry and Chemical Engineering & Institutes of Physical Science and Information Technology, Anhui Province Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials & Key Laboratory of Structure and Functional Regulation of Hybrid Materials of Ministry of Education, Anhui University, Hefei 230601, P. R. China
| | - Yanzhe Zhu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei 230022, P. R. China
| | - Qi Wang
- School of Chemistry and Chemical Engineering & Institutes of Physical Science and Information Technology, Anhui Province Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials & Key Laboratory of Structure and Functional Regulation of Hybrid Materials of Ministry of Education, Anhui University, Hefei 230601, P. R. China
| | - Xianyun Xu
- School of Chemistry and Chemical Engineering & Institutes of Physical Science and Information Technology, Anhui Province Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials & Key Laboratory of Structure and Functional Regulation of Hybrid Materials of Ministry of Education, Anhui University, Hefei 230601, P. R. China
| | - Zonglong Shao
- School of Chemistry and Chemical Engineering & Institutes of Physical Science and Information Technology, Anhui Province Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials & Key Laboratory of Structure and Functional Regulation of Hybrid Materials of Ministry of Education, Anhui University, Hefei 230601, P. R. China
| | - Man Chen
- School of Chemistry and Chemical Engineering & Institutes of Physical Science and Information Technology, Anhui Province Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials & Key Laboratory of Structure and Functional Regulation of Hybrid Materials of Ministry of Education, Anhui University, Hefei 230601, P. R. China
| | - Yan Feng
- School of Chemistry and Chemical Engineering & Institutes of Physical Science and Information Technology, Anhui Province Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials & Key Laboratory of Structure and Functional Regulation of Hybrid Materials of Ministry of Education, Anhui University, Hefei 230601, P. R. China
| | - Xiangming Meng
- School of Chemistry and Chemical Engineering & Institutes of Physical Science and Information Technology, Anhui Province Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials & Key Laboratory of Structure and Functional Regulation of Hybrid Materials of Ministry of Education, Anhui University, Hefei 230601, P. R. China
| |
Collapse
|
10
|
Zhao J, Yang T, Yi J, Hu H, Lai Q, Nie L, Liu M, Chu C, Yang J. AP39 through AMPK-ULK1-FUNDC1 pathway regulates mitophagy, inhibits pyroptosis, and improves doxorubicin-induced myocardial fibrosis. iScience 2024; 27:109321. [PMID: 38558936 PMCID: PMC10981016 DOI: 10.1016/j.isci.2024.109321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 01/10/2024] [Accepted: 02/20/2024] [Indexed: 04/04/2024] Open
Abstract
Doxorubicin induces myocardial injury and fibrosis. Still, no effective interventions are available. AP39 is an H2S donor that explicitly targets mitochondria. This study investigated whether AP39 could improve doxorubicin-induced myocardial fibrosis. Doxorubicin induced significant myocardial fibrosis while suppressing mitophagy-related proteins and elevating pyroptosis-related proteins. Conversely, AP39 reverses these effects, enhancing mitophagy and inhibiting pyroptosis. In vitro experiments revealed that AP39 inhibited H9c2 cardiomyocyte pyroptosis, improved doxorubicin-induced impairment of mitophagy, reduced ROS levels, ameliorated the mitochondrial membrane potential, and upregulated AMPK-ULK1-FUNDC1 expression. In contrast, AMPK inhibitor (dorsomorphin) and ULK1 inhibitor (SBI-0206965) reversed AP39 antagonism of doxorubicin-induced FUNDC1-mediated impairment of mitophagy and secondary cardiomyocyte pyroptosis. These results suggest that mitochondria-targeted H2S can antagonize doxorubicin-induced pyroptosis and impaired mitophagy in cardiomyocytes via AMPK-ULK1-FUNDC1 and ameliorated myocardial fibrosis and remodeling.
Collapse
Affiliation(s)
- Junxiong Zhao
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Ting Yang
- School of Pharmaceutical Science of University of South China, Hengyang 421000, China
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Jiali Yi
- Department of Cardiology, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Hongmin Hu
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Qi Lai
- School of Pharmaceutical Science of University of South China, Hengyang 421000, China
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Liangui Nie
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Maojun Liu
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Chun Chu
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Jun Yang
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| |
Collapse
|
11
|
Zhu C, Xu S, Jiang R, Yu Y, Bian J, Zou Z. The gasdermin family: emerging therapeutic targets in diseases. Signal Transduct Target Ther 2024; 9:87. [PMID: 38584157 PMCID: PMC10999458 DOI: 10.1038/s41392-024-01801-8] [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: 01/15/2024] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 04/09/2024] Open
Abstract
The gasdermin (GSDM) family has garnered significant attention for its pivotal role in immunity and disease as a key player in pyroptosis. This recently characterized class of pore-forming effector proteins is pivotal in orchestrating processes such as membrane permeabilization, pyroptosis, and the follow-up inflammatory response, which are crucial self-defense mechanisms against irritants and infections. GSDMs have been implicated in a range of diseases including, but not limited to, sepsis, viral infections, and cancer, either through involvement in pyroptosis or independently of this process. The regulation of GSDM-mediated pyroptosis is gaining recognition as a promising therapeutic strategy for the treatment of various diseases. Current strategies for inhibiting GSDMD primarily involve binding to GSDMD, blocking GSDMD cleavage or inhibiting GSDMD-N-terminal (NT) oligomerization, albeit with some off-target effects. In this review, we delve into the cutting-edge understanding of the interplay between GSDMs and pyroptosis, elucidate the activation mechanisms of GSDMs, explore their associations with a range of diseases, and discuss recent advancements and potential strategies for developing GSDMD inhibitors.
Collapse
Affiliation(s)
- Chenglong Zhu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
| | - Sheng Xu
- National Key Laboratory of Immunity & Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Ruoyu Jiang
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Naval Medical University, Shanghai, 200433, China
| | - Yizhi Yu
- National Key Laboratory of Immunity & Inflammation, Naval Medical University, Shanghai, 200433, China.
| | - Jinjun Bian
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| | - Zui Zou
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
12
|
Bhatti R, Sato PY. Exploring the role of pyroptosis in the pathogenicity of heart disease. Front Physiol 2024; 15:1357285. [PMID: 38645692 PMCID: PMC11026861 DOI: 10.3389/fphys.2024.1357285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 03/25/2024] [Indexed: 04/23/2024] Open
Abstract
Cell death is an essential cellular mechanism that ensures quality control and whole-body homeostasis. Various modes of cell death have been studied and detailed. Unbalanced cell death can lead to uncontrolled cell proliferation (i.e., tumors) or excessive loss of cells (i.e., ischemia injury tissue loss). Thus, it is imperative for modes of cell death to be balanced and controlled. Here, we will focus on a recent mode of cell death called pyroptosis. While extensive studies have shown the role of this route of cell death in macrophages and monocytes, evidence for pyroptosis have expanded to encompass other pathologies, including cancer and cardiac diseases. Herein, we provide a brief review on pyroptosis and discuss current gaps in knowledge and scientific advances in cardiac pyroptosis in recent years. Lastly, we provide conclusions and prospective on the relevance to various cardiac diseases.
Collapse
Affiliation(s)
| | - Priscila Y. Sato
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
13
|
Fang Z, Han J, Lin L, Ye B, Qu X, Zhang Y, Zhao Y, Xu D, Lin W, Shen S, Min J, Wu G, Huang Z, Liang G. Deubiquitinase OTUD6a drives cardiac inflammation and hypertrophy by deubiquitination of STING. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167061. [PMID: 38342418 DOI: 10.1016/j.bbadis.2024.167061] [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: 09/20/2023] [Revised: 01/31/2024] [Accepted: 02/04/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND Cardiac hypertrophy is a crucial pathological characteristic of hypertensive heart disease and subsequent heart failure. Deubiquitinating enzymes (DUBs) have been found to be involved in the regulation of myocardial hypertrophy. OTU Domain-Containing Protein 6a (OTUD6a) is a recently identified DUB. To date, the potential role of OTUD6a in myocardial hypertrophy has not yet been revealed. METHODS AND RESULTS We examined the up-regulated level of OTUD6a in mouse or human hypertrophic heart tissues. Then, transverse aortic constriction (TAC)- or angiotensin II (Ang II)- induced ventricular hypertrophy and dysfunction were significantly attenuated in OTUD6a gene knockout mice (OTUD6a-/-). In mechanism, we identified that the Stimulator of Interferon Genes (STING) is a direct substrate protein of OTUD6a via immunoprecipitation assay and mass spectrometry. OTUD6a maintains STING stability via clearing the K48-linked ubiquitin in cardiomyocytes. Subsequently, OTUD6a regulates the STING-downstream NF-κB signaling activation and inflammatory gene expression both in vivo and in vitro. Inhibition of STING blocked OTUD6a overexpression-induced inflammatory and hypertrophic responses in cardiomyocytes. CONCLUSION This finding extends our understanding of the detrimental role of OTUD6a in myocardial hypertrophy and identifies STING as a deubiquinating substrate of OTUD6a, indicating that targeting OTUD6a could be a potential strategy for the treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Zimin Fang
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jibo Han
- Department of Cardiology, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Liming Lin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bozhi Ye
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xuefeng Qu
- School of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yu Zhang
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ying Zhao
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Diyun Xu
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wante Lin
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Sirui Shen
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Julian Min
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Gaojun Wu
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Zhouqing Huang
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Guang Liang
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
14
|
Ye B, Fan X, Fang Z, Mao C, Lin L, Wu J, Zheng W, Cai X, Huang W, Lv Y, Han B, Han J. Macrophage-derived GSDMD promotes abdominal aortic aneurysm and aortic smooth muscle cells pyroptosis. Int Immunopharmacol 2024; 128:111554. [PMID: 38262162 DOI: 10.1016/j.intimp.2024.111554] [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: 08/14/2023] [Revised: 12/30/2023] [Accepted: 01/14/2024] [Indexed: 01/25/2024]
Abstract
Macrophage is a vital factor in determining the fate of abdominal aortic aneurysm (AAA). The crosstalk between macrophage and other cells plays a crucial role in the development of aneurysm. Gasdermin D (GSDMD) is a vital executive protein of pyroptosis, which is a novel programmed cell death associated with inflammation. In this study, we identified aortic macrophage as the main expressing cell of GSDMD in AAA. Using Gsdmd-/-ApoE-/- mouse and AAV-F4/80-shGSDMD, we demonstrated the potential role of macrophage-derived GSDMD in AAA and aortic pyroptosis induced by Ang II in vivo. In vitro experiments showed that GSDMD promotes the pyroptosis of mouse primary peritoneal macrophages (MPMs), murine aortic vascular smooth muscle cells (MOVAS) and primary smooth muscle cells. Mechanistically, a mouse cytokine antibody array showed that Gsdmd-/- inhibited LPS + nigericin (LN)- induced secretion of multiple cytokines from MPMs. Furthermore, GSDMD is involved in the crosstalk between MPMs and MOVAS via cytokine secretion. This study provides a novel fundamental insight into macrophage-derived GSDMD in AAA and showed that GSDMD could be a promising therapeutic target for AAA.
Collapse
Affiliation(s)
- Bozhi Ye
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoxi Fan
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zimin Fang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chenxi Mao
- Department of Cardiac Surgery, The 1(st) Affiliated Hospital of Wenzhou Medical University, China
| | - Liming Lin
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jun Wu
- Department of Vascular Surgery, The 2(nd) Affiliated Hospital of Wenzhou Medical University, China
| | - Wenyuan Zheng
- Department of Cardiology, The 1(st) Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Xueli Cai
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weijian Huang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yahui Lv
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Bingjiang Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jibo Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| |
Collapse
|
15
|
Ali SA, Singla DK. Mesenchymal Stem Cell-Derived Exosomes Ameliorate Doxorubicin-Induced Cardiotoxicity. Pharmaceuticals (Basel) 2024; 17:93. [PMID: 38256928 PMCID: PMC10820693 DOI: 10.3390/ph17010093] [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: 12/12/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Doxorubicin (DOX) is an incessantly used chemotherapeutic drug that can cause detrimental dose-dependent effects such as cardiotoxicity and congestive heart failure. Hence, there is a need to discover innovative therapeutic approaches to counteract DOX-induced cardiotoxicity (DIC). MSC-Exos have shown to reduce apoptosis and cardiac fibrosis and promote cardiomyocyte proliferation in myocardial infracted mice. However, the effect of MSC-Exos on ameliorating DOX-induced pyroptosis has not been investigated. In this current study, H9c2 were first exposed to DOX to stimulate pyroptosis, followed by subsequent treatment with MSC-Exos, with further analysis performed through immunocytochemistry, western blotting, and RT-PCR. Our data depicted that post-treatment with MSC-Exos significantly (p < 0.05) reduced the HMGB1/TLR4 axis, inflammasome formation (NLRP3), pyroptotic markers (caspase-1, IL-1β, and IL-18), and the pyroptotic executioner (GSDMD) in DOX-treated H9c2 cells. In conclusion, our data show that MSC-Exos attenuates inflammation-induced pyroptosis in our in vitro DIC model. Our findings indicate that MSC-Exos may serve as a promising therapeutic intervention for mitigating DIC, as they maintain the therapeutic capabilities of MSCs while circumventing the drawbacks associated with traditional stem cell therapy.
Collapse
Affiliation(s)
| | - Dinender K. Singla
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA;
| |
Collapse
|
16
|
Zhong Z, Gao Y, Zhou J, Wang F, Zhang P, Hu S, Wu H, Lou H, Chi J, Lin H, Guo H. Inhibiting mir-34a-5p regulates doxorubicin-induced autophagy disorder and alleviates myocardial pyroptosis by targeting Sirt3-AMPK pathway. Biomed Pharmacother 2023; 168:115654. [PMID: 37806095 DOI: 10.1016/j.biopha.2023.115654] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/27/2023] [Accepted: 10/04/2023] [Indexed: 10/10/2023] Open
Abstract
Doxorubicin (DOX) is a commonly used chemotherapy drug widely applied in various cancers such as breast cancer, leukemia, and sarcomas. However, its usage is limited by cardiotoxicity. Additionally, the cardiac toxicity of DOX accumulates with dose and duration, making it imperative to identify therapeutic targets for DOX-induced cardiomyopathy (DIC). It has been reported that miRNAs are involved in the progression of DIC. Mir-34a-5p has been identified as an early diagnostic marker for DIC. While studies have shown the involvement of mir-34a-5p in DIC apoptosis, it has not been validated in animal models, nor has the potential improvement of DIC by inhibiting mir-34a-5p been confirmed. Autophagy and pyroptosis are key factors in the development of DIC and can serve as therapeutic targets for its treatment. In this study, we found that mir-34a-5p was upregulated in the heart after DOX treatment and that the inhibition of mir-34-5p reduced autophagy and pyroptosis in DIC. We also found that the inhibition of mir-34a-5p inhibited pyroptosis by regulating autophagy and reducing mitochondrial reactive oxygen species. Moreover, we identified Sirtuin3 (Sirt3) as a target gene of mir-34a-5p using a double-luciferase reporter assay. overexpression Sirt3 reduced pyroptosis by alleviating autophagy. Our research findings suggest that inhibiting mir-34a-5p has a beneficial role in alleviating autophagy and pyroptosis in DIC. This provides therapeutic prospects for treating DIC.
Collapse
Affiliation(s)
- Zuoquan Zhong
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China; School of Medicine, Shaoxing University, Shaoxing, China; Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China
| | - Yefei Gao
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Jiedong Zhou
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Fang Wang
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Peipei Zhang
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Songqing Hu
- Zhejiang University School of Medicine, Hangzhou, China
| | - Haowei Wu
- Zhejiang University School of Medicine, Hangzhou, China
| | - Haifei Lou
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Jufang Chi
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China
| | - Hui Lin
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China; Healthy Science Center, The Affiliated Lihuili Hospital of Ningbo University, Ningbo University, Ningbo, Zhejiang 315211, China.
| | - Hangyuan Guo
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China; School of Medicine, Shaoxing University, Shaoxing, China; Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China.
| |
Collapse
|
17
|
Dai S, Chen Y, Fan X, Han J, Zhong L, Zhang Y, Liu Q, Lin J, Huang W, Su L, Huang Z, Ye B. Emodin attenuates cardiomyocyte pyroptosis in doxorubicin-induced cardiotoxicity by directly binding to GSDMD. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 121:155105. [PMID: 37801893 DOI: 10.1016/j.phymed.2023.155105] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 08/15/2023] [Accepted: 09/17/2023] [Indexed: 10/08/2023]
Abstract
BACKGROUND Doxorubicin (Dox), which is an anticancer drug, has significant cardiac toxicity and side effects. Pyroptosis occurs during Dox-induced cardiotoxicity (DIC), and drug inhibition of this process is one therapeutic approach for treating DIC. Previous studies have indicated that emodin can reduce pyroptosis. However, the role of emodin in DIC and its molecular targets remain unknown. HYPOTHESIS/PURPOSE We aimed to clarify the protective role of emodin in mitigating DIC, as well as the mechanisms underlying this effect. METHODS The model of DIC was established via the intraperitoneal administration of Dox at a dosage of 5 mg/kg per week for a span of 4 weeks. Emodin at two different doses (10 and 20 mg/kg) or a vehicle was intragastrically administered to the mice once per day throughout the Dox treatment period. Cardiac function, myocardial injury markers, pathological morphology of the heart, level of pyroptosis and mitochondrial function were assessed. Protein microarray, biolayer interferometry and pull-down assays were used to confirm the target of emodin. Moreover, GSDMD-overexpressing plasmids were transfected into GSDMD-/- mice and HL-1 cells to further verify whether emodin suppressed GSDMD activation. RESULTS Emodin therapy markedly enhanced cardiac function and reduced cardiomyocyte pyroptosis in mice induced by Dox. Mechanistically, emodin binds to GSDMD and inhibits the activation of GSDMD by targeting the Trp415 and Leu290 residues. Moreover, emodin was able to mitigate Dox-induced cardiac dysfunction and myocardial injury in GSDMD-/- mice overexpressing GSDMD, as shown by increased EF and FS, decreased serum levels of CK-MB, LDH and IL-1β and mitigated cell death and cell morphological disorder. Additionally, emodin treatment significantly reduced GSDMD-N expression and plasma membrane disruption in HL-1 cells overexpressing GSDMD induced by Dox. In addition, emodin reduced mitochondrial damage by alleviating Dox-induced GSDMD perforation in the mitochondrial membrane. CONCLUSION Emodin has the potential to attenuate DIC by directly binding to GSDMD to inhibit pyroptosis. Emodin may become a promising drug for prevention and treatment of DIC.
Collapse
Affiliation(s)
- Shanshan Dai
- The Key Laboratory of Emergency and Disaster Medicine of Wenzhou, Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Yunxuan Chen
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Xiaoxi Fan
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Jibo Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Lingfeng Zhong
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Yucong Zhang
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Qingran Liu
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiahui Lin
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weijian Huang
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Lan Su
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China.
| | - Zhouqing Huang
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China.
| | - Bozhi Ye
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China.
| |
Collapse
|
18
|
Gielecińska A, Kciuk M, Yahya EB, Ainane T, Mujwar S, Kontek R. Apoptosis, necroptosis, and pyroptosis as alternative cell death pathways induced by chemotherapeutic agents? Biochim Biophys Acta Rev Cancer 2023; 1878:189024. [PMID: 37980943 DOI: 10.1016/j.bbcan.2023.189024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/22/2023] [Accepted: 11/14/2023] [Indexed: 11/21/2023]
Abstract
For decades, common chemotherapeutic drugs have been established to trigger apoptosis, the preferred immunologically "silent" form of cell death. The primary objective of this review was to show that various FDA-approved chemotherapeutic drugs, including cisplatin, cyclosporine, doxorubicin, etoposide, 5-fluorouracil, gemcitabine, paclitaxel, or vinblastine can trigger necroptosis and pyroptosis. We aimed to provide the advantages and disadvantages of the induction of the given type of cell death by chemotherapeutical agents. Moreover, we give a short overview of the molecular mechanism of each type of cell death and indicate the existing crosstalks between cell death types. Finally, we provide a comparison of cell death types to facilitate the exploration of cell death types induced by other chemotherapeutical agents. Understanding the cell death pathway induced by a drug can lessen side effects and assist the discovery of new combinations with synergistic effects and low systemic toxicity.
Collapse
Affiliation(s)
- A Gielecińska
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Molecular Biotechnology and Genetics, Banacha St. 12/16, 90-237 Lodz, Poland; University of Lodz, Doctoral School of Exact and Natural Sciences, Banacha Street 12/16, 90-237 Lodz, Poland.
| | - M Kciuk
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Molecular Biotechnology and Genetics, Banacha St. 12/16, 90-237 Lodz, Poland
| | - E-B Yahya
- Bioprocess Technology Division, School of Industrial Technology, University Sains Malaysia, Penang 11800, Malaysia
| | - T Ainane
- Superior School of Technology of Khenifra, University of Sultan Moulay Slimane, P.O. Box 170, Khenifra 54000, Morocco
| | - S Mujwar
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India
| | - R Kontek
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Molecular Biotechnology and Genetics, Banacha St. 12/16, 90-237 Lodz, Poland
| |
Collapse
|
19
|
Zhong L, Han J, Fan X, Huang Z, Su L, Cai X, Lin S, Chen X, Huang W, Dai S, Ye B. Novel GSDMD inhibitor GI-Y1 protects heart against pyroptosis and ischemia/reperfusion injury by blocking pyroptotic pore formation. Basic Res Cardiol 2023; 118:40. [PMID: 37782407 DOI: 10.1007/s00395-023-01010-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 10/03/2023]
Abstract
Activation of gasdermin D (GSDMD) and its concomitant cardiomyocyte pyroptosis are critically involved in multiple cardiac pathological conditions. Pharmacological inhibition or gene knockout of GSDMD could protect cardiomyocyte from pyroptosis and dysfunction. Thus, seeking and developing highly potent GSDMD inhibitors probably provide an attractive strategy for treating diseases targeting GSDMD. Through structure-based virtual screening, pharmacological screening and subsequent pharmacological validations, we preliminarily identified GSDMD inhibitor Y1 (GI-Y1) as a selective GSDMD inhibitor with cardioprotective effects. Mechanistically, GI-Y1 binds to GSDMD and inhibits lipid- binding and pyroptotic pore formation of GSDMD-N by targeting the Arg7 residue. Importantly, we confirmed the cardioprotective effect of GI-Y1 on myocardial I/R injury and cardiac remodeling by targeting GSDMD. More extensively, GI-Y1 also inhibited the mitochondrial binding of GSDMD-N and its concomitant mitochondrial dysfunction. The findings of this study identified a new drug (GI-Y1) for the treatment of cardiac disorders by targeting GSDMD, and provide a new tool compound for pyroptosis research.
Collapse
Affiliation(s)
- Lingfeng Zhong
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jibo Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Xiaoxi Fan
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhouqing Huang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lan Su
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xueli Cai
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shuang Lin
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xudong Chen
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weijian Huang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Shanshan Dai
- The Key Laboratory of Emergency and Disaster Medicine of Wenzhou, Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Bozhi Ye
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
20
|
Li X. Doxorubicin-mediated cardiac dysfunction: Revisiting molecular interactions, pharmacological compounds and (nano)theranostic platforms. ENVIRONMENTAL RESEARCH 2023; 234:116504. [PMID: 37356521 DOI: 10.1016/j.envres.2023.116504] [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: 04/22/2023] [Revised: 06/17/2023] [Accepted: 06/23/2023] [Indexed: 06/27/2023]
Abstract
Although chemotherapy drugs are extensively utilized in cancer therapy, their administration for treatment of patients has faced problems that regardless of chemoresistance, increasing evidence has shown concentration-related toxicity of drugs. Doxorubicin (DOX) is a drug used in treatment of solid and hematological tumors, and its function is based on topoisomerase suppression to impair cancer progression. However, DOX can also affect the other organs of body and after chemotherapy, life quality of cancer patients decreases due to the side effects. Heart is one of the vital organs of body that is significantly affected by DOX during cancer chemotherapy, and this can lead to cardiac dysfunction and predispose to development of cardiovascular diseases and atherosclerosis, among others. The exposure to DOX can stimulate apoptosis and sometimes, pro-survival autophagy stimulation can ameliorate this condition. Moreover, DOX-mediated ferroptosis impairs proper function of heart and by increasing oxidative stress and inflammation, DOX causes cardiac dysfunction. The function of DOX in mediating cardiac toxicity is mediated by several pathways that some of them demonstrate protective function including Nrf2. Therefore, if expression level of such protective mechanisms increases, they can alleviate DOX-mediated cardiac toxicity. For this purpose, pharmacological compounds and therapeutic drugs in preventing DOX-mediated cardiotoxicity have been utilized and they can reduce side effects of DOX to prevent development of cardiovascular diseases in patients underwent chemotherapy. Furthermore, (nano)platforms are used comprehensively in treatment of cardiovascular diseases and using them for DOX delivery can reduce side effects by decreasing concentration of drug. Moreover, when DOX is loaded on nanoparticles, it is delivered into cells in a targeted way and its accumulation in healthy organs is prevented to diminish its adverse impacts. Hence, current paper provides a comprehensive discussion of DOX-mediated toxicity and subsequent alleviation by drugs and nanotherapeutics in treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaofeng Li
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai, 200072, China.
| |
Collapse
|
21
|
Chai R, Ye Z, Xue W, Shi S, Wei Y, Hu Y, Wu H. Tanshinone IIA inhibits cardiomyocyte pyroptosis through TLR4/NF-κB p65 pathway after acute myocardial infarction. Front Cell Dev Biol 2023; 11:1252942. [PMID: 37766966 PMCID: PMC10520722 DOI: 10.3389/fcell.2023.1252942] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Background: Tanshinone IIA, derived from Radix Salviae Miltiorrhizae (Salvia miltiorrhiza Bunge), constitutes a significant component of this traditional Chinese medicine. Numerous studies have reported positive outcomes regarding its influence on cardiac function. However, a comprehensive comprehension of the intricate mechanisms responsible for its cardioprotective effects is still lacking. Methods: A rat model of heart failure (HF) induced by acute myocardial infarction (AMI) was established via ligation of the left anterior descending coronary artery. Rats received oral administration of tanshinone IIA (1.5 mg/kg) and captopril (10 mg/kg) for 8 weeks. Cardiac function was assessed through various evaluations. Histological changes in myocardial tissue were observed using staining techniques, including Hematoxylin and Eosin (HE), Masson, and transmission electron microscopy. Tunel staining was used to detect cell apoptosis. Serum levels of NT-pro-BNP, IL-1β, and IL-18 were quantified using enzyme-linked immunosorbent assay (ELISA). Expression levels of TLR4, NF-κB p65, and pyroptosis-related proteins were determined via western blotting (WB). H9C2 cardiomyocytes underwent hypoxia-reoxygenation (H/R) to simulate ischemia-reperfusion (I/R) injury, and cell viability and apoptosis were assessed post treatment with different tanshinone IIA concentrations (0.05 μg/ml, 0.1 μg/ml). ELISA measured IL-1β, IL-18, and LDH expression in the cell supernatant, while WB analysis evaluated TLR4, NF-κB p65, and pyroptosis-related protein levels. NF-κB p65 protein nuclear translocation was observed using laser confocal microscopy. Results: Tanshinone IIA treatment exhibited enhanced cardiac function, mitigated histological cardiac tissue damage, lowered serum levels of NT-pro-BNP, IL-1β, and IL-18, and suppressed myocardial cell apoptosis. Moreover, tanshinone IIA downregulated the expression of TLR4, NF-κB p65, IL-1β, pro-IL-1β, NLRP3, Caspase-1, and GSDMD-N pyroptosis-related proteins in myocardial tissue. Additionally, it bolstered H/R H9C2 cardiomyocyte viability, curbed cardiomyocyte apoptosis, and reduced the levels of TLR4, NF-κB p65, IL-1β, pro-IL-1β, NLRP3, Caspase-1, and GSDMD-N pyroptosis-related proteins in H/R H9C2 cells. Furthermore, it hindered NF-κB p65 protein nuclear translocation. Conclusion: These findings indicate that tanshinone IIA enhances cardiac function and alleviates myocardial injury in HF rats following AMI. Moreover, tanshinone IIA demonstrates potential suppression of cardiomyocyte pyroptosis. These effects likely arise from the inhibition of the TLR4/NF-κB p65 signaling pathway, presenting a promising therapeutic target.
Collapse
Affiliation(s)
| | | | | | | | - Yi Wei
- Department of Cardiology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuanhui Hu
- Department of Cardiology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huaqin Wu
- Department of Cardiology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
22
|
Han J, Fang Z, Han B, Ye B, Lin W, Jiang Y, Han X, Wang X, Wu G, Wang Y, Liang G. Deubiquitinase JOSD2 improves calcium handling and attenuates cardiac hypertrophy and dysfunction by stabilizing SERCA2a in cardiomyocytes. NATURE CARDIOVASCULAR RESEARCH 2023; 2:764-777. [PMID: 39195964 DOI: 10.1038/s44161-023-00313-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 07/03/2023] [Indexed: 08/29/2024]
Abstract
Cardiac hypertrophy leads to myocardial dysfunction and represents a serious threat to global public health security. Deubiquitinating enzymes (DUBs) mainly maintain the stability of substrate proteins and are essential to cardiac pathophysiology. Here, we explored the role and regulating mechanism of a DUB, Josephin domain-containing protein 2 (JOSD2), in cardiac hypertrophy. We found that JOSD2 expression was significantly upregulated in hypertrophic myocardium. Josd2 gene knockout aggravated cardiac dysfunction and hypertrophy in mice, whereas cardiac overexpression of JOSD2 mediated by the AAV9 vector prevented angiotensin II-induced cardiac hypertrophy. A comprehensive proteome-wide quantitative analysis identified sarco/endoplasmic reticulum calcium ATPase 2a (SERCA2a) as a key substrate of JOSD2. Mechanistically, JOSD2 mediates SERCA2a deubiquitination, enhancing the stability of SERCA2a. By regulating SERCA2a, JOSD2 deficiency impairs calcium handling and promotes hypertrophy in primary cardiomyocytes. Our findings highlight the promise of JOSD2 as a beneficial therapeutic target for hypertrophic cardiomyopathy and provide an additional strategy for SERCA2a-targeted therapy.
Collapse
Affiliation(s)
- Jibo Han
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Zimin Fang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bingjiang Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Bozhi Ye
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wante Lin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yucheng Jiang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xue Han
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Xu Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Gaojun Wu
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.
- School of Pharmaceutical Sciences, Hangzhou Normal University, Hangzhou, China.
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China.
| |
Collapse
|
23
|
Sheng SY, Li JM, Hu XY, Wang Y. Regulated cell death pathways in cardiomyopathy. Acta Pharmacol Sin 2023; 44:1521-1535. [PMID: 36914852 PMCID: PMC10374591 DOI: 10.1038/s41401-023-01068-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/20/2023] [Indexed: 03/16/2023] Open
Abstract
Heart disease is a worldwide health menace. Both intractable primary and secondary cardiomyopathies contribute to malignant cardiac dysfunction and mortality. One of the key cellular processes associated with cardiomyopathy is cardiomyocyte death. Cardiomyocytes are terminally differentiated cells with very limited regenerative capacity. Various insults can lead to irreversible damage of cardiomyocytes, contributing to progression of cardiac dysfunction. Accumulating evidence indicates that majority of cardiomyocyte death is executed by regulating molecular pathways, including apoptosis, ferroptosis, autophagy, pyroptosis, and necroptosis. Importantly, these forms of regulated cell death (RCD) are cardinal features in the pathogenesis of various cardiomyopathies, including dilated cardiomyopathy, diabetic cardiomyopathy, sepsis-induced cardiomyopathy, and drug-induced cardiomyopathy. The relevance between abnormity of RCD with adverse outcome of cardiomyopathy has been unequivocally evident. Therefore, there is an urgent need to uncover the molecular and cellular mechanisms for RCD in order to better understand the pathogenesis of cardiomyopathies. In this review, we summarize the latest progress from studies on RCD pathways in cardiomyocytes in context of the pathogenesis of cardiomyopathies, with particular emphasis on apoptosis, necroptosis, ferroptosis, autophagy, and pyroptosis. We also elaborate the crosstalk among various forms of RCD in pathologically stressed myocardium and the prospects of therapeutic applications targeted to various cell death pathways.
Collapse
Affiliation(s)
- Shu-Yuan Sheng
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China
| | - Jia-Min Li
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China
| | - Xin-Yang Hu
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China
| | - Yibin Wang
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China.
- Signature Program in Cardiovascular and Metabolic Diseases, DukeNUS Medical School and National Heart Center of Singapore, Singapore, Singapore.
| |
Collapse
|
24
|
Fang G, Li X, Yang F, Huang T, Qiu C, Peng K, Wang Z, Yang Y, Lan C. Amentoflavone mitigates doxorubicin-induced cardiotoxicity by suppressing cardiomyocyte pyroptosis and inflammation through inhibition of the STING/NLRP3 signalling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 117:154922. [PMID: 37321078 DOI: 10.1016/j.phymed.2023.154922] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 05/16/2023] [Accepted: 06/05/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Doxorubicin (DOX) is a potent anticancer chemotherapeutic agent whose clinical application is substantially constrained by its cardiotoxicity. The pathophysiology of DOX-induced cardiotoxicity manifests as cardiomyocyte pyroptosis and inflammation. Amentoflavone (AMF) is a naturally occurring biflavone possessing anti-pyroptotic and anti-inflammatory properties. However, the mechanism through which AMF alleviates DOX-induced cardiotoxicity remains undetermined. PURPOSE This study aimed at investigating the role of AMF in alleviating DOX-induced cardiotoxicity. STUDY DESIGN AND METHODS To assess the in vivo effect of AMF, DOX was intraperitoneally administered into a mouse model to induce cardiotoxicity. To elucidate the underlying mechanisms, the activities of STING/NLRP3 were quantified using the NLRP3 agonist nigericin and the STING agonist amidobenzimidazole (ABZI). Primary cardiomyocytes isolated from neonatal Sprague-Dawley rats were treated with saline (vehicle) or DOX with or without AMF and/or ABZI. The echocardiogram, haemodynamics, cardiac injury markers, heart/body weight ratio, and pathological alterations were monitored; the STING/NLRP3 pathway-associated proteins were detected by western blot and cardiomyocyte pyroptosis was analysed by immunofluorescence staining of cleaved N-terminal GSDMD and scanning electron microscopy. Furthermore, we evaluated the potential of AMF in compromising the anticancer effects of DOX in human breast cancer cell lines. RESULTS AMF substantially alleviated cardiac dysfunction and reduced heart/body weight ratio and myocardial damage in mice models of DOX-induced cardiotoxicity. AMF effectively suppressed DOX-mediated upregulation of IL-1β, IL-18, TNF-α, and pyroptosis-related proteins, including NLRP3, cleaved caspase-1, and cleaved N-terminal GSDMD. The levels of apoptosis-related proteins, namely Bax, cleaved caspase-3, and BCL-2 were not affected. In addition, AMF inhibited STING phosphorylation in DOX-affected hearts. Intriguingly, the administration of nigericin or ABZI dampened the cardioprotective effects of AMF. The in vitro anti-pyroptotic effect of AMF was demonstrated in attenuating the DOX-induced reduction in cardiomyocyte cell viability, upregulation of cleaved N-terminal GSDMD, and pyroptotic morphology alteration at the microstructural level. AMF exhibited a synergistic effect with DOX to reduce the viability of human breast cancer cells. CONCLUSION AMF alleviates DOX-induced cardiotoxicity by suppressing cardiomyocyte pyroptosis and inflammation via inhibition of the STING/NLRP3 signalling pathway, thereby validating its efficacy as a cardioprotective agent.
Collapse
Affiliation(s)
- Guangyao Fang
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, P.R. China.; Department of Cardiology, General Hospital of Western Theater Command, Chengdu, Sichuan, P.R. China
| | - Xiuchuan Li
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, Sichuan, P.R. China
| | - Fengyuan Yang
- Department of Nephrology, General Hospital of Western Theater Command, Chengdu, Sichuan, P.R. China
| | - Ting Huang
- Department of Medical Oncology, People's Hospital of Luotian County, Huanggang, Hubei, P.R. China
| | - Chenming Qiu
- Department of Burn and Plastic Surgery, General Hospital of Western Theater Command, Chengdu, Sichuan, P.R. China
| | - Ke Peng
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, Sichuan, P.R. China
| | - Ziran Wang
- Department of Orthopedics, 903rd Hospital of PLA, Hangzhou, Zhejiang, P.R. China
| | - Yongjian Yang
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, P.R. China.; Department of Cardiology, General Hospital of Western Theater Command, Chengdu, Sichuan, P.R. China..
| | - Cong Lan
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, P.R. China.; Department of Cardiology, General Hospital of Western Theater Command, Chengdu, Sichuan, P.R. China..
| |
Collapse
|
25
|
Fang Z, Wu G, Sheng J, Ye B, Huang Z, Xu J, Zhang J, Han J, Han B, Xu J. Gasdermin D affects aortic vascular smooth muscle cell pyroptosis and Ang II-induced vascular remodeling. Heliyon 2023; 9:e16619. [PMID: 37303505 PMCID: PMC10248119 DOI: 10.1016/j.heliyon.2023.e16619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 05/18/2023] [Accepted: 05/23/2023] [Indexed: 06/13/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) are primarily responsible for vasoconstriction and the regulation of blood pressure1. Pyroptosis, a particular form of regulated cell death, is involved in multiple vascular injuries, including hypertensive vascular dysfunction. This pyroptotic cell death is mediated by the pore-forming protein of Gasdermin D (GSDMD). This study was designed to examine the direct effect of GSDMD on smooth muscle cell pyroptosis and vascular remodeling. Findings revealed that GSDMD was activated in Angiotensin (Ang) II- treated aortas. We then showed that genetic deletion of Gsdmd reduced vascular remodeling and aorta pyroptosis induced by Ang II in vivo. Aberrant expression of GSDMD by recombinant AAV9 virus carrying Gsdmd cDNA aggravated the level of pyroptosis in aortas of Ang II mice. Gain- and loss-of- function analysis further confirmed that GSDMD regulated the pyroptosis of murine aortic vascular smooth muscle cells (MOVAS) in an in vitro model of tumor necrosis factor (TNF)-α treatment, which was achieved by transfecting expressing plasmid or siRNA, respectively. Overall, this study provided evidence supporting the active involvement of GSDMD in smooth muscle cell pyroptosis and Ang II-induced mice vascular injury. This finding lends credence to GSDMD as a potential therapeutic target for hypertensive vascular remodeling via inhibiting pyroptosis.
Collapse
Affiliation(s)
- Zimin Fang
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gaojun Wu
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jian Sheng
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Bozhi Ye
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhouqing Huang
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianjiang Xu
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jianqin Zhang
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jibo Han
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Bingjiang Han
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jiajun Xu
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| |
Collapse
|
26
|
Han C, Sheng J, Pei H, Sheng Y, Wang J, Zhou X, Li W, Cao C, Yang Y. Environmental toxin chlorpyrifos induces liver injury by activating P53-mediated ferroptosis via GSDMD-mtROS. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 257:114938. [PMID: 37099958 DOI: 10.1016/j.ecoenv.2023.114938] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/16/2023] [Accepted: 04/18/2023] [Indexed: 05/08/2023]
Abstract
AIM We investigate the mechanism whereby chlorpyrifos (CHI), an environmental toxin, causes liver injury by inducing ferroptosis in hepatocytes. METHODS The toxic dose (LD50 = 50 μM) of CHI for inducing AML12 injury in normal mouse hepatocytes was determined, and the ferroptosis-related indices were measured, including the levels of SOD, MDA and GSH-Px, as well as the cellular content of iron ions. JC-1 and DCFH-DA assays were employed to detect the mtROS levels, the levels of mitochondrial proteins (GSDMD, NT-GSDMD), as well as the cellular levels of ferroptosis-related proteins (P53, GPX4, MDM2, SLC7A11). We knocked out the GSDMD and P53 in AML12 and observed the CHI-induced ferroptosis of ALM12 after applying YGC063, an ROS inhibitor. In animal experiments, we explored the effect of CHI on liver injury by using conditional GSDMD-knockout mice (C57BL/6 N-GSDMDem1(flox)Cya) and ferroptosis inhibitor Fer-1. Small molecule-protein docking and Pull-down assay were employed to verify the association between CHI and GSDMD. RESULTS We found that CHI could induce ferroptosis of AML12. CHI promoted the cleavage of GSDMD, leading to upregulation of mitochondrial NT-GSDMD expression, as well as ROS levels. P53 activation promoted the ferroptosis. Knock out of GSDMD and P53 could inhibit the CHI-induced ferroptosis, and YGC063 could also inhibit ferroptosis. In mice experiments, GSDMD knockout or Fer-1 intervention could significantly inhibit the CHI-induced liver injury. CHI promoted the cleavage of GSDMD by binding to its SER234 site. CONCLUSION CHI can bind to GSDMD to promote its cleavage, while NT-GSDMD can open mitochondrial membrane to promote the mtROS release. Cytoplasmic upregulation of ROS levels can facilitate the P53-mediated ferroptosis. GSDMD-mtROS is the primary mechanism whereby CHI induces ferroptosis in hepatocytes.
Collapse
Affiliation(s)
- Chenyang Han
- The Second Affiliated Hospital of Jiaxing University, 314001, China
| | - Jian Sheng
- The Second Affiliated Hospital of Jiaxing University, 314001, China
| | - Hongyan Pei
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Yongjia Sheng
- The Second Affiliated Hospital of Jiaxing University, China
| | - Jin Wang
- The Second Affiliated Hospital of Jiaxing University, China
| | - Xiaohong Zhou
- The Second Affiliated Hospital of Jiaxing University, China
| | - Wenyan Li
- The Second Affiliated Hospital of Jiaxing University, China
| | - Chenxi Cao
- The Second Affiliated Hospital of Jiaxing University, China.
| | - Yi Yang
- The Second Affiliated Hospital of Jiaxing University, China.
| |
Collapse
|
27
|
Wang W, Zhong X, Fang Z, Li J, Li H, Liu X, Yuan X, Huang W, Huang Z. Cardiac sirtuin1 deficiency exacerbates ferroptosis in doxorubicin-induced cardiac injury through the Nrf2/Keap1 pathway. Chem Biol Interact 2023; 377:110469. [PMID: 37030624 DOI: 10.1016/j.cbi.2023.110469] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 03/04/2023] [Accepted: 03/31/2023] [Indexed: 04/10/2023]
Abstract
Doxorubicin (DOX), a broad-spectrum chemotherapeutic agent for various cancers, has limited clinical application because of its serious cardiotoxicity, which is due to different mechanisms, including cardiac ferroptosis and oxidative stress. Some drugs, such as berberine or dioscin, show efficacy in impeding DOX-induced cardiotoxicity by activating Sirtuin 1 (Sirt1). However, there is no direct evidence to clarify the role of Sirt1 in DOX-induced cardiomyopathy and its underlying role in cardiac ferroptosis. In this study, C57BL/6 and cardiac-specific Sirt1-/- knockout mice were used as a DOX-induced cardiotoxicity model. We found that cardiac Sirt1 was downregulated, oxidative stress was increased and ferroptosis were obviously enhanced, as reflected by decreased Glutathione peroxidase 4 (GPX4) and increased Heme oxygenase 1 (Hmox-1), exposure to DOX treatment in mice and H9c2 cells compared with the control. And Sirt1 activation was resistant to cardiac injury induced by DOX, as observed the improvement of cardiac dysfunction, and the reduction of cardiac fibrosis. However, cardiac Sirt1 deficiency aggravated Dox-induced cardiac dysfunction and cardiac remodeling, further downregulated GPX4, upregulated Hmox-1 expression and increased ROS level. In addition, Sirt1-siRNA exacerbated DOX-induced cardiotoxicity in H9c2 cells, which is similar to the results obtained in vivo. Furthermore, DOX decrease Nrf2 translocation from the cytosol to the nucleus, and Sirt1 deficiency further restrain the process, as well as the downstream Keap1 pathways, in DOX-induced cardiotoxicity. This study provides direct evidence that Sirt1 plays a protective role in DOX-induced cardiotoxicity by mediating ferroptosis reduction via the Nrf2/Keap1 pathway.
Collapse
Affiliation(s)
- Weiqi Wang
- Department of Cardiology, The First Affiliated Hospital of WenZhou Medical College, 2 Fuxue Road, WenZhou, ZheJiang, 325000, PR China
| | - Xin Zhong
- Department of Cardiology, The First Affiliated Hospital of WenZhou Medical College, 2 Fuxue Road, WenZhou, ZheJiang, 325000, PR China
| | - Zimin Fang
- Department of Cardiology, The First Affiliated Hospital of WenZhou Medical College, 2 Fuxue Road, WenZhou, ZheJiang, 325000, PR China
| | - Jianmin Li
- Department of Pathology, The First Affiliated Hospital of WenZhou Medical College, 2 Fuxue Road, WenZhou, ZheJiang, 325000, PR China
| | - Hebo Li
- Department of Cardiology, The First Affiliated Hospital of WenZhou Medical College, 2 Fuxue Road, WenZhou, ZheJiang, 325000, PR China
| | - Xuesheng Liu
- Department of Cardiology, The First Affiliated Hospital of WenZhou Medical College, 2 Fuxue Road, WenZhou, ZheJiang, 325000, PR China
| | - Xindi Yuan
- Department of Cardiology, The First Affiliated Hospital of WenZhou Medical College, 2 Fuxue Road, WenZhou, ZheJiang, 325000, PR China
| | - Weijian Huang
- Department of Cardiology, The First Affiliated Hospital of WenZhou Medical College, 2 Fuxue Road, WenZhou, ZheJiang, 325000, PR China
| | - Zhouqing Huang
- Department of Cardiology, The First Affiliated Hospital of WenZhou Medical College, 2 Fuxue Road, WenZhou, ZheJiang, 325000, PR China.
| |
Collapse
|
28
|
Bo L, Wang Y, Li Y, Wurpel JND, Huang Z, Chen ZS. The Battlefield of Chemotherapy in Pediatric Cancers. Cancers (Basel) 2023; 15:cancers15071963. [PMID: 37046624 PMCID: PMC10093214 DOI: 10.3390/cancers15071963] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/12/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
The survival rate for pediatric cancers has remarkably improved in recent years. Conventional chemotherapy plays a crucial role in treating pediatric cancers, especially in low- and middle-income countries where access to advanced treatments may be limited. The Food and Drug Administration (FDA) approved chemotherapy drugs that can be used in children have expanded, but patients still face numerous side effects from the treatment. In addition, multidrug resistance (MDR) continues to pose a major challenge in improving the survival rates for a significant number of patients. This review focuses on the severe side effects of pediatric chemotherapy, including doxorubicin-induced cardiotoxicity (DIC) and vincristine-induced peripheral neuropathy (VIPN). We also delve into the mechanisms of MDR in chemotherapy to the improve survival and reduce the toxicity of treatment. Additionally, the review focuses on various drug transporters found in common types of pediatric tumors, which could offer different therapeutic options.
Collapse
Affiliation(s)
- Letao Bo
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA
| | - Youyou Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA
| | - Yidong Li
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA
| | - John N. D. Wurpel
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA
| | - Zoufang Huang
- Ganzhou Key Laboratory of Hematology, Department of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Correspondence: (Z.H.); (Z.-S.C.); Tel.: +86-138-797-27439 (Z.H.); +1-718-990-1432 (Z.-S.C.); Fax: +1-718-990-1877 (Z.-S.C.)
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA
- Institute for Biotechnology, St. John’s University, Queens, NY 11439, USA
- Correspondence: (Z.H.); (Z.-S.C.); Tel.: +86-138-797-27439 (Z.H.); +1-718-990-1432 (Z.-S.C.); Fax: +1-718-990-1877 (Z.-S.C.)
| |
Collapse
|
29
|
Zhang W, Wang X, Tang Y, Huang C. Melatonin alleviates doxorubicin-induced cardiotoxicity via inhibiting oxidative stress, pyroptosis and apoptosis by activating Sirt1/Nrf2 pathway. Biomed Pharmacother 2023; 162:114591. [PMID: 36965257 DOI: 10.1016/j.biopha.2023.114591] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/13/2023] [Accepted: 03/22/2023] [Indexed: 03/27/2023] Open
Abstract
Melatonin confers cardioprotective effects on multiple cardiovascular diseases, including doxorubicin-induced cardiomyopathy. The effectiveness of melatonin in mitigating myocardial injuries caused by Doxorubicin through enhancement of mitochondrial function is already established, however, the role of melatonin in regulating the Sirtuin-1 (Sirt1)/Nuclear factor E2-associated factor 2 (Nrf2) pathway in lessening the onset of Doxorubicin-induced cardiomyopathy is yet to be elucidated. To address this, H9C2 cardiomyocytes and C57BL/6 mice were employed to construct in vitro and in vivo models of Dox-induced myocardial impairments, respectively. Results showed that Dox markedly evoked oxidative stress, pyroptosis and apoptosis both in vitro and in vivo, which were significantly alleviated by melatonin administration. Mechanistically, melatonin attenuated Dox-induced downregulation of Sirt1 and Nrf2, and both inhibition of Sirt1 and Nrf2 significantly reversed the cardioprotective effects of melatonin. In conclusion, our studies suggest that the activation of the Sirt1/Nrf2 pathway is the underlying mechanism behind melatonin's ability to curtail oxidative stress, pyroptosis, and apoptosis in Dox-induced cardiomyopathy. These promising results demonstrated the potential application of melatonin as a treatment for doxorubicin-induced cardiac injury.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China.
| |
Collapse
|
30
|
Zeng H, Yang Y, Tou F, Zhan Y, Liu S, Zou P, Chen Y, Shao L. Bone marrow stromal cell-derived exosomes improve oxidative stress and pyroptosis in doxorubicin-induced myocardial injury in vitro by regulating the transcription of GSDMD through the PI3K-AKT-Foxo1 pathway. Immun Inflamm Dis 2023; 11:e810. [PMID: 36988259 PMCID: PMC10042126 DOI: 10.1002/iid3.810] [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: 11/08/2022] [Revised: 01/13/2023] [Accepted: 01/20/2023] [Indexed: 03/29/2023] Open
Abstract
OBJECTIVES Doxorubicin (DOX) can contribute to severe myocardial injury, and bone marrow stromal cells (BMSC)-exosomes (Exos) improves acute myocardial infarction. Hence, this research investigated whether BMSC-Exos alleviated DOX-induced myocardial injury. METHODS BMSC-derived Exos were isolated and identified, and the optimal concentration of DOX was confirmed. H9C2 cells were treated with DOX and BMSC-Exos or in combination with the protein kinase B (AKT) inhibitor. Reactive oxygen species (ROS) and JC-1 were detected to assess oxidative stress (OS) and mitochondrial membrane damage, respectively. In addition, the expression of pyroptosis-related molecules was measured. The expression of phosphatidylinositol 3 kinase (PI3K)-AKT pathway-related proteins and the phosphorylation and acetylation of forkhead box O1 (Foxo1) in the cell nucleus and cytoplasm were tested. Last, interactions between Foxo1 and gasdermin D (GSDMD) were assessed. RESULTS BMSC-Exo treatment increased viability and mitochondrial membrane potential and reduced lactic dehydrogenase release and ROS levels in DOX-treated H9C2 cells. Furthermore, the addition of BMSC-Exos suppressed DOX-induced activation and upregulation of NLRP3 and apoptosis-associated speck-like protein containing A CARD (ASC) and in vitro cleavage of caspase-1, GSDMD, interleukin (IL)-1β, and IL-18 proteins. Additionally, BMSC-Exo treatment enhanced the expression of phosphorylated (p)-PI3K, p-AKT, and p-mTOR in DOX-treated H9C2 cells and the levels of phosphorylated Foxo1 in the cytoplasm of DOX-treated H9C2 cells. Foxo1 was enriched in the promoter region of GSDMD. Moreover, the AKT inhibitor API-2 annulled the effects of BMSC-Exos on OS, pyroptosis, and Foxo1 phosphorylation in DOX-treated H9C2 cells. CONCLUSIONS BMSC-Exos phosphorylated Foxo1 and inactivated Foxo1 transcription via the PI3K-AKT pathway to diminish GSDMD expression, thus restraining DOX-induced pyroptosis and OS of myocardial cells.
Collapse
Affiliation(s)
- Hong Zeng
- Department of Cardiology, Jiangxi Provincial People's HospitalThe First Affiliated Hospital of Nanchang Medical CollegeNanchangJiangxiPeople's Republic of China
| | - Yong Yang
- Department of Cardiology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiPeople's Republic of China
| | - Fangfang Tou
- Jiangxi Provincial People's HospitalThe First Affiliated Hospital of Nanchang Medical CollegeNanchangJiangxiPeople's Republic of China
| | - Yuliang Zhan
- Department of Cardiology, Jiangxi Provincial People's HospitalThe First Affiliated Hospital of Nanchang Medical CollegeNanchangJiangxiPeople's Republic of China
| | - Songtao Liu
- Department of Cardiology, Jiangxi Provincial People's HospitalThe First Affiliated Hospital of Nanchang Medical CollegeNanchangJiangxiPeople's Republic of China
| | - Pengtao Zou
- Department of Cardiology, Jiangxi Provincial People's HospitalThe First Affiliated Hospital of Nanchang Medical CollegeNanchangJiangxiPeople's Republic of China
| | - Yanmei Chen
- Department of Cardiology, Jiangxi Provincial People's HospitalThe First Affiliated Hospital of Nanchang Medical CollegeNanchangJiangxiPeople's Republic of China
| | - Liang Shao
- Department of Cardiology, Jiangxi Provincial People's HospitalThe First Affiliated Hospital of Nanchang Medical CollegeNanchangJiangxiPeople's Republic of China
| |
Collapse
|
31
|
Nifuroxazide mitigates doxorubicin-induced cardiovascular injury: Insight into oxidative/NLRP3/GSDMD-mediated pyroptotic signaling modulation. Life Sci 2023; 314:121311. [PMID: 36549350 DOI: 10.1016/j.lfs.2022.121311] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
Doxorubicin (DOX) is a widely used powerful anthracycline for treatment of many varieties of malignancies; however its cumulative and dose-dependent cardio-toxicity has been limited its clinical use. In the current study, in vivo and in vitro (neonatal rat's cardiomyocytes) experiments were conducted to identify the impact of nifuroxazide (NIFU) on DOX-induced cardiomyopathy, vascular injury, and hemato-toxcity and plot the underlying regulatory mechanisms. Cardiovascular injury was induced in vivo by I.P. injection of an overall dose of DOX (21 mg/kg) administered (3.5 mg/kg) twice weekly for 21 days. NIFU (10 and 30 mg/kg) was administered orally once daily for 21 days, 1 week after DOX injection initiation. In vivo experiments confirmed NIFU to restore blood cells counts and hemoglobin concentration. Moreover, NIFU normalized the myocardial functional status as confirmed by ECG examination and myocardial injury markers; CK-MB, LDH, and AST. NIFU restored the balance between TAC and both of ROS and MDA and down-regulated the protein expression of TLR4, NF-kB, TXNIP, NLR-family pyrin domain containing 3 (NLRP3), caspase-1, IL-1β, and GSDMD-N terminal, with inhibition of the up-stream of NLRP3 and the down-stream DOX-induced pyroptosis. The in vitro assay confirmed well preserved cardiomyocytes' architecture, amelioration of NLRP3/IL-1 β-mediated cell pyroptosis, enhanced cell viability, and improved spontaneous beating. Moreover, NIFU normalized the disturbed aortic oxidant-antioxidant balance; enhanced eNOS- mediated endothelial relaxation, and down regulated IL-1β expression. Thus, NIFU may be proposed to serve as a cardioprotective agent to attenuate DOX-induced cardio-toxicity and vascular injury.
Collapse
|
32
|
Lv XF, Wen RQ, Liu K, Zhao XK, Pan CL, Gao X, Wu X, Zhi XD, Ren CZ, Chen QL, Lu WJ, Bai TY, Li YD. Role and molecular mechanism of traditional Chinese medicine in preventing cardiotoxicity associated with chemoradiotherapy. Front Cardiovasc Med 2022; 9:1047700. [PMID: 36419486 PMCID: PMC9678083 DOI: 10.3389/fcvm.2022.1047700] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 10/20/2022] [Indexed: 08/12/2023] Open
Abstract
Cardiotoxicity is a serious complication of cancer therapy. It is the second leading cause of morbidity and mortality in cancer survivors and is associated with a variety of factors, including oxidative stress, inflammation, apoptosis, autophagy, endoplasmic reticulum stress, and abnormal myocardial energy metabolism. A number of studies have shown that traditional Chinese medicine (TCM) can mitigate chemoradiotherapy-associated cardiotoxicity via these pathways. Therefore, this study reviews the effects and molecular mechanisms of TCM on chemoradiotherapy-related cardiotoxicity. In this study, we searched PubMed for basic studies on the anti-cardiotoxicity of TCM in the past 5 years and summarized their results. Angelica Sinensis, Astragalus membranaceus Bunge, Danshinone IIA sulfonate sodium (STS), Astragaloside (AS), Resveratrol, Ginsenoside, Quercetin, Danggui Buxue Decoction (DBD), Shengxian decoction (SXT), Compound Danshen Dripping Pill (CDDP), Qishen Huanwu Capsule (QSHWC), Angelica Sinensis and Astragalus membranaceus Bunge Ultrafiltration Extract (AS-AM),Shenmai injection (SMI), Xinmailong (XML), and nearly 60 other herbs, herbal monomers, herbal soups and herbal compound preparations were found to be effective as complementary or alternative treatments. These preparations reduced chemoradiotherapy-induced cardiotoxicity through various pathways such as anti-oxidative stress, anti-inflammation, alleviating endoplasmic reticulum stress, regulation of apoptosis and autophagy, and improvement of myocardial energy metabolism. However, few clinical trials have been conducted on these therapies, and these trials can provide stronger evidence-based support for TCM.
Collapse
Affiliation(s)
- Xin-Fang Lv
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Ruo-Qing Wen
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
| | - Kai Liu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Xin-Ke Zhao
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Chen-Liang Pan
- The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiang Gao
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Xue Wu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
- Lanzhou University Second Hospital, Lanzhou, China
| | - Xiao-Dong Zhi
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Chun-Zhen Ren
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
| | - Qi-Lin Chen
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
| | - Wei-Jie Lu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
| | - Ting-Yan Bai
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
| | - Ying-Dong Li
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
33
|
Gasdermin D mediates endoplasmic reticulum stress via FAM134B to regulate cardiomyocyte autophagy and apoptosis in doxorubicin-induced cardiotoxicity. Cell Death Dis 2022; 13:901. [PMID: 36289195 PMCID: PMC9606128 DOI: 10.1038/s41419-022-05333-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 10/05/2022] [Accepted: 10/10/2022] [Indexed: 01/23/2023]
Abstract
Cardiomyocyte pyroptosis and apoptosis play a vital role in the pathophysiology of several cardiovascular diseases. Our recent study revealed that gasdermin D (GSDMD) can promote myocardial I/R injury via the caspase-11/GSDMD pathway. We also found that GSDMD deletion attenuated myocardial I/R and MI injury by reducing cardiomyocyte apoptosis and pyroptosis. However, how GSDMD mediates cardiomyocyte apoptosis and protects myocardial function remains unclear. Here, we found that doxorubicin (DOX) treatment resulted in increased apoptosis and pyroptosis in cardiomyocytes and that caspase-11/GSDMD could mediate DOX-induced cardiotoxicity (DIC) injury. Interestingly, GSDMD overexpression promoted cardiomyocyte apoptosis, which was attenuated by GSDMD knockdown. Notably, GSDMD overexpression exacerbated DIC injury, impaired cardiac function in vitro and in vivo, and enhanced DOX-induced cardiomyocyte autophagy. Mechanistically, GSDMD regulated the activity of FAM134B, an endoplasmic reticulum autophagy receptor, by pore formation on the endoplasmic reticulum membrane via its N-terminus, thus activating endoplasmic reticulum stress. In turn, FAM134B interacted with autophagic protein LC3, thus inducing cardiac autophagy, promoting cardiomyocyte apoptosis, and aggravating DIC. These results suggest that GSDMD promotes autophagy and induces cardiomyocyte apoptosis by modulating the reaction of FAM134B and LC3, thereby promoting DIC injury. Targeted regulation of GSDMD may be a new target for the prevention and treatment of DIC.
Collapse
|
34
|
Habimana O, Modupe Salami O, Peng J, Yi GH. Therapeutic Implications of Targeting Pyroptosis in Cardiac-related Etiology of Heart Failure. Biochem Pharmacol 2022; 204:115235. [PMID: 36044938 DOI: 10.1016/j.bcp.2022.115235] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/26/2022]
Abstract
Heart failure remains a considerable clinical and public health problem, it is the dominant cause of death from cardiovascular diseases, besides, cardiovascular diseases are one of the leading causes of death worldwide. The survival of patients with heart failure continues to be low with 45-60% reported deaths within five years. Apoptosis, necrosis, autophagy, and pyroptosis mediate cardiac cell death. Acute cell death is the hallmark pathogenesis of heart failure and other cardiac pathologies. Inhibition of pyroptosis, autophagy, apoptosis, or necrosis reduces cardiac damage and improves cardiac function in cardiovascular diseases. Pyroptosis is a form of inflammatory deliberate cell death that is characterized by the activation of inflammasomes such as NOD-like receptors (NLR), absent in melanoma 2 (AIM2), interferon-inducible protein 16 (IFI-16), and their downstream effector cytokines: Interleukin IL-1β and IL-18 leading to cell death. Recent studies have shown that pyroptosis is also the dominant cell death process in cardiomyocytes, cardiac fibroblasts, endothelial cells, and immune cells. It plays a crucial role in the pathogenesis of cardiac diseases that contribute to heart failure. This review intends to summarize the therapeutic implications targeting pyroptosis in the main cardiac pathologies preceding heart failure.
Collapse
Affiliation(s)
- Olive Habimana
- International College, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China
| | | | - Jinfu Peng
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China; Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China
| | - Guang-Hui Yi
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China; Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China.
| |
Collapse
|