1
|
Chen S, Li P, Shi K, Tang S, Zhang W, Peng C, Li T, Xie H, Liu C, Zhou J. Tanshinone IIA promotes ferroptosis in cutaneous melanoma via STAT1-mediated upregulation of PTGS2 expression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156702. [PMID: 40222167 DOI: 10.1016/j.phymed.2025.156702] [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: 12/27/2024] [Revised: 03/10/2025] [Accepted: 03/26/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Melanoma is highly aggressive, metastatic with a poor prognosis. Despite significant advances in targeted therapies and immunotherapies, their efficiency limited by drug resistance. Tanshinone IIA (Tan IIA), a bioactive compound derived from Traditional Chinese plant, exhibits significant anticancer potential, which still needs more research in its complex regulatory mechanisms. PURPOSE This study aimed to elucidate the putative targets and regulatory mechanisms of Tan IIA in anti-melanoma, with a focus on its role in inducing ferroptosis. STUDY DESIGN We designed the experiment to explore the effects of Tan IIA on melanoma through both in vitro and in vivo experiments and to investigate the underlying mechanisms through transcriptomics combining network pharmacology analysis. METHOD Ferroptosis monitored by Malondialdehyde (MDA), Fe2+, reactive oxygen species (ROS) and glutathione (GSH) in vivo and in vitro. RNA sequence was performed to explore the key regulatory pathways involved in Tan IIA-induced ferroptosis. Chromatin immunoprecipitation (ChIP) and Luciferase assays were used to validate transcription factor responsible for prostaglandin-endoperoxide synthase 2 (PTGS2) regulation. Additionally, RT-qPCR, western blot, IF, IHC were aimed to evaluate the expression of target gene. RESULT Tan IIA markedly suppresses melanoma growth in a xenograft model. The same effect performed on inhibition melanoma cells and promotion to ferroptosis with accumulation of ROS, MDA, and Fe²⁺levels and GSH consumption. RNA sequencing and public database analysis revealed that Tan IIA regulates PTGS2, the critical marker of ferroptosis, and PTGS2-knockdown attenuates Tan IIA -induced ferroptosis in melanoma cells. Furthermore, we identified that Tan IIA stimulate signal transducer and activator of transcription 1 (STAT1), a transcription factor, promoting PTGS2 expression and localized in the cell cytoplasm. Moreover, downregulation of the transcription factor STAT1 lead to PTGS2 downregulation and also inhibit ferroptosis in melanoma. CONCLUSION This study, the first to link Tan IIA-induced ferroptosis to the STAT1/PTGS2 axis in melanoma, identifies STAT1 and PTGS2 as novel therapeutic targets for melanoma, which demonstrates the potential of natural compounds Tan IIA in overcoming drug resistance and integrates traditional medicine with advanced molecular techniques for mechanistic exploration.
Collapse
Affiliation(s)
- Shuyue Chen
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Peiting Li
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Ke Shi
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Shijie Tang
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Wancong Zhang
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Tianyu Li
- Department of Burns and Plastic Surgery, Nanshi Hospital of Nanyang, Nanyang, Henan Province 473000, China
| | - Huiqing Xie
- Department of Rehabilitation, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Can Liu
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China.
| | - Jianda Zhou
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China.
| |
Collapse
|
2
|
Zhang Y, Yao Y, Zhang Q, Yang B. Traditional Chinese Medicine for Inhibiting Ferroptosis in Ischemic-Related Diseases. Basic Clin Pharmacol Toxicol 2025; 136:e70039. [PMID: 40296341 DOI: 10.1111/bcpt.70039] [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: 12/31/2024] [Revised: 03/28/2025] [Accepted: 04/06/2025] [Indexed: 04/30/2025]
Abstract
Ischemic-related diseases, such as myocardial infarction and stroke, are primarily driven by a deficit in oxygen supply leading to cellular damage and death. Ferroptosis has emerged as an important mechanism contributing to the progression of ischemic injury, characterized by iron-dependent lipid peroxidation. This review aims to provide a comprehensive overview of the significant mechanisms underlying ferroptosis in ischemic conditions and explores the potential effects of traditional Chinese medicines (TCMs) and their extracts. Numerous compounds extracted from TCMs, including flavonoids, polyphenols and terpenes, exhibit potent antiferroptotic effects by activating nuclear factor erythroid 2-related factor 2, upregulating glutathione peroxidase 4, inhibiting lipid peroxidation and so on. These properties render TCMs a promising candidate for developing novel ferroptosis therapeutic strategies. This review underscores the importance of investigating the interactions between ferroptosis and TCMs within the context of ischemic diseases. These findings provide valuable insights for future research to identify targets associated with ferroptosis regulation, thereby expanding the pharmacological perspective of TCMs in treating ischemic diseases and revealing the potential of novel therapeutic strategies. Additionally, this highlights the relevance of integrating traditional and modern medical approaches in addressing complex health issues.
Collapse
Affiliation(s)
- Yukun Zhang
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing, China
| | - Yang Yao
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing, China
| | - Qiaoling Zhang
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing, China
| | - Baoxue Yang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
3
|
Mai L, Liu J, Wu H, Wang H, Lin Z, Rao S, Sun W, Tan A, Lin Y, Chen B. Enhanced inhibition of neuronal ferroptosis and regulation of microglial polarization with multifunctional traditional Chinese medicine active ingredients-based selenium nanoparticles for treating spinal cord injury. Mater Today Bio 2025; 32:101758. [PMID: 40270889 PMCID: PMC12017924 DOI: 10.1016/j.mtbio.2025.101758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 03/23/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
Spinal cord injury (SCI) is a devastating condition that results in the loss of sensory and motor functions. The complex pathogenesis of SCI contributes to the limited availability of effective therapies. Two major factors exacerbating secondary injury in SCI are neuronal ferroptosis and microglial inflammatory polarization. Tanshinone IIA (TSIIA) has demonstrated a significant anti-ferroptosis effect by inhibiting lipid peroxidation, while tetramethylpyrazine (TMP) exhibits remarkable anti-inflammatory properties by promoting the shift of microglial polarization from the M1 to the M2 phenotype. However, most drugs currently under development primarily target a single aspect of this multifaceted condition, which is insufficient for comprehensive treatment. Selenium nanoparticles have emerged as a promising therapeutic strategy due to their ability to encapsulate various agents, effectively targeting diverse pathophysiological mechanisms while offering favorable water solubility and low toxicity. In this study, we developed a novel nanocarrier functionalized with astragalus polysaccharide (APS) and loaded with TSIIA and TMP. Results from both in vitro and in vivo studies indicate that TSIIA/TMP/APS@Se NPs possess anti-ferroptosis properties and can regulate microglial polarization, potentially enhancing functional recovery following SCI. In summary, this study presents a promising alternative strategy for treating SCI, highlighting its significant potential for future clinical applications.
Collapse
Affiliation(s)
- Luoqi Mai
- Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Jinggong Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
- Orthopedics Department, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Huimei Wu
- Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
- Dermatology Department, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Hongshen Wang
- Orthopedics Department, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Zhidong Lin
- Orthopedics Department, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Siyuan Rao
- Orthopedics Department, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Wenxi Sun
- Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Aowei Tan
- Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Yongpeng Lin
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
- Orthopedics Department, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Bolai Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
- Orthopedics Department, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| |
Collapse
|
4
|
Gong G, Wan W, Zhang X, Chen X, Yin J. Management of ROS and Regulatory Cell Death in Myocardial Ischemia-Reperfusion Injury. Mol Biotechnol 2025; 67:1765-1783. [PMID: 38852121 DOI: 10.1007/s12033-024-01173-y] [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: 12/13/2023] [Accepted: 04/02/2024] [Indexed: 06/10/2024]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is fatal to patients, leading to cardiomyocyte death and myocardial remodeling. Reactive oxygen species (ROS) and oxidative stress play important roles in MIRI. There is a complex crosstalk between ROS and regulatory cell deaths (RCD) in cardiomyocytes, such as apoptosis, pyroptosis, autophagy, and ferroptosis. ROS is a double-edged sword. A reasonable level of ROS maintains the normal physiological activity of myocardial cells. However, during myocardial ischemia-reperfusion, excessive ROS generation accelerates myocardial damage through a variety of biological pathways. ROS regulates cardiomyocyte RCD through various molecular mechanisms. Targeting the removal of excess ROS has been considered an effective way to reverse myocardial damage. Many studies have applied antioxidant drugs or new advanced materials to reduce ROS levels to alleviate MIRI. Although the road from laboratory to clinic has been difficult, many scholars still persevere. This article reviews the molecular mechanisms of ROS inhibition to regulate cardiomyocyte RCD, with a view to providing new insights into prevention and treatment strategies for MIRI.
Collapse
Affiliation(s)
- Ge Gong
- Department of Geriatrics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 211002, China
| | - Wenhui Wan
- Department of Geriatrics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 211002, China
| | - Xinghu Zhang
- Department of Geriatrics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 211002, China
| | - Xiangxuan Chen
- Department of Cardiology, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211100, China.
| | - Jian Yin
- Department of Orthopedics, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211100, China.
- Department of Orthopedics, Jiangning Clinical Medical College of Jiangsu Medical Vocational College, Nanjing, 211100, China.
- Department of Orthopedics, Jiangning Clinical Medical College of Nanjing Medical University Kangda College, Nanjing, 211100, China.
| |
Collapse
|
5
|
Li T, Xu X, Zhou T, Xie X, Peng Y, He L, He J, Luo W. ETS1 modulates ferroptosis to affect the process of myocardial ischemia-reperfusion injury via PIM3. Exp Cell Res 2025; 448:114550. [PMID: 40189183 DOI: 10.1016/j.yexcr.2025.114550] [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: 01/14/2025] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 04/13/2025]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is a common complication of cardiovascular disease and its pathogenesis remains unclear. ETS1 (E26 transformation-specific sequence-1) is a transcription factor that plays an important regulatory role in vascular development and generation. Therefore, this study aims to explore the role of ETS1 in MIRI and its potential molecular mechanisms. An OGD/R-induced H9C2 cardiomyocyte model was established, and cell viability was determined by CCK8 and changes in SOD, MDA and GSH levels by ELISA; expression of ETS1, PIM3 and ferroptosis-related indices were determined by immunofluorescence, Western blot and qPCR; at the same time, a mouse MIRI model was established to assess changes in myocardial injury and changes in ferroptosis after knockdown of ETS1. In the OGD/R-induced H9C2 cell model, cell viability was significantly lower than that of the control group, and the level of intracellular ferroptosis was significantly enhanced. Further research has revealed that in the OGD/R-induced H9C2 model, the expression of ETS1 is significantly upregulated. Knockdown of ETS1 can reverse the myocardial cell injury induced by OGD/R. Mechanistically, ETS1 promotes the progression of MIRI by targeting and regulating PIM3, thereby exacerbating ferroptosis. Additionally, in a mouse MIRI model, the knockdown of ETS1 significantly enhances the expression of GPX4, SLC7A11, and FTH1 proteins, inhibits the ferroptosis process, and thereby improves MIRI in mice. The research results indicate that ETS1 promotes the MIRI process through regulating ferroptosis of cardiomyocytes mediated by PIM3. This discovery provides important scientific evidence for further elucidating the mechanisms underlying MIRI and developing therapeutic strategies.
Collapse
Affiliation(s)
- Ting Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Xuefeng Xu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Tao Zhou
- Department of Cardiovascular Medicine, The First Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Xuan Xie
- Department of Cardiovascular Medicine, The First Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Yutao Peng
- Department of Cardiovascular Medicine, The First Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Li He
- Department of Cardiovascular Medicine, The First Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Jin He
- Department of Cardiovascular Medicine, The First Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, People's Republic of China.
| | - Wangsheng Luo
- Department of Cardiovascular Medicine, The First Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, People's Republic of China.
| |
Collapse
|
6
|
Wu Z, Xu Y, Guo X, Zhang Z, Wang J, Tang Y. Assessment of Tanshinone IIA Derivatives for Cardioprotection in Myocardial Ischemic Injury. PLANTA MEDICA 2025. [PMID: 40127683 DOI: 10.1055/a-2565-8285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Tanshinone ⅡA (TSA), a component of traditional Chinese medicine, effectively protects against myocardial injury. However, its clinical application is limited by poor water solubility and a short half-life. In this study, we report on four TSA derivatives designed and synthesized by our research group. The protective activity against hypoxia-reoxygenation injury in cells was evaluated, and derivative Ⅰ-3 was selected for in vivo experiments to verify its myocardial protective activity in rats with myocardial infarction. The results demonstrated that these four compounds could protect neonatal rat cardiomyocytes from hypoxia-reoxygenation injury. Among the derivatives, Ⅰ-3 showing superior protective effects, we found that Ⅰ-3 has enhanced metabolic stability and an extended half-life. Ⅰ-3 exhibited superior biological activity, effectively reducing the heart infarction area, alleviating myocardial hypertrophy, and enhancing cardiac pumping function. Ⅰ-3 reported in the present work represents a novel and effective derivative of TSA, showing great potential for the treatment of myocardial ischemia (MI).
Collapse
Affiliation(s)
- Zhiwu Wu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P. R. China
| | - Ying Xu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P. R. China
| | - Ximing Guo
- School of Pharmacy, China Pharmaceutical University, Nanjing, P. R. China
| | - Zhilan Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing, P. R. China
| | - Jinxin Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing, P. R. China
| | - Yiqun Tang
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P. R. China
| |
Collapse
|
7
|
Zheng X, Tang X, Xu Y, Zhu H, Zhong L, Chen C, Cui J, Zhou J. Sodium aescinate induces hepatotoxicity through apoptosis and ferroptosis by inhibiting the Nrf2/CTH pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119608. [PMID: 40064321 DOI: 10.1016/j.jep.2025.119608] [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: 12/30/2024] [Revised: 02/08/2025] [Accepted: 03/07/2025] [Indexed: 03/15/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The seed of Aesculus wilsonii Rehd., also known as Suoluozi in China, is a traditional Chinese herb included in the Pharmacopoeia of China (2020). Sodium aescinate (SA) is derived from the Aesculus wilsonii Rehd.'s seeds and is extensively used in clinical practice. AIM OF THE STUDY The study investigated the involvement of the Nrf2/CTH pathway in SA-induced hepatotoxicity and explored potential strategies for alleviating SA-induced liver damage. MATERIALS AND METHODS The ICR mice and AML12 mouse hepatocytes were exposed to SA. The levels of Fe2+, cysteine (Cys), glutathione (GSH), hydrogen sulfide (H2S), ROS, lipid peroxides and caspase-3 activity were assessed. The effects of SA on signaling pathways related to ferroptosis and apoptosis were examined. Furthermore, genetic modification or agonists of Nrf2 and CTH were co-treated with SA. RESULTS SA triggered ferroptosis and apoptosis in AML12 cells and mouse livers, characterized by a decline in Cys, GSH, and H2S levels, as well as accumulation of Fe2+, ROS and lipid peroxides, mitochondrial dysfunction, and chromatin condensation. SA decreased Nrf2, CTH, and Bcl-2 levels, elevated Bax levels, and activated caspase-9/3. Overexpression of Nrf2 or CTH, or NAC supplementation alleviated SA-induced ferroptosis by upregulating Cys and GSH levels. Overexpression of Nrf2 or CTH, or NaHS supplementation increased H2S levels, which reduced the interaction between p616-Drp1 and VDAC1 by enhancing Drp1 S-sulfenylation, thereby alleviating SA-induced mitochondrial-dependent apoptosis. Furthermore, DMF or Met mitigated SA-induced hepatotoxicity by activating the Nrf2/CTH pathway. CONCLUSIONS SA triggers oxidative stress, mitochondrial dysfunction, apoptosis, and ferroptosis, ultimately leading to liver damage by suppressing the Nrf2/CTH pathway.
Collapse
Affiliation(s)
- Xin Zheng
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Xinyi Tang
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Yinan Xu
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Haiyan Zhu
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Lianwei Zhong
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Chen Chen
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Jiajun Cui
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Jie Zhou
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China.
| |
Collapse
|
8
|
Shahrokhi H, Asili J, Tayarani-Najaran Z, Boozari M. Signaling pathways behind the biological effects of tanshinone IIA for the prevention of cancer and cardiovascular diseases. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03857-x. [PMID: 39937254 DOI: 10.1007/s00210-025-03857-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 01/24/2025] [Indexed: 02/13/2025]
Abstract
Tanshinone IIA (Tan IIA) is a well-known fat-soluble diterpenoid found in Salvia miltiorrhiza, recognized for its various biological effects. The molecular signaling pathways of Tan IIA have been investigated in different diseases, including the anti-inflammatory, hepatoprotective, renoprotective, neuroprotective effects, and fibrosis prevention. This article provides a brief overview of the signaling pathways related to anti-cancer and cardioprotective effects of Tan IIA. It shows that Tan IIAs anti-cancer ability has good expectation through multiplicity mechanisms affecting various aspects' tumor biology. The major pathways involved in its anti-cancer effects include inhibition of PI3/Akt, MAPK, and p53/p21 signaling which leads to enhancement of immune responses and increased radiation sensitivity. Some essential pathways responsible for cardioprotective effects induced by Tan IIA are PI3/AKT activation, MAPK, and SIRT1 promoting protection against ischemia/reperfusion injury in myocardial cells as well as inhibiting pathological remodeling processes. Finally, the article underscores the complex and specific signaling pathways influenced by Tan IIA. The PI3/Akt and MAPK pathways play critical roles in the anti-cancer and cardioprotective effects of Tan IIA. Particularly, Tan IIA suppresses the proliferation of malignancies in cancerous cells but stimulates protective mechanisms in normal cardiovascular cells. These findings highlight the importance of investigating molecular signaling pathways in evaluating the therapeutic potential of natural products. Studying about signaling pathways is vital in understanding the therapeutic aspects of Tan IIA and its derivatives as anti-cancer and cardio-protective agents. Further research is necessary to understand these complex mechanisms.
Collapse
Affiliation(s)
- Homa Shahrokhi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Javad Asili
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Tayarani-Najaran
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Motahareh Boozari
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Chuan'ai C, Haolong L, Pengpeng A, Yang Y, Chunyan L, Yumiao Y. VDAC1 Cleavage Promotes Autophagy in Renal Tubular Epithelial Cells With Hypoxia/Reoxygenation Injury. Nephrology (Carlton) 2025; 30:e70000. [PMID: 39853618 DOI: 10.1111/nep.70000] [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: 05/06/2024] [Revised: 12/24/2024] [Accepted: 01/07/2025] [Indexed: 01/26/2025]
Abstract
AIM To study the effect and elucidate the underlying mechanisms of VDAC1-ΔC on autophagy in renal tubular epithelial cells injured by hypoxia/reoxygenation. METHODS C57/BL6 mice were randomly divided into groups: sham operation group, IRI 1d group and IRI 2d group. The inner canthal blood of mice was collected to detect the levels of serum creatinine and urea nitrogen and kidney tissues were sampled, and sections were stained with Periodic acid-Schiff for morphological evaluation. The expression of VDAC1 in kidney tissue was detected by Western blot. An immortalised human proximal tubular epithelial cell line, HK-2 cells, were subjected to hypoxia/reoxygenation treatment. HK-2 cells were incubated under hypoxia for 6 h, followed by 6 and 24 h of reoxygenation, cells were divided into four groups: H6/R0 group, H6/R6 group, H6/R24 group and control group. The release of LDH and cytosolic ROS were assessed, the expression of autophagy-related proteins LC3 and p62 was detected by Western blot, autophagy flux was monitored by transfecting mRFP-GFP-LC3 lentivirus in HK2 cells, and cells were pretreated with bafilomycin A1 to further monitor the autophagy flux. VDAC1-cleavage-defective mutant in HK-2 cells silencing VDAC1 was established to examine the effect of VDAC1 cleavage on autophagy and hypoxia/reoxygenation injury. RESULTS In vivo, IRI 1d/2d promoted the disorder of renal tubular structure and the cleavage of VDAC1 in kidney tissue; in vitro, hypoxia/reoxygenation promoted cytosolic ROS accumulation, LDH release, VDAC1 cleavage and induced autophagy and autophagic flux; reduced VDAC1 cleavage inhibited autophagy; and decreased cytosolic ROS accumulation and LDH release, thus alleviated cell injury. CONCLUSION In renal tubular epithelial cells injured by H/R, VDAC1 cleavage was increased, triggering an autophagic response, and VDAC1 cleavage promoted autophagy to regulate cell injury.
Collapse
Affiliation(s)
- Chen Chuan'ai
- Department of Quality Management, Tianjin Blood Center, Tianjin, China
| | - Li Haolong
- Department of Quality Management, Tianjin Blood Center, Tianjin, China
| | - An Pengpeng
- Department of Quality Management, Tianjin Blood Center, Tianjin, China
| | - Yu Yang
- Department of Quality Management, Tianjin Blood Center, Tianjin, China
| | - Liu Chunyan
- Department of Quality Management, Tianjin Blood Center, Tianjin, China
| | - Yang Yumiao
- Department of Quality Management, Tianjin Blood Center, Tianjin, China
| |
Collapse
|
10
|
Wang K, Zhu Q, Liu W, Wang L, Li X, Zhao C, Wu N, Ma C. Mitochondrial apoptosis in response to cardiac ischemia-reperfusion injury. J Transl Med 2025; 23:125. [PMID: 39875870 PMCID: PMC11773821 DOI: 10.1186/s12967-025-06136-8] [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: 10/10/2024] [Accepted: 01/13/2025] [Indexed: 01/30/2025] Open
Abstract
In patients with acute myocardial infarction (AMI), thrombolytic therapy and revascularization strategies allow complete recanalization of occluded epicardial coronary arteries. However, approximately 35% of patients still experience myocardial ischemia/reperfusion (I/R) injury, which contributing to increased AMI mortality. Therefore, an accurate understanding of myocardial I/R injury is important for preventing and treating AMI. The death of each cell (cardiomyocytes, endothelial cells, vascular smooth muscle cells, cardiac fibroblasts, and mesenchymal stem cells) after myocardial ischemia/reperfusion is associated with apoptosis due to mitochondrial dysfunction. Abnormal opening of the mitochondrial permeability transition pore, aberrant mitochondrial membrane potential, Ca2+ overload, mitochondrial fission, and mitophagy can lead to mitochondrial dysfunction, thereby inducing mitochondrial apoptosis. The manifestation of mitochondrial apoptosis varies according to cell type. Here, we reviewed the characteristics of mitochondrial apoptosis in cardiomyocytes, endothelial cells, vascular smooth muscle cells, cardiac fibroblasts, and mesenchymal stem cells following myocardial ischemia/reperfusion.
Collapse
Affiliation(s)
- Kaixin Wang
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, China
- Clinical Medical Research Center of Imaging in Liaoning Province, Shenyang, China
| | - Qing Zhu
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, China
- Clinical Medical Research Center of Imaging in Liaoning Province, Shenyang, China
| | - Wen Liu
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, China
- Clinical Medical Research Center of Imaging in Liaoning Province, Shenyang, China
| | - Linyuan Wang
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, China
- Clinical Medical Research Center of Imaging in Liaoning Province, Shenyang, China
| | - Xinxin Li
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, China
- Clinical Medical Research Center of Imaging in Liaoning Province, Shenyang, China
| | - Cuiting Zhao
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, China
- Clinical Medical Research Center of Imaging in Liaoning Province, Shenyang, China
| | - Nan Wu
- The Central Laboratory of The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Chunyan Ma
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, China.
- Clinical Medical Research Center of Imaging in Liaoning Province, Shenyang, China.
| |
Collapse
|
11
|
Wu T, Li D, Chen Q, Kong D, Zhu H, Zhou H, Zhang Q, Cui G. Identification of VDAC1 as a cardioprotective target of Ginkgolide B. Chem Biol Interact 2025; 406:111358. [PMID: 39716534 DOI: 10.1016/j.cbi.2024.111358] [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: 09/21/2024] [Revised: 11/18/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024]
Abstract
Ginkgolide B (GB), a compound derived from Ginkgo biloba, exhibits significant cardioprotective properties, although its precise molecular target has yet to be identified. In this study, we synthesized a biotin-labeled GB probe (GB-biotin) to identify the molecular targets of GB. Our experiments demonstrated that treatment with GB or GB-biotin reduced mitochondrial injury, restored mitochondrial membrane potential, and decreased cell apoptosis in a concentration-dependent manner. Additionally, GB increased mitochondrial oxygen consumption rate, indicating improved mitochondrial bioenergetics. Proteomic analysis revealed that the voltage-dependent anion channel 1 (VDAC1) is a key protein that interacts with GB-biotin. This finding was further confirmed through cellular thermal shift assay (CETSA) and molecular docking, which revealed hydrogen bond formation between GB and VDAC1. Furthermore, overexpression of VDAC1 diminished the protective effects of GB, highlighting the crucial role of VDAC1 in GB-mediated cardioprotection. These findings identify VDAC1 as a therapeutic target for GB in vitro, providing valuable insights into the cardioprotective mechanisms of GB and the development of novel cardioprotective strategies.
Collapse
Affiliation(s)
- Tingbiao Wu
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, China
| | - Deyao Li
- Guangzhou Henovcom Biosciences Co., Ltd., Guangzhou, 510535, China
| | - Qiuyu Chen
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, China
| | - Dezhi Kong
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, 050000, China
| | - Hongxuan Zhu
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, China
| | - Hefeng Zhou
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, China
| | - Qingwen Zhang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, 999078, China
| | - Guozhen Cui
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, China.
| |
Collapse
|
12
|
Cao Y, Zhao H, Lin S, Chen J, Xiong J, Zeng Z, Long Z, Su Y, Zhong Y, Zhao L, Zhang M, Wu J, Zhou Y, Zhou J. Danshen injection ameliorates unilateral ureteral obstruction-induced renal fibrosis by inhibiting ferroptosis via activating SIRT1/GPX4 pathway. Front Pharmacol 2025; 15:1503628. [PMID: 39872048 PMCID: PMC11770031 DOI: 10.3389/fphar.2024.1503628] [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: 09/29/2024] [Accepted: 12/26/2024] [Indexed: 01/29/2025] Open
Abstract
Introduction The pathogenesis of renal fibrosis is related to blood stasis, and the method of promoting blood circulation and removing blood stasis is often used as the treatment principle. Danshen injection (DSI) is a commonly used drug for promoting blood circulation and removing blood stasis in clinic. However, whether DSI slows the progression of renal fibrosis or the potential mechanism is uncertain. Methods We investigated renal fibrosis models using UUO mice and TGF-β stimulation in HK-2 cells. Results Our findings revealed that DSI or Fer-1 alleviated kidney injury by ameliorating renal morphology injury and pathological injury in vivo. Besides, DSI or Fer-1 inhibited renal fibrosis in vivo and in TGF-β-induced HK-2 cells. Furthermore, ferroptosis was lessened under DSI or Fer-1 treatment. More importantly, the DSI active ingredients (danshensu, salvianolic acid B, protocatechuic aldehyde, caffeic acid and tanshinone IIA) could bind to SIRT1. The protein levels of SIRT1 and GPX4 were downregulated accompanied by the incremental concentrations of TGF-β or Erastin, which were repaired by DSI or Fer-1 intervention. However, the inhibition of ferroptosis and renal fibrosis owing to DSI were reversed by SIRT1 inhibitor EX527. Conclusion Taken together, our results indicated that DSI could protect against ferroptosis to attenuate renal fibrosis by activating the SIRT1/GPX4 pathway. It is expected to be a potential agent to treat renal fibrosis.
Collapse
Affiliation(s)
- Yiwen Cao
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Huan Zhao
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shuyin Lin
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Junqi Chen
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Jingli Xiong
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhijun Zeng
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Ziyu Long
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yingru Su
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yingqi Zhong
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lingru Zhao
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Mingshan Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Junbiao Wu
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yuan Zhou
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jiuyao Zhou
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
13
|
Xu Y, Wang YR, Peng WP, Bu HM, Zhou Y, Wu Q. Tanshinone IIA Alleviates Pulmonary Fibrosis by Inhibiting Pyroptosis of Alveolar Epithelial Cells Through the MAPK Signaling Pathway. Phytother Res 2025; 39:282-297. [PMID: 39520221 DOI: 10.1002/ptr.8372] [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: 05/27/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 11/16/2024]
Abstract
The current dearth of safe and efficacious pharmaceutical interventions for pulmonary fibrosis (PF) has prompted investigations into alternative treatments. This study aim to investigate the underlying mechanisms of Tanshinone IIA in the treatment of PF. PF was induced in a mouse model by intratracheal infusion of bleomycin (BLM), followed by gavage administration of varying concentrations of Tanshinone IIA. Lung tissue was obtained for pathological slides, proteomic and transcriptomic analyses. The target was predicted and analyzed using network pharmacology. Initially, an in vitro model in A549 cells was established by adding BLM, followed by treatment with varying concentrations of Tanshinone IIA. Subsequently, NAC and the ERK inhibitor, U0126, were individually introduced. Treatment with Tanshinone IIA in vivo decreased lung tissue lesions. Proteomic, transcriptomic, and network pharmacology analyses suggested that Tanshinone IIA may offer therapeutic benefits for PF by mitigating oxidative stress damage via the MAPK signaling pathway. In vitro studies demonstrated that BLM treatment in A549 cells induced exposure of the N-terminal end of the pyroptosis core protein GSDMD, and elevated oxidative stress levels in A549 cells, concomitant with the upregulation of P-ERK protein expression. Subsequent administration of Tanshinone IIA, NAC, and U0126 reduced the number of A549 cells undergoing pyroptosis, decreased oxidative stress levels, and decreased P-ERK protein expression. These findings suggested that Tanshinone IIA potentially delays the progression of PF. The mechanism of action involves the inhibition of oxidative stress and reduced epithelial cell pyroptosis via the MAPK-related pathway. The findings may provide a new reference for treatment of PF.
Collapse
Affiliation(s)
- Yong Xu
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yi-Ran Wang
- Department of Physiology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Wen-Pan Peng
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Hui-Min Bu
- Department of Physiology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Yao Zhou
- Department of Pathophysiology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Qi Wu
- Department of Physiology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
14
|
Sang Y, Du J, Zulikala D, Sang Z. Mechanistic analysis of Tanshinone IIA's regulation of the ATM/GADD45/ORC signaling pathway to reduce myocardial ischemia-reperfusion injury. Front Pharmacol 2024; 15:1510380. [PMID: 39776578 PMCID: PMC11703710 DOI: 10.3389/fphar.2024.1510380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Background By far, one of the best treatments for myocardial ischemia is reperfusion therapy. The primary liposoluble component of Danshen, a traditional Chinese herbal remedy, Tanshinone ⅡA, has been shown to have cardiac healing properties. The purpose of this work is to investigate the processes by which Tanshinone ⅡA influences myocardial ischemia-reperfusion injury (MIRI) in the H9C2 cardiac myoblast cell line, as well as the association between Tanshinone ⅡA and MIRI. Methods and results The cardiac cells were divided into a normal group, a model group and Tanshinone ⅡA treatment groups. After 4 h of culture with the deprivation of oxygen and glucose, the cells were incubated normally for 2 h. The success of the model and the capacity of Tanshinone ⅡA to heal cardiac damage were validated by the outcomes of cell viability, morphology, and proliferation. The efficacy of Tanshinone ⅡA in treating MIRI was further confirmed by the scratch assay and biomarker measurement. The differentially expressed genes were examined using transcriptome sequencing. The Ataxia-Telangiectasia Mutated (ATM)/Growth Arrest and DNA Damage (GADD45)/Origin Recognition Complex (ORC) signaling pathway was identified as being crucial to this process by KEGG pathway analysis and GO enrichment. Molecular docking and RT-qPCR were used to confirm our results. The crucial function of the ATM/GADD45/ORC pathway was further confirmed by the addition of an ATM inhibitor, which inhibited the expression of ATM. Conclusion Tanshinone ⅡA can relieve the myocardial ischemia-reperfusion injury in cardiac cells by activating the ATM/GADD45/ORC pathway.
Collapse
Affiliation(s)
- Yiwei Sang
- Nature Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast, United Kingdom
| | - Jiangnan Du
- Nature Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast, United Kingdom
| | - Dilimulati Zulikala
- Nature Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast, United Kingdom
| | - Zhongqiang Sang
- Dermatology Department, Shanghai Zhongye Hospital, Shanghai, China
| |
Collapse
|
15
|
Yuan Y, Huang H, Hu T, Zou C, Qiao Y, Fang M, Liu J, Lai S. Curcumin pretreatment attenuates myocardial ischemia/reperfusion injury by inhibiting ferroptosis, autophagy and apoptosis via HES1. Int J Mol Med 2024; 54:110. [PMID: 39364745 PMCID: PMC11517743 DOI: 10.3892/ijmm.2024.5434] [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: 05/01/2024] [Accepted: 09/18/2024] [Indexed: 10/05/2024] Open
Abstract
The early restoration of hemodynamics/reperfusion in acute myocardial infarction (AMI) is an effective therapeutic strategy to reduce sudden death and improve patient prognosis. However, reperfusion induces additional cardiomyocyte damage and cardiac tissue dysfunction. In this context, turmeric‑derived curcumin (Cur) has been shown to exhibit a protective effect against myocardial ischemia/reperfusion injury (I/RI). The molecular mechanism of its activity, however, remains unclear. The current study investigated the protective effect of Cur and its molecular mechanism via in vitro experiments. The Cell Counting Kit‑8 and lactate dehydrogenase (LDH) assay kit were used to assess the cell viability and cytotoxicity. The contents of malondialdehyde (MDA), superoxide dismutase (SOD), glutathione peroxidase, glutathione (GSH)/glutathione disulfide (GSSG), total iron, ferrous iron, caspase‑3 and reactive oxygen species (ROS) were measured using an appropriate kit. Western blotting was used to detect the expression of relevant proteins. The levels of apoptosis, mitochondrial permeability transition pore (MPTP) opening, and mitochondrial membrane potential (MMP) were detected by flow cytometry. The study findings indicated that anoxia/reoxygenation (A/R) injury significantly decreased cell viability, increased in LDH and caspase‑3 activities, induced ferroptosis, increased apoptosis and overactivated autophagy. However, pretreatment with Cur or ferrostatin‑1 (Fer‑1, a ferroptosis inhibitor) significantly increased A/R‑reduced cell viability, SOD, glutathione peroxidase activity, GSH/GSSH ratio and HES1 and glutathione peroxidase 4 protein expression; attenuated A/R‑induced LDH, MDA, total iron, ferrous iron, prostaglandin‑endoperoxide synthase 2 protein expression and prevented ROS overproduction and MMP loss. In addition, Cur inhibited caspase‑3 activity, upregulated the Bcl‑2/Bax ratio, reduced apoptotic cell number and inhibited MPTP over‑opening. Furthermore, Cur increased P62, LC3II/I, NDUFB8 and UQCRC2 expression and upregulated the p‑AMPK/AMPK ratio. However, erastin (a ferroptosis activator), pAD/HES1‑short hairpin RNA, rapamycin (an autophagy activator) and Compound C (an AMPK inhibitor) blocked the protective effect of Cur. In conclusion, Cur pretreatment inhibited ferroptosis, autophagy overactivation and oxidative stress; improved mitochondrial dysfunction; maintained energy homeostasis; attenuated apoptosis; and ultimately protected the myocardium from A/R injury via increased HES1 expression.
Collapse
Affiliation(s)
- Yong Yuan
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Huang Huang
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Tie Hu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Chenchao Zou
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yamei Qiao
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ming Fang
- Department of Emergency, Gaoxin Branch of The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jichun Liu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Songqing Lai
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
16
|
Han D, Chen YB, Zhao K, Li HZ, Chen XY, Zhu GZ, Tu C, Gao JW, Zhuang JS, Wu ZY, Zhong ZM. Tanshinone IIA alleviates inflammation-induced skeletal muscle atrophy by regulating mitochondrial dysfunction. Arch Biochem Biophys 2024; 762:110215. [PMID: 39547552 DOI: 10.1016/j.abb.2024.110215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 11/17/2024]
Abstract
Skeletal muscle atrophy, characterized by loss of muscle mass and function, is often linked to systemic inflammation. Tanshinone IIA (Tan IIA), a major active constituent of Salvia miltiorrhiza, has anti-inflammatory and antioxidant properties. However, the effect of Tan IIA on inflammation-induced skeletal muscle atrophy remains unclear. Here, a mice model of the inflammatory muscle atrophy was established using lipopolysaccharide (LPS). Tan IIA intervention significantly increased muscle mass and strength, improved muscle fiber size, and maintained the integrity of skeletal muscle mitochondrial morphology in LPS-treated mice. Myotubes derived from myosatellite cells (MUSCs) were exposed to LPS in vitro. Tan IIA treatment inhibited LPS-induced muscle protein degradation and increased myotube diameter. Notably, Tan IIA attenuated LPS-induced inflammatory response and hyperactive mitophagy both in vivo and in vitro. In addition, Tan IIA treatment effectively diminished oxidative stress, inhibited the accumulation of mitochondrial reactive oxygen species (mtROS), and attenuated mitochondrial fission in LPS-treated myotubes. Reducing mtROS production helped to inhibit LPS-induced excessive mitophagy and myotubes atrophy. Together, our results reveal that Tan IIA can protect against inflammation-induced skeletal muscle atrophy by regulating mitochondrial dysfunction, presenting innovative potential therapeutics for skeletal muscle atrophy.
Collapse
Affiliation(s)
- Dong Han
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yun-Biao Chen
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kai Zhao
- Department of Orthopaedics, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Hong-Zhou Li
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xing-Yu Chen
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guo-Zheng Zhu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chen Tu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jia-Wen Gao
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jing-Shen Zhuang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhi-Yong Wu
- Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhao-Ming Zhong
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
17
|
Tian L, Liu Q, Guo H, Zang H, Li Y. Fighting ischemia-reperfusion injury: Focusing on mitochondria-derived ferroptosis. Mitochondrion 2024; 79:101974. [PMID: 39461581 DOI: 10.1016/j.mito.2024.101974] [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: 07/04/2024] [Revised: 09/12/2024] [Accepted: 10/12/2024] [Indexed: 10/29/2024]
Abstract
Ischemia-reperfusion injury (IRI) is a major cause of mortality and morbidity. Current treatments for IRI have limited efficacy and novel therapeutic strategies are needed. Mitochondrial dysfunction not only initiates IRI but also plays a significant role in ferroptosis pathogenesis. Recent studies have highlighted that targeting mitochondrial pathways is a promising therapeutic approach for ferroptosis-induced IRI. The association between ferroptosis and IRI has been reviewed many times, but our review provides the first comprehensive overview with a focus on recent mitochondrial research. First, we present the role of mitochondria in ferroptosis. Then, we summarize the evidence on mitochondrial manipulation of ferroptosis in IRI and review recent therapeutic strategies aimed at targeting mitochondria-related ferroptosis to mitigate IRI. We hope our review will provide new ideas for the treatment of IRI and accelerate the transition from bench to bedside.
Collapse
Affiliation(s)
- Lei Tian
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Qian Liu
- Department of Anesthesiology, Zigong First People's Hospital, Zigong Academy of Medical Sciences, Zigong, China
| | - Hong Guo
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Honggang Zang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Yulan Li
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
18
|
Zhu X, Qin Z, Zhou M, Li C, Jing J, Ye W, Gan X. The Role of Mitochondrial Permeability Transition in Bone Metabolism, Bone Healing, and Bone Diseases. Biomolecules 2024; 14:1318. [PMID: 39456250 PMCID: PMC11506728 DOI: 10.3390/biom14101318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/11/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Bone is a dynamic organ with an active metabolism and high sensitivity to mitochondrial dysfunction. The mitochondrial permeability transition pore (mPTP) is a low-selectivity channel situated in the inner mitochondrial membrane (IMM), permitting the exchange of molecules of up to 1.5 kDa in and out of the IMM. Recent studies have highlighted the critical role of the mPTP in bone tissue, but there is currently a lack of reviews concerning this topic. This review discusses the structure and function of the mPTP and its impact on bone-related cells and bone-related pathological states. The mPTP activity is reduced during the osteogenic differentiation of mesenchymal stem cells (MSCs), while its desensitisation may underlie the mechanism of enhanced resistance to apoptosis in neoplastic osteoblastic cells. mPTP over-opening triggers mitochondrial swelling, regulated cell death, and inflammatory response. In particular, mPTP over-opening is involved in dexamethasone-induced osteoblast dysfunction and bisphosphonate-induced osteoclast apoptosis. In vivo, the mPTP plays a significant role in maintaining bone homeostasis, with many bone disorders linked to its excessive opening. Genetic deletion or pharmacological inhibition of the over-opening of mPTP has shown potential in enhancing bone injury recovery and alleviating bone diseases. Here, we review the findings on the relationship of the mPTP and bone at both the cellular and disease levels, highlighting novel avenues for pharmacological approaches targeting mitochondrial function to promote bone healing and manage bone-related disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xueqi Gan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (X.Z.)
| |
Collapse
|
19
|
Wang J, Chen L, Zheng Q, Chen S, Hou Z, Liu P. Indirubin induces apoptosis in ovarian cancer cells via the mitochondrial pathway. Am J Transl Res 2024; 16:6119-6129. [PMID: 39544767 PMCID: PMC11558396 DOI: 10.62347/iofy5604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/25/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVE To investigate the pro-apoptotic effects of Indirubin, a traditional Chinese medicine, on ovarian cancer SKOV3 cell line and to explore its underlying mechanisms. METHODS Ovarian cancer SKOV3 cells were divided into a control group (cells cultured normally) and an experimental group (cells cultured in medium containing Indirubin). SKOV3 cells at the logarithmic phase were treated with Indirubin at various concentrations. Cell proliferation was assessed using the Cell Counting Kit-8 (CCK-8) assay and 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide (MTT) assay, while apoptosis was detected by flow cytometry, TdT-mediated dUTP Nick-End Labeling (TUNEL) staining, and immunofluorescence. Transcriptome sequencing was conducted to screen for apoptosis-related factors. qPCR and western blot were used to detect the mRNA and protein expression on added of molecules related to mitochondrial permeability transition pores. RESULTS MTT assay showed that Indirubin inhibited the growth of SKOV3 cells in both plate and sphere cultures, with IC50 values of 3.003 μM (plate culture) and 4.253 μM (sphere culture), respectively. Indirubin showed a lower inhibitory concentration than cisplatin (IC50 3.687 μM in plate culture and 7.023 μM in sphere culture), and its effect was comparable to adriamycin. Flow cytometry revealed an increase in apoptosis rates in SKOV3 cells treated with Indirubin. Transcriptome sequencing indicated significant changes in the transcription of various apoptosis-related genes, particularly those involved in the mitochondrial apoptosis pathway, such as Bcl-2-associated X protein (Bax) and Bcl2 associated agonist of cell death (Bad). After Indirubin treatment, the mRNA and protein expression levels of mitochondrial channel-related genes Cyclophilin D (CyPD), adenine nucleotide translocator 1 (ANT1), and voltage-dependent anion channel (VADC) were significantly elevated (all P < 0.05). By regulating the mitochondrial membrane permeability through the Bcl-2 family members, Indirubin promoted apoptosis in SKOV3 cells. CONCLUSION Indirubin inhibits the proliferation and promotes the apoptosis of ovarian cancer cells, exerting an anti-tumor effect. Its pro-apoptotic action is closely related to the mitochondrial apoptosis pathway.
Collapse
Affiliation(s)
- Jinhua Wang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong UniversityJinan 250012, Shandong, China
- Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical UniversityFuzhou 350001, Fujian, China
| | - Lihong Chen
- Department of Obstetrics and Gynecology, Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical UniversityFuzhou 350001, Fujian, China
| | - Qiaomei Zheng
- Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical UniversityFuzhou 350001, Fujian, China
| | - Shaozhan Chen
- Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical UniversityFuzhou 350001, Fujian, China
| | - Zhidan Hou
- Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical UniversityFuzhou 350001, Fujian, China
| | - Peishu Liu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong UniversityJinan 250012, Shandong, China
| |
Collapse
|
20
|
Wen J, Li L, Yang Y, Ou D, Yang J, Xie J, Du W, Tong Y. Phytochemicals targeting ferroptosis in cardiovascular diseases: Recent advances and therapeutic perspectives. Phytother Res 2024; 38:4386-4405. [PMID: 38973263 DOI: 10.1002/ptr.8278] [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: 01/02/2024] [Revised: 06/03/2024] [Accepted: 06/08/2024] [Indexed: 07/09/2024]
Abstract
Ferroptosis is a form of iron-dependent regulatory cell death that is related to the pathogenesis and progression of various cardiovascular diseases, such as arrhythmia, diabetic cardiomyopathy, myocardial infarction, myocardial ischemia/reperfusion injury, and heart failure. This makes it a promising therapeutic target for cardiovascular diseases. It is interesting that a significant number of cardiovascular disease treatment drugs derived from phytochemicals have been shown to target ferroptosis, thus producing cardioprotective effects. This study offers a concise overview of the initiation and control mechanisms of ferroptosis. It discusses the core regulatory factors of ferroptosis as potential new therapeutic targets for various cardiovascular diseases, elucidating how ferroptosis influences the progression of cardiovascular diseases. In addition, this review systematically summarizes the regulatory effects of phytochemicals on ferroptosis, emphasizing their potential mechanisms and clinical applications in treating cardiovascular diseases. This study provides a reference for further elucidating the molecular mechanisms of phytochemicals in treating cardiovascular diseases. This may accelerate their application in the treatment of cardiovascular diseases and is worth further research in this field.
Collapse
Affiliation(s)
- Jianxia Wen
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Lu Li
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Yi Yang
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Dinglin Ou
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Junjie Yang
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Jiachen Xie
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Wenya Du
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Yuling Tong
- School of Medicine and Food, Sichuan Vocational College of Health and Rehabilitation, Zigong, China
| |
Collapse
|
21
|
Cong J, Li JY, Zou W. Mechanism and treatment of intracerebral hemorrhage focus on mitochondrial permeability transition pore. Front Mol Neurosci 2024; 17:1423132. [PMID: 39156127 PMCID: PMC11328408 DOI: 10.3389/fnmol.2024.1423132] [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: 04/25/2024] [Accepted: 07/15/2024] [Indexed: 08/20/2024] Open
Abstract
Intracerebral hemorrhage (ICH) is the second most common subtype of stroke, characterized by high mortality and a poor prognosis. Despite various treatment methods, there has been limited improvement in the prognosis of ICH over the past decades. Therefore, it is imperative to identify a feasible treatment strategy for ICH. Mitochondria are organelles present in most eukaryotic cells and serve as the primary sites for aerobic respiration and energy production. Under unfavorable cellular conditions, mitochondria can induce changes in permeability through the opening of the mitochondrial permeability transition pore (mPTP), ultimately leading to mitochondrial dysfunction and contributing to various diseases. Recent studies have demonstrated that mPTP plays a role in the pathological processes associated with several neurodegenerative diseases including Parkinson's disease, Alzheimer's disease, Huntington's disease, ischemic stroke and ischemia-reperfusion injury, among others. However, there is limited research on mPTP involvement specifically in ICH. Therefore, this study comprehensively examines the pathological processes associated with mPTP in terms of oxidative stress, apoptosis, necrosis, autophagy, ferroptosis, and other related mechanisms to elucidate the potential mechanism underlying mPTP involvement in ICH. This research aims to provide novel insights for the treatment of secondary injury after ICH.
Collapse
Affiliation(s)
- Jing Cong
- The First School of Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jing-Yi Li
- The Second School of Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wei Zou
- Molecular Biology Laboratory of Clinical Integrated of Traditional Chinese and Western Medicine of Heilong Jiang Province, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
22
|
Hu T, Yu WP, Wang XQ, Wang ZY, Xu ZQ, Hu FJ, Liu JC, Yu F, Wang LJ. Activation of PPAR-α attenuates myocardial ischemia/reperfusion injury by inhibiting ferroptosis and mitochondrial injury via upregulating 14-3-3η. Sci Rep 2024; 14:15246. [PMID: 38956068 PMCID: PMC11219969 DOI: 10.1038/s41598-024-64638-9] [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: 03/01/2024] [Accepted: 06/11/2024] [Indexed: 07/04/2024] Open
Abstract
This study aimed to explore the effects of peroxisome proliferator-activated receptor α (PPAR-α), a known inhibitor of ferroptosis, in Myocardial ischemia/reperfusion injury (MIRI) and its related mechanisms. In vivo and in vitro MIRI models were established. Our results showed that activation of PPAR-α decreased the size of the myocardial infarct, maintained cardiac function, and decreased the serum contents of creatine kinase-MB (CK-MB), lactate dehydrogenase (LDH), and Fe2+ in ischemia/reperfusion (I/R)-treated mice. Additionally, the results of H&E staining, DHE staining, TUNEL staining, and transmission electron microscopy demonstrated that activation of PPAR-α inhibited MIRI-induced heart tissue and mitochondrial damage. It was also found that activation of PPAR-α attenuated MIRI-induced ferroptosis as shown by a reduction in malondialdehyde, total iron, and reactive oxygen species (ROS). In vitro experiments showed that intracellular contents of malondialdehyde, total iron, LDH, reactive oxygen species (ROS), lipid ROS, oxidized glutathione disulphide (GSSG), and Fe2+ were reduced by the activation of PPAR-α in H9c2 cells treated with anoxia/reoxygenation (A/R), while the cell viability and GSH were increased after PPAR-α activation. Additionally, changes in protein levels of the ferroptosis marker further confirmed the beneficial effects of PPAR-α activation on MIRI-induced ferroptosis. Moreover, the results of immunofluorescence and dual-luciferase reporter assay revealed that PPAR-α achieved its activity via binding to the 14-3-3η promoter, promoting its expression level. Moreover, the cardioprotective effects of PPAR-α could be canceled by pAd/14-3-3η-shRNA or Compound C11 (14-3-3η inhibitor). In conclusion, our results indicated that ferroptosis plays a key role in aggravating MIRI, and PPAR-α/14-3-3η pathway-mediated ferroptosis and mitochondrial injury might be an effective therapeutic target against MIRI.
Collapse
Affiliation(s)
- Tie Hu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Wen-Peng Yu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xiu-Qi Wang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Zi-Yao Wang
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Zhi-Qiang Xu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Fa-Jia Hu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Ji-Chun Liu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Fan Yu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| | - Li-Jun Wang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
23
|
Zhao ST, Qiu ZC, Zeng RY, Zou HX, Qiu RB, Peng HZ, Zhou LF, Xu ZQ, Lai SQ, Wan L. Exploring the molecular biology of ischemic cardiomyopathy based on ferroptosis‑related genes. Exp Ther Med 2024; 27:221. [PMID: 38590563 PMCID: PMC11000445 DOI: 10.3892/etm.2024.12509] [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: 10/05/2023] [Accepted: 02/21/2024] [Indexed: 04/10/2024] Open
Abstract
Ischemic cardiomyopathy (ICM) is a serious cardiac disease with a very high mortality rate worldwide, which causes myocardial ischemia and hypoxia as the main damage. Further understanding of the underlying pathological processes of cardiomyocyte injury is key to the development of cardioprotective strategies. Ferroptosis is an iron-dependent form of regulated cell death characterized by the accumulation of lipid hydroperoxides to lethal levels, resulting in oxidative damage to the cell membrane. The current understanding of the role and regulation of ferroptosis in ICM is still limited, especially in the absence of evidence from large-scale transcriptomic data. Through comprehensive bioinformatics analysis of human ICM transcriptome data obtained from the Gene Expression Omnibus database, the present study identified differentially expressed ferroptosis-related genes (DEFRGs) in ICM. Subsequently, their potential biological mechanisms and cross-talk were analyzed, and hub genes were identified by constructing protein-protein interaction networks. Ferroptosis features such as reactive oxygen species generation, changes in ferroptosis marker proteins, iron ion aggregation and lipid oxidation, were identified in the H9c2 anoxic reoxygenation injury model. Finally, the diagnostic ability of Gap junction alpha-1 (GJA1), Solute carrier family 40 member 1 (SLC40A1), Alpha-synuclein (SNCA) were identified through receiver operating characteristic curves and the expression of DEFRGs was verified in an in vitro model. Furthermore, potential drugs (retinoic acid) that could regulate ICM ferroptosis were predicted based on key DEFRGs. The present article presents new insights into the role of ferroptosis in ICM, investigating the regulatory role of ferroptosis in the pathological process of ICM and advocating for ferroptosis as a potential novel therapeutic target for ICM based on evidence from the ICM transcriptome.
Collapse
Affiliation(s)
- Shi-Tao Zhao
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Cardiovascular Surgical Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhi-Cong Qiu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Cardiovascular Surgical Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Rui-Yuan Zeng
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Cardiovascular Surgical Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Hua-Xi Zou
- Department of Cardiovascular Surgery, The Second Affiliated Hospita, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330200, P.R. China
| | - Rong-Bin Qiu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Cardiovascular Surgical Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Han-Zhi Peng
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Cardiovascular Surgical Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Lian-Fen Zhou
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Cardiovascular Surgical Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhi-Qiang Xu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Cardiovascular Surgical Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Song-Qing Lai
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Cardiovascular Surgical Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Li Wan
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Cardiovascular Surgical Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
24
|
Hu F, Hu T, Qiao Y, Huang H, Zhang Z, Huang W, Liu J, Lai S. Berberine inhibits excessive autophagy and protects myocardium against ischemia/reperfusion injury via the RhoE/AMPK pathway. Int J Mol Med 2024; 53:49. [PMID: 38577949 PMCID: PMC10999226 DOI: 10.3892/ijmm.2024.5373] [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: 03/20/2024] [Indexed: 04/06/2024] Open
Abstract
Several studies have shown that berberine (BBR) is effective in protecting against myocardial ischemia‑reperfusion injury (MI/RI). However, the precise molecular mechanism remains elusive. The present study observed the mechanism and the safeguarding effect of BBR against hypoxia/reoxygenation (H/R) myocardial injury in H9c2 cells. BBR pretreatment significantly improved the decrease of cell viability, P62 protein, Rho Family GTPase 3 (RhoE) protein, ubiquinone subunit B8 protein, ubiquinol‑cytochrome c reductase core protein U, the Bcl‑2‑associated X protein/B‑cell lymphoma 2 ratio, glutathione (GSH) and the GSH/glutathione disulphide (GSSG) ratio induced by H/R, while reducing the increase in lactate dehydrogenase, microtubule‑associated protein 1 light 3 protein, caspase‑3 activity, reactive oxygen species, GSSG and malonaldehyde caused by H/R. Transmission electron microscopy and LysoTracker Red DND‑99 staining results showed that BBR pretreatment inhibited H/R‑induced excessive autophagy by mediating RhoE. BBR also inhibited mitochondrial permeability transition, maintained the stability of the mitochondrial membrane potential, reduced the apoptotic rate, and increased the level of caspase‑3. However, the protective effects of BBR were attenuated by pAD/RhoE‑small hairpin RNA, rapamycin (an autophagy activator) and compound C (an AMP‑activated protein kinase inhibitor). These new findings suggested that BBR protects the myocardium from MI/RI by inhibiting excessive autophagy, maintaining mitochondrial function, improving the energy supply and redox homeostasis, and attenuating apoptosis through the RhoE/AMP‑activated protein kinase pathway.
Collapse
Affiliation(s)
- Fajia Hu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Tie Hu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yamei Qiao
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Huang Huang
- Institute of Cardiovascular Surgical Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zeyu Zhang
- Institute of Nanchang University Trauma Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Wenxiong Huang
- Institute of Cardiovascular Surgical Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jichun Liu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Songqing Lai
- Institute of Cardiovascular Surgical Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|