1
|
Ji YW, Wen XY, Tang HP, Su WT, Xia ZY, Lei SQ. Necroptosis: a significant and promising target for intervention of cardiovascular disease. Biochem Pharmacol 2025; 237:116951. [PMID: 40268251 DOI: 10.1016/j.bcp.2025.116951] [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: 12/20/2024] [Revised: 03/18/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025]
Abstract
Due to changes in dietary structures, population aging, and the exacerbation of metabolic risk factors, the incidence of cardiovascular disease continues to rise annually, posing a significant health burden worldwide. Cell death plays a crucial role in the onset and progression of cardiovascular diseases. As a regulated endpoint encountered by cells under adverse stress conditions, the execution of necroptosis is regulated by classicalpathways, the calmodulin-dependent protein kinases (CaMK) pathway, and mitochondria-dependent pathways, and implicated in various cardiovascular diseases, including atherosclerosis, myocardial infarction, myocardial ischemia-reperfusion injury (IRI), heart failure, diabetic cardiomyopathy, dilated cardiomyopathy, hypertrophic cardiomyopathy, chemotherapy drug-induced cardiomyopathy, and abdominal aortic aneurysm (AAA). To further investigate potential therapeutic targets for cardiovascular diseases, we also analyzed the main molecules and their inhibitors involved in necroptosis in an effort to uncover insights for treatment.
Collapse
Affiliation(s)
- Yan-Wei Ji
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin-Yu Wen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - He-Peng Tang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wa-Ting Su
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhong-Yuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shao-Qing Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
2
|
Chaudhary R, Suhan TK, Wu C, Alzamrooni A, Madamanchi N, Abdel-Latif A. Housing temperature influences metabolic phenotype of heart failure with preserved ejection fraction in J vs N strain C57BL/6 mice. Mol Cell Endocrinol 2025; 598:112457. [PMID: 39788312 PMCID: PMC11820722 DOI: 10.1016/j.mce.2025.112457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/04/2025] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
Preclinical heart failure studies rely heavily on mouse models despite their higher metabolic and heart rates compared to humans. This study examines how mouse strain (C57BL/6J vs. C57BL/6N) and housing temperature (23 °C vs. 30 °C) affect a well-established two-hit HFpEF model using high-fat diet with L-NAME treatment in male C57BL/6 mouse. Metabolic parameters and cardiac function were assessed at baseline, week 5, and week 15. Thermoneutral housing (30 °C) reduced early diastolic dysfunction in the J strain and altered metabolic profiles in both strains, decreasing energy expenditure and fat oxidation. The J strain specifically showed reduced respiratory exchange ratio and glucose oxidation at 30 °C. While physical activity remained constant across groups, both strains exhibited increased cardiac fibrosis and inflammatory gene expression under HFD + L-NAME, independent of housing temperature. These findings reveal strain-specific physiological adaptations to housing temperature, emphasizing the need to consider environmental conditions in heart failure research carefully.
Collapse
Affiliation(s)
- Rajesh Chaudhary
- Division of Cardiology, Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48105, USA; Ann Arbor VA Healthcare System, 2215 Fuller Rd, Ann Arbor, MI, 48105, USA
| | - Tahra K Suhan
- Division of Cardiology, Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48105, USA; Ann Arbor VA Healthcare System, 2215 Fuller Rd, Ann Arbor, MI, 48105, USA
| | - Chao Wu
- University of Michigan, Ann Arbor, MI, 48105, USA
| | - Afnan Alzamrooni
- Division of Cardiology, Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48105, USA; Ann Arbor VA Healthcare System, 2215 Fuller Rd, Ann Arbor, MI, 48105, USA
| | - Nageswara Madamanchi
- Division of Cardiology, Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Ahmed Abdel-Latif
- Division of Cardiology, Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48105, USA; Ann Arbor VA Healthcare System, 2215 Fuller Rd, Ann Arbor, MI, 48105, USA.
| |
Collapse
|
3
|
Jia Y, Liu Y, Zuo Y, Zhang J, Li Y, Liu X, Lv S. The Potential Therapeutic Prospect of PANoptosis in Heart Failure. J Inflamm Res 2024; 17:9147-9168. [PMID: 39583864 PMCID: PMC11585275 DOI: 10.2147/jir.s485901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 11/15/2024] [Indexed: 11/26/2024] Open
Abstract
Heart failure (HF) represents a serious manifestation or advanced stage of various cardiac diseases. HF continues to impose a significant global disease burden, characterized by high rates of hospitalization and fatality. Furthermore, the pathogenesis and pathophysiological processes underlying HF remain incompletely understood, complicating its prevention and treatment strategies. One significant pathophysiological mechanism associated with HF is the systemic inflammatory response. PANoptosis, a novel mode of inflammatory cell death, has been extensively studied in the context of infectious diseases, neurodegenerative disorders, cancers, and other inflammatory conditions. Recent investigations have revealed that PANoptosis-related genes are markedly dysregulated in HF specimens. Consequently, the PANoptosis-mediated inflammatory response may represent a potential mechanism and therapeutic target for HF. This paper conducts a comprehensive analysis of the molecular pathways that drive PANoptosis. We discuss its role and potential therapeutic targets in HF, thereby providing valuable insights for clinical treatment and the development of novel therapies.
Collapse
Affiliation(s)
- Yunfeng Jia
- Department of Geriatrics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, 300381, People’s Republic of China
| | - Yayi Liu
- Department of Geriatrics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, 300381, People’s Republic of China
| | - Yiming Zuo
- Department of Geriatrics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, 300381, People’s Republic of China
| | - Junping Zhang
- Department of Geriatrics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, 300381, People’s Republic of China
| | - Yanyang Li
- Department of Integrated Traditional and Western Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People’s Republic of China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People’s Republic of China
| | - Xuezheng Liu
- Department of Geriatrics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, 300381, People’s Republic of China
| | - Shichao Lv
- Department of Geriatrics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, 300381, People’s Republic of China
| |
Collapse
|
4
|
Scott XO, Kerr NA, Sanchez-Molano J, de Rivero Vaccari JP, Hadad R, De La Cruz A, Larsson HP, Dietrich WD, Keane RW. Catecholamine-Induced Inflammasome Activation in the Heart Following Photothrombotic Stroke. Transl Stroke Res 2024:10.1007/s12975-024-01311-3. [PMID: 39556309 DOI: 10.1007/s12975-024-01311-3] [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: 07/31/2024] [Revised: 10/23/2024] [Accepted: 11/11/2024] [Indexed: 11/19/2024]
Abstract
Cerebrovascular stroke patients exhibit an increased incidence of cardiac arrhythmias. The pathomechanisms underlying post-traumatic cardiac dysfunction include a surge of catecholamines and an increased systemic inflammatory response, but whether inflammasome activation contributes to cardiac dysfunction remains unexplored. Here, we used a mouse model of photothrombotic stroke (PTS) to investigate the role of inflammasome activation in post-stroke cardiac dysfunction by catecholamines and to evaluate the effectiveness of the inflammasome inhibitor IC100 on inflammasome activation. To evaluate functional electrophysiological changes in the heart by catecholamine treatment, we recorded action potential duration in excised zebrafish hearts with and without IC100 treatment. We show that PTS induced AIM2 inflammasome activation in atria and ventricles that was significantly reduced by administration of IC100. Injection of epinephrine into naïve mice induced a significant increase in AIM2, IL-1b and caspase-8 in atria. Treatment of excised zebrafish hearts with epinephrine shortened the action potential duration and this shortening that was reduced by IC100. These findings indicate that stroke initiates a catecholamine surge that induces inflammasome activation and pyroptosis in the heart that is blocked by IC100, thus providing a framework for the development of therapeutics for stroke-related cardiovascular injury.
Collapse
Affiliation(s)
- Xavier O Scott
- Department of Molecular Physiology and Cellular Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Nadine A Kerr
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, 33136, USA
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juliana Sanchez-Molano
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, 33136, USA
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Molecular Physiology and Cellular Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, 33136, USA
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Roey Hadad
- Department of Molecular Physiology and Cellular Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, 33136, USA
| | - Alicia De La Cruz
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - H Peter Larsson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - W Dalton Dietrich
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, 33136, USA
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Robert W Keane
- Department of Molecular Physiology and Cellular Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA.
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, 33136, USA.
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
5
|
Emeka PM, Badger-Emeka LI, Thirugnanasambantham K, Alatawi AS. Rutin-Activated Nuclear Factor Erythroid 2-Related Factor 2 (Nrf2) Attenuates Corneal and Heart Damage in Mice. Pharmaceuticals (Basel) 2024; 17:1523. [PMID: 39598433 PMCID: PMC11597448 DOI: 10.3390/ph17111523] [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: 09/30/2024] [Revised: 11/06/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Corneal degeneration is a form of progressive cell death caused by multiple factors, such as diabetic retinopathy. It is the most well-known neural degenerative disease caused by macular degeneration in the aged and those with retinitis pigmentosa. Myocardial infarction is becoming a more common burden, causing cardiomyocyte degeneration, ischemia, and heart tissue death. This study examined the preventive effects of rutin on isoproterenol (ISO)-induced oxidative damage (that is, inflammation) on rabbit corneal epithelial cells and mouse heart injuries. Methods: These investigations involved a cytotoxicity test, biochemical analysis, qRT-PCR, Western blotting, and mouse cardiac histopathology. Results: The results showed that rutin enhanced ADH7 and ALDH1A1, retinoic acid signaling components in SIRC1 rabbit corneal cell lines. The production of NO by ocular epithelial cells was significantly reduced. It reduced cTnT and cTnI, CK-MB, and LDH contents in mouse cardiac tissue. The nuclear expressions of Nrf2, Sirt, and HO-1 were all increased by rutin. Docking studies revealed a good interaction between rutin and the Keap protein, enhancing Nrf2 nuclear activity. Conclusions: This showed that rutin can potentially enhance ADH7 and ALDH1A1 corneal signaling components, preventing corneal degeneration and mitigating ISO-induced myocardial infarction (MI) via Keap/Nrf2 expressions.
Collapse
Affiliation(s)
- Promise M. Emeka
- Department of Pharmaceutical Science, College of Clinical Pharmacy, King Faisal University, Al Ahsa 31982, Saudi Arabia;
| | - Lorina I. Badger-Emeka
- Department of Biomedical Science, College of Medicine, King Faisal University, Al Ahsa 31982, Saudi Arabia;
| | | | - Abdulaziz S. Alatawi
- Department of Pharmaceutical Science, College of Clinical Pharmacy, King Faisal University, Al Ahsa 31982, Saudi Arabia;
| |
Collapse
|
6
|
Wu S, Li M, Chen F, Zeng Y, Xu C. Inhibition of β2-adrenergic receptor regulates necroptosis in prostate cancer cell. Heliyon 2024; 10:e31865. [PMID: 38845899 PMCID: PMC11153256 DOI: 10.1016/j.heliyon.2024.e31865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 06/09/2024] Open
Abstract
As the malignant tumor with the highest incidence in male, prostate cancer poses a significant threat to the reproductive health of elderly men. Our previous studies have shown that promoting necroptosis of cancer cells can effectively inhibit cancer cell proliferation. This study includes lentivirus-mediated knockdown of β2AR which resulted in stable transfectants that exhibited an increased ability to form clones compared to that of the negative control group. In the protein and mRNA levels, necroptosis associated RIP and mixed lineage kinase domain-like (MLKL) were significantly higher in the treatment group than they were in the control group. Furthermore, cells treated with propranolol exhibited necrotic morphology as observed by transmission electron microscopy. The combination of β2AR suppression and necroptosis inhibitors resulted in a more potent suppression of cell proliferation compared to that observed in the control and negative control groups. Additionally, it elevated in the necrosis rate as determined by flow cytometry. Immunofluorescence staining revealed enhanced RIP and MLKL expression in the sh-β2AR group compared to levels in the negative control group. Co-immunoprecipitation experiments detected an interaction between β2AR and RIP. MLKL and RIPK3 levels were significantly higher in xenograft tumor sections from the sh-β2AR group compared to levels in the sh-NC group. To conclude, our research indicates the proliferation of PC-3 and DU-145 cprostate cancer cells can be suppressed by inhibiting β2AR, and this occurs through the RIP/MLKL-mediated pathway of necroptosis.
Collapse
Affiliation(s)
| | | | - Fangfang Chen
- Institution of Life Science, Chongqing Medical University, Chongqing, China
| | - Yan Zeng
- Institution of Life Science, Chongqing Medical University, Chongqing, China
| | - Chen Xu
- Corresponding author. Institution of Life Science, Chongqing Medical University, 1 Medical College Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
7
|
Cai M, Wu P, Ni W, Huang D, Wang X. mTORC1 hyperactivation and resultant suppression of macroautophagy contribute to the induction of cardiomyocyte necroptosis by catecholamine surges. Physiol Rep 2024; 12:e15966. [PMID: 38444056 PMCID: PMC10915131 DOI: 10.14814/phy2.15966] [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: 12/21/2023] [Revised: 02/17/2024] [Accepted: 02/26/2024] [Indexed: 03/07/2024] Open
Abstract
Previous studies revealed a controversial role of mechanistic target of rapamycin complex 1 (mTORC1) and mTORC1-regulated macroautophagy in isoproterenol (ISO)-induced cardiac injury. Here we investigated the role of mTORC1 and potential underlying mechanisms in ISO-induced cardiomyocyte necrosis. Two consecutive daily injections of ISO (85 mg/kg, s.c.) or vehicle control (CTL) were administered to C57BL/6J mice with or without rapamycin (RAP, 5 mg/kg, i.p.) pretreatment. Western blot analyses showed that myocardial mTORC1 signaling and the RIPK1-RIPK3-MLKL necroptotic pathway were activated, mRNA expression analyses revealed downregulation of representative TFEB target genes, and Evan's blue dye uptake assays detected increased cardiomyocyte necrosis in ISO-treated mice. However, RAP pretreatment prevented or significantly attenuated the ISO-induced cardiomyocyte necrosis, myocardial inflammation, downregulation of TFEB target genes, and activation of the RIPK1-RIPK3-MLKL pathway. LC3-II flux assays confirmed the impairment of myocardial autophagic flux in the ISO-treated mice. In cultured neonatal rat cardiomyocytes, mTORC1 signaling was also activated by ISO, and inhibition of mTORC1 by RAP attenuated ISO-induced cytotoxicity. These findings suggest that mTORC1 hyperactivation and resultant suppression of macroautophagy play a major role in the induction of cardiomyocyte necroptosis by catecholamine surges, identifying mTORC1 inhibition as a potential strategy to treat heart diseases with catecholamine surges.
Collapse
Affiliation(s)
- Mingqi Cai
- Heart CenterShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- Division of Basic Biomedical SciencesSanford School of Medicine of the University of South DakotaVermillionSouth DakotaUSA
| | - Penglong Wu
- Division of Basic Biomedical SciencesSanford School of Medicine of the University of South DakotaVermillionSouth DakotaUSA
- Department of Cardiology, Xiamen Cardiovascular Hospital of Xiamen University, School of MedicineXiamen UniversityXiamenChina
| | - Wei Ni
- Department of Cardiology, Xiamen Cardiovascular Hospital of Xiamen University, School of MedicineXiamen UniversityXiamenChina
| | - Dong Huang
- Heart CenterShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xuejun Wang
- Division of Basic Biomedical SciencesSanford School of Medicine of the University of South DakotaVermillionSouth DakotaUSA
| |
Collapse
|
8
|
Peng Y, Qin D, Wang Y, Gao W, Xu X. Pharmacological inhibition of ICOS attenuates the protective effect of exercise on cardiac fibrosis induced by isoproterenol. Eur J Pharmacol 2024; 965:176327. [PMID: 38224847 DOI: 10.1016/j.ejphar.2024.176327] [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: 08/27/2023] [Revised: 12/14/2023] [Accepted: 01/09/2024] [Indexed: 01/17/2024]
Abstract
AIMS To investigate the cardioprotective mechanism of exercise or exercise combined with inducible costimulatory molecules (ICOS) monoclonal antibody (mAb) therapy against isoproterenol (ISO)-induced cardiac remodeling. MAIN METHODS Totally 24 male C57BL/6J mice were randomly divided into four groups: the control group (normal saline treatment), ISO group (subcutaneous injection of isoproterenol, 10 mg/kg/day, once daily for 5 consecutive days), the exercise with subcutaneous ISO injection group (EPI), and the exercise with injected with ISO and ICOS mAb group (EPII). The mice in EPI and EPII group were trained on a small animal treadmill for 4 weeks (13 m/min, 0% grade, 60min/day). KEY FINDINGS Exercise significantly attenuated CD45+, Mac-2 inflammatory cell infiltration, cardiac fibrosis and inhibited the RIPK1/RIPK3/MLKL/CaMKII and cardiomyocyte pyroptosis pathways to counter ISO-induced severe cardiac injury. The administration of the ICOS mAb may inhibit the cardioprotection of exercise against ISO-induced heart damage. Compared to those in EPI, our data showed that the increasing levels of myocardial fibrosis, the leukocyte infiltration of cardiac tissue and proteins expression of cardiac myocyte necrosis and pyroptosis signaling pathways in the EPII group. SIGNIFICANCE Our results demonstrated that exercise decreased leukocyte infiltration in heart, inhibited the cardiomyocyte pyroptosis and necroptosis signaling pathways, and attenuated inflammatory responses to alleviate ISO-induced cardiac fibrosis. However, the antifibrotic effects of combined treatment with exercise and ICOS mAb intervention did not exhibit synergistic enhancement.
Collapse
Affiliation(s)
- Yong Peng
- School of Kinesiology, Shanghai University of Sport, Shanghai, China; Jiangsu Collaborative Innovation Center for Sports and Health Project, Nanjing Sport Institute, Nanjing, Jiangsu, China; Key Laboratory of Exercise Training and Rehabilitation for Jiangsu Province, Nanjing Sport Institute, Nanjing, Jiangsu, China
| | - Di Qin
- School of Sport and Health, Nanjing Sport Institute, Nanjing, Jiangsu, China
| | - Yudi Wang
- School of Physical Education and Nursing, Chengdu College of Arts and Sciences, Chengdu, Sichuan, China
| | - Wenyue Gao
- School of Sport and Health, Nanjing Sport Institute, Nanjing, Jiangsu, China
| | - Xin Xu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China.
| |
Collapse
|
9
|
Chen Y, Guo X, Zeng Y, Mo X, Hong S, He H, Li J, Steinmetz R, Liu Q. Ferroptosis contributes to catecholamine-induced cardiotoxicity and pathological remodeling. Free Radic Biol Med 2023; 207:227-238. [PMID: 37499888 PMCID: PMC10529955 DOI: 10.1016/j.freeradbiomed.2023.07.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/06/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
High levels of circulating catecholamines cause cardiac injury, pathological remodeling, and heart failure, but the underlying mechanisms remain elusive. Here we provide both in vitro and in vivo evidence that excessive β-adrenergic stimulation induces ferroptosis in cardiomyocytes, revealing a novel mechanism for catecholamine-induced cardiotoxicity and remodeling. We found that isoproterenol, a synthetic catecholamine, promoted glutathione depletion and glutathione peroxidase 4 (GPX4) degradation in cardiomyocytes, leading to GPX4 inactivation and enhanced lipid peroxidation. Isoproterenol also promoted heme oxygenase 1 (HO-1) expression by downregulating the transcription suppressor BTB and CNC homology 1 (Bach1), leading to increased labile iron accumulation through heme degradation. Moreover, isoproterenol markedly induced the accumulation of free iron and lipid reactive oxygen species (ROS) in the mitochondria, while targeted inhibition of iron overload and ROS accumulation within mitochondria effectively inhibited ferroptosis in cardiomyocytes. Importantly, isoproterenol administration markedly induced ferroptosis in the myocardium in vivo, associated with elevated non-heme iron accumulation driven by HO-1 upregulation. Strikingly, blockade of ferroptosis with ferrostatin-1 or inhibition of HO-1 activity with zinc protoporphyrin (ZnPP) effectively alleviated cardiac necrosis, pathological remodeling, and heart failure induced by isoproterenol administration. Taken together, our results reveal that catecholamine stimulation primarily induces ferroptotic cell death in cardiomyocyte through GPX4 and Bach1-HO-1 dependent signaling pathways. Targeting ferroptosis may represent a novel therapeutic strategy for catecholamine overload-induced myocardial injury and heart failure.
Collapse
Affiliation(s)
- Yi Chen
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Xiaoyun Guo
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Yachang Zeng
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Xiaoliang Mo
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Siqi Hong
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Hui He
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Jing Li
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Rachel Steinmetz
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Qinghang Liu
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
10
|
Borodzicz-Jażdżyk S, Kołodzińska A, Czarzasta K, Wojciechowska M, Główczyńska R, Szczepankiewicz B, Puchalska L, Opolski G, Cudnoch-Jędrzejewska A. Inflammatory Forms of Cardiomyocyte Cell Death in the Rat Model of Isoprenaline-Induced Takotsubo Syndrome. Biomedicines 2023; 11:2060. [PMID: 37509699 PMCID: PMC10377582 DOI: 10.3390/biomedicines11072060] [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: 06/18/2023] [Revised: 07/15/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Takotsubo syndrome (TTS) is associated with inflammatory response, therefore the aim of the study was to evaluate the presence and dynamics of inflammatory-associated forms of cell death, necroptosis, and pyroptosis in the female rat model of isoprenaline (ISO)-induced TTS. TTS was induced in female Sprague Dawley rats (n = 36) by ISO 150 mg/kg intraperitoneally. Animals were divided into four groups: TTSO (TTS+ovariectomy; n = 10), TTSP (TTS+sham operation; n = 10), CO (0.9% NaCl+ovariectomy; n = 8), CP (0.9% NaCl+sham operation; n = 8). Histopathological analysis, evaluation of plasma concentration, and myocardial expression of pyroptosis- and necroptosis-associated proteins were performed. TTSO and TTSP groups had higher plasma concentrations of interleukin-1β in comparison with the controls. Low myocardial protein expression of mixed lineage kinase domain-like pseudokinase (MLKL), caspase-1 (Casp-1), and calcium/calmodulin-dependent kinase type II isoform delta (CAMKIIδ) was visible 6 and/or 12 h post-ISO. Twenty-four hours post-ISO, high myocardial and vascular protein expression of CAMKIIδ was visible in TTSO but not TTSP rats, while high myocardial expression of MLKL and Casp-1 was visible both in TTSO and TTSP rats. The course of TTS is associated with activation of inflammatory-associated programmed cell death, necroptosis, and pyroptosis, therefore inflammation may be a primary response occurring simultaneously with cardiomyocyte death in TTS.
Collapse
Affiliation(s)
- Sonia Borodzicz-Jażdżyk
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1b Banacha Street, 02-097 Warsaw, Poland
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 1a Banacha Street, 02-097 Warsaw, Poland
| | - Agnieszka Kołodzińska
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 1a Banacha Street, 02-097 Warsaw, Poland
| | - Katarzyna Czarzasta
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1b Banacha Street, 02-097 Warsaw, Poland
| | - Małgorzata Wojciechowska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1b Banacha Street, 02-097 Warsaw, Poland
| | - Renata Główczyńska
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 1a Banacha Street, 02-097 Warsaw, Poland
| | - Benedykt Szczepankiewicz
- Department of Pathology, Medical University of Warsaw, 7 Pawińskiego Street, 02-106 Warsaw, Poland
| | - Liana Puchalska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1b Banacha Street, 02-097 Warsaw, Poland
| | - Grzegorz Opolski
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 1a Banacha Street, 02-097 Warsaw, Poland
| | - Agnieszka Cudnoch-Jędrzejewska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1b Banacha Street, 02-097 Warsaw, Poland
| |
Collapse
|
11
|
Necroptosis in heart disease: Molecular mechanisms and therapeutic implications. J Mol Cell Cardiol 2022; 169:74-83. [PMID: 35597275 DOI: 10.1016/j.yjmcc.2022.05.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/09/2022] [Accepted: 05/12/2022] [Indexed: 01/11/2023]
Abstract
Cell death is a crucial event underlying cardiac ischemic injury, pathological remodeling, and heart failure. Unlike apoptosis, necrosis had long been regarded as a passive and unregulated process. However, recent studies demonstrate that a significant subset of necrotic cell death is actively mediated through regulated pathways - a process known as "regulated necrosis". As a form of regulated necrosis, necroptosis is mediated by death receptors and executed through the activation of receptor interacting protein kinase 3 (RIPK3) and its downstream substrate mixed lineage kinase-like domain (MLKL). Recent studies have provided compelling evidence that necroptosis plays an important role in myocardial homeostasis, ischemic injury, pathological remodeling, and heart failure. Moreover, it has been shown that genetic and pharmacological manipulations of the necroptosis signaling pathway elicit cardioprotective effects. Important progress has also been made regarding the molecular mechanisms that regulate necroptotic cell death in vitro and in vivo. In this review, we discuss molecular and cellular mechanisms of necroptosis, potential crosstalk between necroptosis and other cell death pathways, functional implications of necroptosis in heart disease, and new therapeutic strategies that target necroptosis signaling.
Collapse
|