1
|
Mei Y, Chen X, Shi S, Lin W, Cheng Z, Fan X, Wu W, Han J, Huang W, Ye B, Dai S. GI-Y2, a novel gasdermin D inhibitor, attenuates sepsis-induced myocardial dysfunction by inhibiting gasdermin D-mediated pyroptosis in macrophages. Br J Pharmacol 2025. [PMID: 40165368 DOI: 10.1111/bph.70040] [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: 06/20/2024] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND AND PURPOSE Myocardial dysfunction is a significant complication associated with sepsis. However, there are currently no specific and effective treatments available. Inhibiting gasdermin D (GSDMD)-mediated pyroptosis has shown promise in mitigating sepsis-induced myocardial dysfunction. The GSDMD inhibitor Y2 (GI-Y2) has been demonstrated to directly bind to GSDMD. Nonetheless, it remains uncertain whether GI-Y2 offers a cardioprotective effect in the context of sepsis-induced myocardial dysfunction. EXPERIMENTAL APPROACH A mouse model of sepsis was created using lipopolysaccharide (LPS), caecal ligation and puncture. Following treatment with GI-Y2 or macrophage membrane-encapsulated GI-Y2 nanoparticles (GI-Y2@MM-NPs), myocardial dysfunction and pyroptosis levels in heart tissues were assessed. Transcriptome sequencing revealed the molecular mechanism of GI-Y2 in treating septic cardiomyopathy. KEY RESULTS We observed that GI-Y2 alleviated myocardial dysfunction and attenuated cardiac inflammation in mice induced by LPS, caecal ligation and puncture. GI-Y2 reduced macrophage pyroptosis and attenuated macrophage-mediated cardiomyocyte injury induced by LPS/nigericin. Concurrently, we confirmed the protective effect of GI-Y2 against LPS-induced cardiac dysfunction was abolished in the absence of GSDMD. Additionally, GI-Y2 attenuated the mitochondrial damage induced by LPS by inhibiting GSDMD in the mitochondria. Furthermore, we developed GI-Y2@MM-NPs to enhance the targeting capability of GI-Y2 towards macrophages in heart tissues and demonstrated its protective effect in vivo. CONCLUSION AND IMPLICATIONS These findings indicate that GI-Y2 alleviates septic myocardial injury and dysfunction by specifically targeting GSDMD, thereby inhibiting GSDMD-mediated pyroptosis and mitochondrial damage. Both GI-Y2 and GI-Y2@MM-NPs may serve as promising therapeutic options for addressing septic myocardial dysfunction.
Collapse
Affiliation(s)
- Yiling Mei
- The Key Laboratory of Emergency and Disaster Medicine of Wenzhou, Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xudong Chen
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Cardiology, Ningbo Hangzhou Bay Hospital, Ningbo, Zhejiang, China
| | - Si Shi
- First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wante Lin
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhenfeng Cheng
- Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Xiaoxi Fan
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenqi Wu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jibo Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Weijian Huang
- The Key Laboratory of Emergency and Disaster Medicine of Wenzhou, Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bozhi Ye
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shanshan Dai
- The Key Laboratory of Emergency and Disaster Medicine of Wenzhou, Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
2
|
Shi R, Sun W, Yin S, Sun T, Yang N, Zhang H, Yao Y, Lai K, Chen X, Yu P. Qihuang Zhuyu formula modulates mitochondrial function to inhibit platelet activation via endocannabinoid signaling mediated by the SIRT1/PPARα pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119006. [PMID: 39500463 DOI: 10.1016/j.jep.2024.119006] [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: 08/16/2024] [Revised: 10/12/2024] [Accepted: 10/26/2024] [Indexed: 11/18/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Developing effective drugs is urgent for preventing platelet activation and atherosclerotic diseases. Qihuang Zhuyu formula (QHZYF) is a curative medical plants formula in clinic while its molecular mechanism remains to be further elucidated. AIM OF THE STUDY To investigate how QHZYF, via the sirtuin 1 (SIRT1)/peroxisome proliferator-activated receptor alpha (PPARα) pathway, mediates the endocannabinoid system, regulates mitochondrial function, and inhibits platelet activation, thereby offering novel strategies for the prevention and treatment of cardiovascular diseases. METHODS The chemical constituents of QHZYF were characterized by ultra-performance liquid chromatography tandem with quadrupole time-of-flight mass spectrometry (UPLC-Q/TOF-MS). For the in vivo experiments, an atherosclerosis rat model was developed. Platelet activation and endocannabinoid levels were assessed by ELISA, while the expression of PPARα was evaluated by immunofluorescence. Additionally, the expression of endocannabinoid-degrading enzymes was analyzed by Western blotting. In the in vitro studies, the protein-protein interaction between SIRT1 and PPARα was initially investigated using co-immunoprecipitation (Co-IP). Thereafter, platelets were co-cultured with human umbilical vein endothelial cells (HUVECs) stimulated by oxidized low-density lipoprotein (ox-LDL). The mitochondrial function of platelets was examined by flow cytometry. Platelet activation and endocannabinoid levels were quantified using ELISA, and the expression of PPARα, endocannabinoid-degrading enzymes and proteins reflecting mitochondrial function were determined by Western blotting. Subsequently, QHZYF was incorporated into the aforementioned system, and the entire experimental protocol was replicated to explore how QHZYF influences platelet activation through the SIRT1/PPARα pathway-mediated endocannabinoid system. RESULTS In the in vivo experiments, it was observed that QHZYF significantly augmented the levels of PPARα, concurrently with a decrease in the expression of endocannabinoid-degrading enzymes. This was paralleled by a rise in endogenous cannabinoid levels and a reduction in platelet activation, a process that was found to be regulated by SIRT1. The co-immunoprecipitation (Co-IP) analysis substantiated the cooperative interaction between SIRT1 and PPARα, underscoring their role in mediating the endocannabinoid system, regulating mitochondrial function, and consequently influencing platelet activation. In the in vitro studies, a co-culture system involving platelets and HUVECs stimulated with ox-LDL was employed. It was noted that the SIRT1/PPARα pathway mediates the endocannabinoid system in regulating mitochondrial function, which in turn affects platelet activation. The incorporation of QHZYF-containing serum into this system produced outcomes consistent with those observed in the in vivo experiments. These findings suggest that QHZYF operates by modulating the endocannabinoid system via the SIRT1/PPARα pathway, thereby controlling mitochondrial function and inhibiting platelet activation. CONCLUSION SIRT1 may mediate endocannabinoid signaling through its interaction with PPARα, regulating mitochondrial function and subsequently influencing platelet activation. QHZYF effectively modulates the endocannabinoid system via the SIRT1/PPARα pathway, thereby inhibiting platelet activation.
Collapse
Affiliation(s)
- Ruijie Shi
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Department of Cardiology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weixin Sun
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China; Department of Cardiology, Yancheng Hospital of Traditional Chinese Medicine, Yancheng, China
| | - Siyuan Yin
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Department of Cardiology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tong Sun
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Department of Cardiology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ning Yang
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Haosheng Zhang
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China; Institute of Modern Biology, Nanjing University, Nanjing, China
| | - Yun Yao
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China; Jiangning District Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Kailun Lai
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Department of Cardiology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaohu Chen
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Department of Cardiology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.
| | - Peng Yu
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Department of Cardiology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.
| |
Collapse
|
3
|
Chen J, Zhang WJ, Liu XY, Hu TP, Gao N, Li ZH, Wang Y, Zhang GQ. KW-2449 Ameliorates Cardiac Dysfunction in a Rat Model of Sepsis-Induced Cardiomyopathy. Inflammation 2025:10.1007/s10753-024-02223-y. [PMID: 39843680 DOI: 10.1007/s10753-024-02223-y] [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: 10/10/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025]
Abstract
KW-2449 is a novel multitargeted kinase inhibitor that has been reported to alleviate chronic inflammation and altered immunity during the treatment of autoimmune diseases. The aim of the study was to investigate the effect of KW-2449 on sepsis-induced cardiomyopathy (SIC). A rat model of moderate SIC was induced using the cecal ligation and puncture (CLP) method. KW-2449 was administered to rats at 10 mg/kg for 3 consecutive days by intraperitoneal injection. At 24 hours after CLP, echocardiography, electrocardiogram, and hemodynamic analyses were performed. Blood and cardiac tissues were collected for further analysis. RNA sequencing (RNA-seq) analyses were used to identify the key genes affected by KW-2449 treatment during SIC. KW-2449 improved the liver dysfunction in septic rats. KW-2449 significantly improved left ventricular (LV) systolic function and hemodynamics compared to the CLP group. KW-2449 suppressed the systemic inflammatory response, decreased myocardial inflammation and cell apoptosis in the CLP rats. RNA-seq analyses indicated that there were a total of 2256 differentially expressed genes in the CLP group compared to the Control group, among which 63 genes were down-regulated and 59 genes were up-regulated by KW-2449. Specifically, Pparα was identified as a key target gene of KW-2449 in the treatment of SIC by RNA-seq analysis.KW-2449 also significantly upregulated the protein expression of Pparα in the LV tissue of septic rats. KW-2449 reduced systemic inflammation, cardiac inflammation, and improved cardiac dysfunction in the CLP-induced SIC rat model. The underlying mechanism of the cardio-protective role of KW-2449 in the CLP-induced SIC might be related to Pparα.
Collapse
Affiliation(s)
- Jie Chen
- Department of Emergency, China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China
| | - Wei-Jian Zhang
- Department of Emergency, China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China
| | - Xiao-Yu Liu
- Department of Emergency, China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China
| | - Tian-Peng Hu
- Department of Emergency, China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China
| | - Nan Gao
- Department of Emergency, China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China
| | - Zhong-Hao Li
- Department of Emergency, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yu Wang
- Department of Emergency, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Guo-Qiang Zhang
- Department of Emergency, China-Japan Friendship Hospital, Beijing, 100029, China.
| |
Collapse
|
4
|
Mosa AK, Majeed S, Ghafil FA, Hadi NR. Potential cardioprotective effect of trimetazidine in mice model of endotoxemia: role of AMPK-Nrf2. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2025; 78:35-44. [PMID: 40023854 DOI: 10.36740/wlek/190820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
OBJECTIVE Aim: To clarify the potential cardioprotective effect of Trimetazidine against experimentally sepsis-caused endotoxic cardiac injury damage in mice. PATIENTS AND METHODS Materials and Methods: 24 Mice were divided into four groups (n=6): Sham group, CLP group DMSO group, trimetazidine-treated group 50 mg/kg IP, 1hr before CLP, then the animals were sacrificed 24 hr after CLP and tissue sample was taken for measurement of TNF-α, TNF-αr1, IL-1β, HO-1, MPO, caspase-11, F2-isoprostane and serum troponin by ELISA and gene expression of AMPK-Nrf2 by qpcr and histopathological study. RESULTS Results: trimetazidine treated group showed significant changes as compared with clp group regarding TNF-α, TNF-αr1, IL-1β, HO-1, MPO, CASPASE-11, F2-ISOPROSTANE as well as affect tissue mRNA expression of AMPK-Nrf2 genes p<0.05. CONCLUSION Conclusions: We evaluate that Trimetazidine has cardio protective effects due to its anti-inflammatory and anti-oxidative action. Also, trimetazidine showed a cardio-protective effect as they affect tissue mRNA expression of AMPK-Nrf2 genes.
Collapse
Affiliation(s)
- Alaa Kadhum Mosa
- PHARMACOLOGY AND THERAPEUTIC DEPARTMENT, FACULTY OF MEDICINE, UNIVERSITY OF KUFA, KUFA, IRAQ
| | - Sahar Majeed
- PHARMACOLOGY AND THERAPEUTIC DEPARTMENT, FACULTY OF MEDICINE, UNIVERSITY OF KUFA, KUFA, IRAQ
| | | | - Najah Rayish Hadi
- PHARMACOLOGY AND THERAPEUTIC DEPARTMENT, FACULTY OF MEDICINE, UNIVERSITY OF KUFA, KUFA, IRAQ
| |
Collapse
|
5
|
Singh D, Oladimeji-Salami J, Akindele AJ. New insights on pharmacological and therapeutic potentials of trimetazidine beyond anti-anginal drug: A comprehensive review. Eur J Pharmacol 2024; 985:177062. [PMID: 39427862 DOI: 10.1016/j.ejphar.2024.177062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/27/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
Trimetazidine (TMZ) is a beneficial and well-tolerable anti-anginal drug which has protective action towards ischemia and reperfusion injury. TMZ performs its anti-ischemic effect by modifying cardiac metabolism without shifting the hemodynamic functions, so it represents an outstanding complementary perspective to the general angina treatment. TMZ possesses a positive impact on the inflammatory profile, and also endothelial function furthermore displays various benefits through minimising the number, as well as the intensity of angina strikes and ameliorating the clinical indication and symptoms of myocardium ischemia. It is administrated as monotherapy along with a combination of different antianginal drugs. Apart from anti-angina action, in recent years TMZ has shown various pharmacological activities such as neuroprotective, renal protective, hepato-protective, cardio-protective effects, and other beneficial pharmacological activities. We select to write the present review article to cover the different pharmacological and therapeutic potentials of TMZ.
Collapse
Affiliation(s)
- Dhirendra Singh
- Department of Pharmacology, M.M College of Pharmacy, Maharishi Markandeshwar Mullana, Ambala, Haryana, India.
| | - Joy Oladimeji-Salami
- Medical Biotechnology Department, National Biotechnology Research and Development Agency, Abuja, Nigeria
| | - Abidemi James Akindele
- Department of Pharmacology, Therapeutics & Toxicology, Faculty of Basic Medical Sciences, College of Medicine, University of Lagos, Idi-Araba, P.M.B. 12003, Lagos, Nigeria.
| |
Collapse
|
6
|
Ebrahim NA, Elnagar MR, El-Gamal R, Habotta OA, Albadawi EA, Albadrani M, Bahashwan AS, Hassan HM. Melatonin mitigates doxorubicin induced chemo brain in a rat model in a NRF2/p53-SIRT1 dependent pathway. Heliyon 2024; 10:e38081. [PMID: 39386846 PMCID: PMC11462207 DOI: 10.1016/j.heliyon.2024.e38081] [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: 05/14/2024] [Revised: 09/03/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
Cancer is a critical health problem, and chemotherapy administration is mandatory for its eradication. However, chemotherapy like doxorubicin (Dox) has serious side-effects including cognitive impairment or chemo brain. Melatonin is a neuroprotective agent that has antioxidant, and anti-inflammatory effects. We aimed to explore melatonin's effect on Dox-induced chemo brain to discover new mechanisms associated with Dox-induced neurotoxicity and try to prevent its occurrence. Thirty-two male albino rats had been equally divided into four groups; control, melatonin-administrated, Dox-induced chemo brain, and melatonin + Dox treated. On the 9th day, brain had been excised after scarification and had been assessed for reactive oxygen species measurement, histopathological analysis, immunohistochemical, gene and protein expressions for the nuclear factor erythroid 2-related factor 2 (Nrf2), p53 and Silent information regulator 2 homolog 1 (SIRT1). Our results show that melatonin coadministration diminished Dox induced hippocampal and prefrontal cortex (PFC) cellular degeneration. It alleviated Nitric Oxide (NO) level and reversed the decline of antioxidant enzyme activities. It also upregulated Nrf2, SIRT1 and downregulated p53 gene expression in rats receiving Dox. Moreover, melatonin elevated the protein expression level of Nrf2, SIRT1 and reduced p53 corresponding to immunohistochemical results. The data suggested that melatonin can mitigate Dox-induced neurotoxicity by aggravating the endogenous antioxidants and inducing neurogenesis through activation of Nrf2/p53-SIRT1signaling pathway in adult rats' PFC. These effects were associated with Nrf2, SIRT1 activation and p53 inhibition. This could be guidance to add melatonin as an adjuvant supplement to Dox regimens to limit its adverse effect on the brain function.
Collapse
Affiliation(s)
- Neven A. Ebrahim
- Department of Basic Medical Sciences, College of Medicine, Taibah University, Madinah, Saudi Arabia
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed R. Elnagar
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Al-Azhar University, Egypt
- Pharmacology Department, College of Pharmacy, The Islamic University, Najaf, 54001, Iraq
| | - Randa El-Gamal
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
- Medical Experimental Research Centre (MERC), Faculty of Medicine, Mansoura University, Egypt
- Department of Medical Biochemistry, Faculty of Medicine, Horus University, New Damietta, Egypt
- Department of Medical Biochemistry, Faculty of Medicine, New Mansoura University, Mansoura, Egypt
| | - Ola Ali Habotta
- Forensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, Mansoura University, Egypt
| | - Emad A. Albadawi
- Department of Basic Medical Sciences, College of Medicine, Taibah University, Madinah, Saudi Arabia
| | - Muayad Albadrani
- Department of Family and Community Medicine and Medical Education, College of Medicine, Taibah University, Madinah, Saudi Arabia
| | - Abdulrahman S. Bahashwan
- Department of Basic Medical Sciences, College of Medicine, Taibah University, Madinah, Saudi Arabia
| | - Hend M. Hassan
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Human Anatomy and Embryology Department, Faculty of Medicine, New Mansoura University, Egypt
| |
Collapse
|
7
|
Yang XR, Wen R, Yang N, Zhang TN. Role of sirtuins in sepsis and sepsis-induced organ dysfunction: A review. Int J Biol Macromol 2024; 278:134853. [PMID: 39163955 DOI: 10.1016/j.ijbiomac.2024.134853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 08/22/2024]
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. Sepsis causes a high mortality rate and current treatment focuses on supportive therapies but lacks specific therapeutic targets. Notably, sirtuins (SIRTs) shows potential clinical application in the treatment of sepsis. It has been demonstrated that SIRTs, the nicotinamide adenine dinucleotide+(NAD+)-dependent deacetylases that regulate key signaling pathways in eukaryotes and prokaryotes, are involved in a variety of biological processes. To date, seven mammalian yeast Sir2 homologs have been identified. SIRTs can regulate inflammation, oxidative stress, apoptosis, autophagy, and other pathways that play important roles in sepsis-induced organ dysfunction. However, the existing studies on SIRTs in sepsis are too scattered, and there is no relevant literature to integrate them. This review innovatively summarizes the different mechanisms of SIRTs in sepsis organ dysfunction according to the different systems, and focuses on SIRT agonists, inhibitors, and targeted drugs that have been proved to be effective in the treatment of sepsis, so as to integrate the clinical research and basic research closely. We searched PubMed for all literature related to SIRTs and sepsis since its inception using the following medical subject headings: sirtuins, SIRTs, and sepsis. Data on the mechanisms of SIRTs in sepsis-induced organ damage and their potential as targets for disease treatment were extracted.
Collapse
Affiliation(s)
- Xin-Ru Yang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ri Wen
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ni Yang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Tie-Ning Zhang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
8
|
Shen S, Li J, Wei Z, Liu Y, Kang L, Gu R, Sun X, Xu B, Li Q. Immune-response gene 1 deficiency aggravates inflammation-triggered cardiac dysfunction by inducing M1 macrophage polarization and aggravating Ly6C high monocyte recruitment. Biol Direct 2024; 19:86. [PMID: 39350193 PMCID: PMC11441264 DOI: 10.1186/s13062-024-00521-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 08/28/2024] [Indexed: 10/04/2024] Open
Abstract
The immune response gene 1 (IRG1) and its metabolite itaconate are implicated in modulating inflammation and oxidative stress, with potential relevance to sepsis-induced myocardial dysfunction (SIMD). This study investigates their roles in SIMD using both in vivo and in vitro models. Mice were subjected to lipopolysaccharide (LPS)-induced sepsis, and cardiac function was assessed in IRG1 knockout (IRG1-/-) and wild-type mice. Exogenous 4-octyl itaconate (4-OI) supplementation was also examined for its protective effects. In vitro, bone marrow-derived macrophages and RAW264.7 cells were treated with 4-OI following Nuclear factor, erythroid 2 like 2 (NRF2)-small interfering RNA administration to elucidate the underlying mechanisms. Our results indicate that IRG1 deficiency exacerbates myocardial injury during sepsis, while 4-OI administration preserves cardiac function and reduces inflammation. Mechanistic insights reveal that 4-OI activates the NRF2/HO-1 pathway, promoting macrophage polarization and attenuating inflammation. These findings underscore the protective role of the IRG1/itaconate axis in SIMD and suggest a therapeutic potential for 4-OI in modulating macrophage responses.
Collapse
Affiliation(s)
- Song Shen
- Department of Cardiology of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jianhui Li
- Department of Cardiology of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhonghai Wei
- Department of Cardiology of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yihai Liu
- Department of Cardiology of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Lina Kang
- Department of Cardiology of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Rong Gu
- Department of Cardiology of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Xuan Sun
- Department of Cardiology of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Biao Xu
- Department of Cardiology of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| | - QiaoLing Li
- Department of Cardiology of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
9
|
Bi C, Wang D, Hao B, Yang T. Snhg14/miR-181a-5p axis-mediated "M1" macrophages aggravate LPS-induced myocardial cell injury. Heliyon 2024; 10:e37104. [PMID: 39309894 PMCID: PMC11414504 DOI: 10.1016/j.heliyon.2024.e37104] [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: 04/17/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
An increasing number of studies have suggested that macrophages participate in sepsis-induced myocardial injury. Our study highlights the function and mechanism of the lncRNA Snhg14 in "M1" polarized macrophage-mediated myocardial cell damage. Lipopolysaccharide (LPS) was used to treat H9c2 cells to construct an in vitro myocardial injury model. M1 and M2 polarization of RAW264.7 cells were induced and the exosomes were obtained from the supernatant through ultracentrifugation. Moreover, cecal ligation and puncture (CLP) surgery was implemented to establish a mouse sepsis-induced myocardial injury model, and Snhg14 was knocked down with sh-Snhg14. The results showed that the conditioned medium (CM) and the exosomes (Exo) of M1 macrophages substantially augmented LPS-induced apoptosis and oxidative stress in myocardial cells. Notably, M1-CM and M1-Exo contributed to nearly 50 % of myocardial cell viability decline. Snhg14 was highly expressed in M1 macrophages and exosomes derived from M1-MΦ (M1-Exo). Snhg14 overexpression aggravated myocardial cell damage and increased 10 to 50 times expression of proinflammatory cytokines in MΦ. Snhg14 knockdown reversed M1-Exo-mediated myocardial cell damage and inhibited the production of proinflammatory cytokines (50 %-75 % decline) of MΦ. Moreover, Snhg14 targeted and inhibited miR-181a-5p expression. miR-181a-5p upregulation partly reversed Snhg4 overexpression-mediated myocardial cell damage and MΦ activation. In vivo, sh-Snhg14 dramatically ameliorated cardiac damage in septic mice by enhancing miR-181a-5p and inhibiting the HMGB1/NF-κB pathway. In conclusion, "M1" macrophage-derived exosomal Snhg14 aggravates myocardial cell damage by modulating the miR-181a-5p/HMGB1/NF-κB pathway.
Collapse
Affiliation(s)
- Chenglong Bi
- Department of Cardiology, Shandong University Zibo Central Hospital, Zibo, 255000, Shandong, China
| | - Dejin Wang
- Department of Cardiology, Shandong University Zibo Central Hospital, Zibo, 255000, Shandong, China
| | - Bin Hao
- Cardiovascular Surgery, Shandong University Zibo Central Hospital, Zibo, 255000, China
| | - Tianxiao Yang
- Department of Cardiology, Shandong University Zibo Central Hospital, Zibo, 255000, Shandong, China
| |
Collapse
|
10
|
Jiang J, Shu H, Wang DW, Hui R, Li C, Ran X, Wang H, Zhang J, Nie S, Cui G, Xiang D, Shao Q, Xu S, Zhou N, Li Y, Gao W, Chen Y, Bian Y, Wang G, Xia L, Wang Y, Zhao C, Zhang Z, Zhao Y, Wang J, Chen S, Jiang H, Chen J, Du X, Chen M, Sun Y, Li S, Ding H, Ma X, Zeng H, Lin L, Zhou S, Ma L, Tao L, Chen J, Zhou Y, Guo X. Chinese Society of Cardiology guidelines on the diagnosis and treatment of adult fulminant myocarditis. SCIENCE CHINA. LIFE SCIENCES 2024; 67:913-939. [PMID: 38332216 DOI: 10.1007/s11427-023-2421-0] [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: 03/21/2023] [Accepted: 07/25/2023] [Indexed: 02/10/2024]
Abstract
Fulminant myocarditis is an acute diffuse inflammatory disease of myocardium. It is characterized by acute onset, rapid progress and high risk of death. Its pathogenesis involves excessive immune activation of the innate immune system and formation of inflammatory storm. According to China's practical experience, the adoption of the "life support-based comprehensive treatment regimen" (with mechanical circulation support and immunomodulation therapy as the core) can significantly improve the survival rate and long-term prognosis. Special emphasis is placed on very early identification,very early diagnosis,very early prediction and very early treatment.
Collapse
Affiliation(s)
- Jiangang Jiang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hongyang Shu
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dao Wen Wang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Rutai Hui
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Chenze Li
- Zhongnan Hospital of Wuhan University, Wuhan, 430062, China
| | - Xiao Ran
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hong Wang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jing Zhang
- Fuwai Huazhong Cardiovascular Hospital, Zhengzhou, 450003, China
| | - Shaoping Nie
- Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Guanglin Cui
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dingcheng Xiang
- Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, 510010, China
| | - Qun Shao
- Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Shengyong Xu
- Union Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ning Zhou
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuming Li
- Taida Hospital, Tianjin, 300457, China
| | - Wei Gao
- Peking University Third Hospital, Beijing, 100191, China
| | - Yuguo Chen
- Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yuan Bian
- Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Guoping Wang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Liming Xia
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yan Wang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chunxia Zhao
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhiren Zhang
- Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yuhua Zhao
- Kanghua Hospital, Dongguan, Guangzhou, 523080, China
| | - Jianan Wang
- Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Shaoliang Chen
- Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Hong Jiang
- Renmin Hospital of Wuhan University, Wuhan, 430060, Wuhan, China
| | - Jing Chen
- Renmin Hospital of Wuhan University, Wuhan, 430060, Wuhan, China
| | - Xianjin Du
- Renmin Hospital of Wuhan University, Wuhan, 430060, Wuhan, China
| | - Mao Chen
- West China Hospital, Sichuan University, Chengdu, 610044, China
| | - Yinxian Sun
- First Hospital of China Medical University, Shenyang, 110002, China
| | - Sheng Li
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hu Ding
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xueping Ma
- General Hospital of Ningxia Medical University, Yinchuan, 750003, China
| | - Hesong Zeng
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li Lin
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shenghua Zhou
- The Second Xiangya Hospital, Central South University, Changsha, 410012, China
| | - Likun Ma
- The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230002, China
| | - Ling Tao
- The First Affiliated Hospital of Air Force Medical University, Xi'an, 710032, China
| | - Juan Chen
- Central Hospital of Wuhan City, Wuhan, 430014, China
| | - Yiwu Zhou
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaomei Guo
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
11
|
Majid Z, Muhammad-Baqir B, Al-Shimerty DF, Hadi NR. The possible cardioprotective effect of ghrelin during experimental endotoxemia in mice. J Med Life 2024; 17:486-491. [PMID: 39144689 PMCID: PMC11320619 DOI: 10.25122/jml-2023-0228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 10/04/2023] [Indexed: 08/16/2024] Open
Abstract
This study aimed to evaluate the cardioprotective effects of ghrelin in septic mice, focusing on its anti-inflammatory and antioxidant properties. Thirty-five male Swiss mice (8-12 weeks old, 23-33g) were randomly assigned to five groups (n = 7 each): (1) Normal, fed usual diets, (2) Sham, subjected to anesthesia and laparotomy, (3) Sepsis, subjected to cecal ligation and puncture, (4) Vehicle, given an equivalent volume of intraperitoneal saline injections immediately after cecal ligation and puncture, and (5) Ghrelin-treated, administered 80 µg/kg ghrelin intraperitoneal injections immediately following cecal ligation and puncture. Serum levels of tumor necrosis factor-alpha (TNF-α), macrophage migration inhibitory factor (MIF), toll-like receptor 4 (TLR4), and 8-epi-prostaglandin F2 alpha (8-epi-PGF2α) were measured. The extent of cardiac damage was also evaluated histologically. The mean serum levels of TNF-α, MIF, TLR4, and 8-epi-PGF2α levels were significantly higher in the sepsis and vehicle groups than in the normal and sham groups. The levels were significantly lower in the ghrelin-treated group than in the vehicle and sepsis groups. Histological analysis revealed normal myocardial architecture in the normal and sham groups, whereas the sepsis and vehicle groups had severe myocardial injury. The ghrelin-treated group displayed histological features similar to the sham group, indicating reduced myocardial damage. Ghrelin ameliorated sepsis-induced cardiotoxicity in mice by exhibiting strong anti-inflammatory and antioxidant effects. These findings suggest that ghrelin may be a promising therapeutic candidate for the prevention of sepsis-induced cardiotoxicity.
Collapse
Affiliation(s)
| | | | | | - Najah Rayish Hadi
- Department of Pharmacology & Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq
| |
Collapse
|
12
|
You J, Li Y, Chong W. The role and therapeutic potential of SIRTs in sepsis. Front Immunol 2024; 15:1394925. [PMID: 38690282 PMCID: PMC11058839 DOI: 10.3389/fimmu.2024.1394925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 04/03/2024] [Indexed: 05/02/2024] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by the host's dysfunctional response to infection. Abnormal activation of the immune system and disturbance of energy metabolism play a key role in the development of sepsis. In recent years, the Sirtuins (SIRTs) family has been found to play an important role in the pathogenesis of sepsis. SIRTs, as a class of histone deacetylases (HDACs), are widely involved in cellular inflammation regulation, energy metabolism and oxidative stress. The effects of SIRTs on immune cells are mainly reflected in the regulation of inflammatory pathways. This regulation helps balance the inflammatory response and may lessen cell damage and organ dysfunction in sepsis. In terms of energy metabolism, SIRTs can play a role in immunophenotypic transformation by regulating cell metabolism, improve mitochondrial function, increase energy production, and maintain cell energy balance. SIRTs also regulate the production of reactive oxygen species (ROS), protecting cells from oxidative stress damage by activating antioxidant defense pathways and maintaining a balance between oxidants and reducing agents. Current studies have shown that several potential drugs, such as Resveratrol and melatonin, can enhance the activity of SIRT. It can help to reduce inflammatory response, improve energy metabolism and reduce oxidative stress, showing potential clinical application prospects for the treatment of sepsis. This review focuses on the regulation of SIRT on inflammatory response, energy metabolism and oxidative stress of immune cells, as well as its important influence on multiple organ dysfunction in sepsis, and discusses and summarizes the effects of related drugs and compounds on reducing multiple organ damage in sepsis through the pathway involving SIRTs. SIRTs may become a new target for the treatment of sepsis and its resulting organ dysfunction, providing new ideas and possibilities for the treatment of this life-threatening disease.
Collapse
Affiliation(s)
- Jiaqi You
- Department of Emergency, The First Hospital of China Medical University, Shenyang, China
| | - Yilin Li
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Wei Chong
- Department of Emergency, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
13
|
Zheng MH, Liu WJ, Yang J. Effect of early stepwise cardiopulmonary rehabilitation on function and quality of life in sepsis patients. World J Clin Cases 2024; 12:729-736. [PMID: 38322673 PMCID: PMC10841144 DOI: 10.12998/wjcc.v12.i4.729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/04/2023] [Accepted: 01/08/2024] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND Sepsis, as a non-limiting host infection disease, can be accompanied by serious complications such as organ failure, which seriously threatens patient quality of life. AIM To investigate the effect of early stepwise cardiopulmonary rehabilitation on cardiopulmonary function and quality of life in patients evacuated from mechanical ventilation with sepsis. METHODS A total of 80 patients with sepsis who were hospitalized in our hospital from January 2021 to January 2022 were selected and divided into the observation group (n = 40) and the control group (n = 40) according to the random number table method. The observation group was treated with early stepwise cardiopulmonary rehabilitation, and the control group was treated with a conventional treatment regimen. Cardiac function indexes (central venous pressure, cardiac troponin I, B-type brain natriuretic peptide), lung function indicators (diaphragmatic mobility, changes in central venous oxygen saturation, oxygenation index), and quality of life (Quality of Life Evaluation Scale) were compared between the two groups after treatment. RESULTS After treatment, the central venous pressure, diaphragm mobility, central venous oxygen saturation, oxygenation index, and Quality of Life Evaluation Scale scores in the observation group were higher than those in the control group, and the differences were statistically significant (P < 0.05). The observation group was less than that of the control group for other parameters, and the differences were statistically significant (P < 0.05). CONCLUSION Early stepwise cardiopulmonary rehabilitation can effectively enhance cardiac and pulmonary function and improve the quality of life in patients evacuated from mechanical ventilation with sepsis.
Collapse
Affiliation(s)
- Ming-Hui Zheng
- Division of Cardiovascular First Ward Departments of Internal Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, Hubei Province, China
| | - Wen-Jun Liu
- Department of Respiratory Medicine, PuRen Hospital Affiliated to Wuhan University of Science and Technology/Respiratory Medicine, Wuhan 430000, Hubei Province, China
| | - Juan Yang
- Department of Cardiology, Huanggang Central Hospital, Huanggang 438000, Hubei Province, China
| |
Collapse
|
14
|
Liu D, Li L, Li Z. Anemonin inhibits sepsis-induced acute kidney injury via mitigating inflammation and oxidative stress. Biotechnol Appl Biochem 2023; 70:1983-2001. [PMID: 37592376 DOI: 10.1002/bab.2504] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/08/2023] [Indexed: 08/19/2023]
Abstract
Elevated inflammation and oxidative stress (OS) are the main pathologic features of acute kidney injury (AKI)-caused by sepsis. Here, we made an investigation into the protective effects of the natural compound Anemonin (ANE) on sepsis-induced AKI both in vitro and in vivo. Lipopolysaccharide (LPS) was applied to construct an in vitro AKI model in renal tubular epithelial cells, and the septic C57BL/6J mouse model was constructed via cecal ligation and puncture (CLP). Cell viability and apoptosis were detected. The levels of p53, Bax, Bcl2, Caspase3, Caspase8, Caspase9, AMP-activated protein kinase (AMPK), Sirt-1, and forkhead box O3 were determined by Western Blot or RT-PCR. The reactive oxygen species level and OS markers were measured. Furthermore, the pathological changes of kidneys were evaluated by hematoxylin-eosin staining and immunohistochemistry. As per the information presented, ANE improved LPS-elicited apoptosis, inflammatory response, and OS in a dose-dependent pattern in renal tubular epithelial cells. Besides, ANE activated the AMPK/Sirt-1 pathway, and the AMPK inhibitor (Compound C) and Sirt-1 inhibitor (EX-527) significantly attenuated ANE-mediated protection on renal tubular epithelial cells. In vivo, ANE mitigated the levels of serum creatinine and urea nitrogen in the CLP-induced mouse sepsis model, reduced the renal tissue injury score, and attenuated OS, inflammation, and apoptosis levels in the kidney. Taken together, this study suggested that ANE has protective effects in sepsis-triggered AKI through repressing inflammation, OS, and cell apoptosis by activating the AMPK/Sirt-1 pathway.
Collapse
Affiliation(s)
- Dan Liu
- Department of Nephrology, First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Li Li
- Department of Nephrology, First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Zengyan Li
- Department of Nephrology, First Affiliated Hospital of Baotou Medical College, Baotou, China
| |
Collapse
|
15
|
Bortolasci CC, Kidnapillai S, Spolding B, Truong TTT, Connor T, Swinton C, Panizzutti B, Liu ZSJ, Sanigorski A, Dean OM, Crowley T, Richardson M, Bozaoglu K, Vlahos K, Cowdery S, Watmuff B, Steyn SF, Wolmarans DW, Engelbrecht BJ, Perry C, Drummond K, Pang T, Jamain S, Gray L, McGee SL, Harvey BH, Kim JH, Leboyer M, Berk M, Walder K. Use of a gene expression signature to identify trimetazidine for repurposing to treat bipolar depression. Bipolar Disord 2023; 25:661-670. [PMID: 36890661 PMCID: PMC10946906 DOI: 10.1111/bdi.13319] [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] [Indexed: 03/10/2023]
Abstract
OBJECTIVES The aim of this study was to repurpose a drug for the treatment of bipolar depression. METHODS A gene expression signature representing the overall transcriptomic effects of a cocktail of drugs widely prescribed to treat bipolar disorder was generated using human neuronal-like (NT2-N) cells. A compound library of 960 approved, off-patent drugs were then screened to identify those drugs that affect transcription most similar to the effects of the bipolar depression drug cocktail. For mechanistic studies, peripheral blood mononuclear cells were obtained from a healthy subject and reprogrammed into induced pluripotent stem cells, which were then differentiated into co-cultured neurons and astrocytes. Efficacy studies were conducted in two animal models of depressive-like behaviours (Flinders Sensitive Line rats and social isolation with chronic restraint stress rats). RESULTS The screen identified trimetazidine as a potential drug for repurposing. Trimetazidine alters metabolic processes to increase ATP production, which is thought to be deficient in bipolar depression. We showed that trimetazidine increased mitochondrial respiration in cultured human neuronal-like cells. Transcriptomic analysis in induced pluripotent stem cell-derived neuron/astrocyte co-cultures suggested additional mechanisms of action via the focal adhesion and MAPK signalling pathways. In two different rodent models of depressive-like behaviours, trimetazidine exhibited antidepressant-like activity with reduced anhedonia and reduced immobility in the forced swim test. CONCLUSION Collectively our data support the repurposing of trimetazidine for the treatment of bipolar depression.
Collapse
Affiliation(s)
- Chiara C. Bortolasci
- IMPACTThe Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin UniversityGeelongAustralia
| | - Srisaiyini Kidnapillai
- IMPACTThe Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin UniversityGeelongAustralia
| | - Briana Spolding
- IMPACTThe Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin UniversityGeelongAustralia
| | - Trang T. T. Truong
- IMPACTThe Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin UniversityGeelongAustralia
| | - Timothy Connor
- IMPACTThe Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin UniversityGeelongAustralia
| | - Courtney Swinton
- IMPACTThe Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin UniversityGeelongAustralia
| | - Bruna Panizzutti
- IMPACTThe Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin UniversityGeelongAustralia
| | - Zoe S. J. Liu
- IMPACTThe Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin UniversityGeelongAustralia
| | - Andrew Sanigorski
- IMPACTThe Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin UniversityGeelongAustralia
| | - Olivia M. Dean
- IMPACTThe Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin UniversityGeelongAustralia
- The Florey Institute of Neuroscience and Mental HealthParkvilleAustralia
| | - Tamsyn Crowley
- IMPACTThe Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin UniversityGeelongAustralia
- Bioinformatics Core Research Facility (BCRF)Deakin UniversityGeelongAustralia
| | - Mark Richardson
- Bioinformatics Core Research Facility (BCRF)Deakin UniversityGeelongAustralia
| | - Kiymet Bozaoglu
- Murdoch Children's Research InstituteParkvilleVictoriaAustralia
- Department of PaediatricsUniversity of MelbourneParkvilleVictoriaAustralia
| | - Katerina Vlahos
- Murdoch Children's Research InstituteParkvilleVictoriaAustralia
| | - Stephanie Cowdery
- IMPACTThe Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin UniversityGeelongAustralia
| | - Brad Watmuff
- IMPACTThe Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin UniversityGeelongAustralia
| | - Stephan F. Steyn
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health SciencesNorth‐West UniversityPotchefstroomSouth Africa
| | - De Wet Wolmarans
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health SciencesNorth‐West UniversityPotchefstroomSouth Africa
| | - Barend J. Engelbrecht
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health SciencesNorth‐West UniversityPotchefstroomSouth Africa
| | - Christina Perry
- The Florey Institute of Neuroscience and Mental HealthParkvilleAustralia
| | - Katherine Drummond
- The Florey Institute of Neuroscience and Mental HealthParkvilleAustralia
| | - Terence Pang
- The Florey Institute of Neuroscience and Mental HealthParkvilleAustralia
| | - Stéphane Jamain
- Univ Paris Est Créteil, INSERM, IMRB, Translational Neuropsychiatry, AP‐HP, DMU IMPACT, FHU ADAPTFondation FondaMentalCréteilFrance
| | - Laura Gray
- IMPACTThe Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin UniversityGeelongAustralia
- The Florey Institute of Neuroscience and Mental HealthParkvilleAustralia
| | - Sean L. McGee
- IMPACTThe Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin UniversityGeelongAustralia
| | - Brian H. Harvey
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health SciencesNorth‐West UniversityPotchefstroomSouth Africa
- SAMRC Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Mental Health and Neuroscience InstituteUniversity of Cape TownCape TownSouth Africa
| | - Jee Hyun Kim
- IMPACTThe Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin UniversityGeelongAustralia
- The Florey Institute of Neuroscience and Mental HealthParkvilleAustralia
| | - Marion Leboyer
- Univ Paris Est Créteil, INSERM, IMRB, Translational Neuropsychiatry, AP‐HP, DMU IMPACT, FHU ADAPTFondation FondaMentalCréteilFrance
| | - Michael Berk
- IMPACTThe Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin UniversityGeelongAustralia
- The Florey Institute of Neuroscience and Mental HealthParkvilleAustralia
- Orygen, The National Centre of Excellence in Youth Mental HealthParkvilleAustralia
| | - Ken Walder
- IMPACTThe Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin UniversityGeelongAustralia
| |
Collapse
|
16
|
Khanra S, Reddy P, Giménez-Palomo A, Park CHJ, Panizzutti B, McCallum M, Arumugham SS, Umesh S, Debnath M, Das B, Venkatasubramanian G, Ashton M, Turner A, Dean OM, Walder K, Vieta E, Yatham LN, Pacchiarotti I, Reddy YCJ, Goyal N, Kesavan M, Colomer L, Berk M, Kim JH. Metabolic regulation to treat bipolar depression: mechanisms and targeting by trimetazidine. Mol Psychiatry 2023; 28:3231-3242. [PMID: 37386057 PMCID: PMC10618096 DOI: 10.1038/s41380-023-02134-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 05/14/2023] [Accepted: 06/13/2023] [Indexed: 07/01/2023]
Abstract
Bipolar disorder's core feature is the pathological disturbances in mood, often accompanied by disrupted thinking and behavior. Its complex and heterogeneous etiology implies that a range of inherited and environmental factors are involved. This heterogeneity and poorly understood neurobiology pose significant challenges to existing drug development paradigms, resulting in scarce treatment options, especially for bipolar depression. Therefore, novel approaches are needed to discover new treatment options. In this review, we first highlight the main molecular mechanisms known to be associated with bipolar depression-mitochondrial dysfunction, inflammation and oxidative stress. We then examine the available literature for the effects of trimetazidine in said alterations. Trimetazidine was identified without a priori hypothesis using a gene-expression signature for the effects of a combination of drugs used to treat bipolar disorder and screening a library of off-patent drugs in cultured human neuronal-like cells. Trimetazidine is used to treat angina pectoris for its cytoprotective and metabolic effects (improved glucose utilization for energy production). The preclinical and clinical literature strongly support trimetazidine's potential to treat bipolar depression, having anti-inflammatory and antioxidant properties while normalizing mitochondrial function only when it is compromised. Further, trimetazidine's demonstrated safety and tolerability provide a strong rationale for clinical trials to test its efficacy to treat bipolar depression that could fast-track its repurposing to address such an unmet need as bipolar depression.
Collapse
Affiliation(s)
- Sourav Khanra
- Department of Psychiatry, Central Institute of Psychiatry, Ranchi, Jharkhand, India
| | - Preethi Reddy
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, Karnataka, India
| | - Anna Giménez-Palomo
- Bipolar and Depressive Disorders Unit, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Mental Health Biomedical Research Networking Center (CIBERSAM), Madrid, Spain
| | - Chun Hui J Park
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Bruna Panizzutti
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Madeleine McCallum
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Shyam Sundar Arumugham
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, Karnataka, India
| | - Shreekantiah Umesh
- Department of Psychiatry, Central Institute of Psychiatry, Ranchi, Jharkhand, India
| | - Monojit Debnath
- Department of Human Genetics, NIMHANS, Bengaluru, Karnataka, India
| | - Basudeb Das
- Department of Psychiatry, Central Institute of Psychiatry, Ranchi, Jharkhand, India
| | - Ganesan Venkatasubramanian
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, Karnataka, India
| | - Melanie Ashton
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Alyna Turner
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Olivia M Dean
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Ken Walder
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Eduard Vieta
- Bipolar and Depressive Disorders Unit, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Mental Health Biomedical Research Networking Center (CIBERSAM), Madrid, Spain
| | - Lakshmi N Yatham
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Isabella Pacchiarotti
- Bipolar and Depressive Disorders Unit, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Mental Health Biomedical Research Networking Center (CIBERSAM), Madrid, Spain
| | - Y C Janardhan Reddy
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, Karnataka, India
| | - Nishant Goyal
- Department of Psychiatry, Central Institute of Psychiatry, Ranchi, Jharkhand, India
| | - Muralidharan Kesavan
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, Karnataka, India
| | - Lluc Colomer
- Bipolar and Depressive Disorders Unit, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Mental Health Biomedical Research Networking Center (CIBERSAM), Madrid, Spain
| | - Michael Berk
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia.
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia.
| | - Jee Hyun Kim
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia.
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
17
|
Ye S, Huang H, Xiao Y, Han X, Shi F, Luo W, Chen J, Ye Y, Zhao X, Huang W, Wang Y, Lai D, Liang G, Fu G. Macrophage Dectin-1 mediates Ang II renal injury through neutrophil migration and TGF-β1 secretion. Cell Mol Life Sci 2023; 80:184. [PMID: 37340199 DOI: 10.1007/s00018-023-04826-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 05/10/2023] [Accepted: 05/26/2023] [Indexed: 06/22/2023]
Abstract
Macrophage activation has been shown to play an essential role in renal fibrosis and dysfunction in hypertensive chronic kidney disease. Dectin-1 is a pattern recognition receptor that is also involved in chronic noninfectious diseases through immune activation. However, the role of Dectin-1 in Ang II-induced renal failure is still unknown. In this study, we found that Dectin-1 expression on CD68 + macrophages was significantly elevated in the kidney after Ang II infusion. We assessed the effect of Dectin-1 on hypertensive renal injury using Dectin-1-deficient mice infused by Angiotensin II (Ang II) at 1000 ng/kg/min for 4 weeks. Ang II-induced renal dysfunction, interstitial fibrosis, and immune activation were significantly attenuated in Dectin-1-deficient mice. A Dectin-1 neutralizing antibody and Syk inhibitor (R406) were used to examine the effect and mechanism of Dectin-1/Syk signaling axle on cytokine secretion and renal fibrosis in culturing cells. Blocking Dectin-1 or inhibiting Syk significantly reduced the expression and secretion of chemokines in RAW264.7 macrophages. The in vitro data showed that the increase in TGF-β1 in macrophages enhanced the binding of P65 and its target promotor via the Ang II-induced Dectin-1/Syk pathway. Secreted TGF-β1 caused renal fibrosis in kidney cells through Smad3 activation. Thus, macrophage Dectin-1 may be involved in the activation of neutrophil migration and TGF-β1 secretion, thereby promoting kidney fibrosis and dysfunction.
Collapse
Affiliation(s)
- Shiju Ye
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 311399, Zhejiang, China
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - He Huang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
| | - Yun Xiao
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
| | - Xue Han
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 311399, Zhejiang, China
| | - Fengjie Shi
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
| | - Wu Luo
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 311399, Zhejiang, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jiawen Chen
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
| | - Yang Ye
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
| | - Xia Zhao
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 311399, Zhejiang, China
| | - Weijian Huang
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Dongwu Lai
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
| | - Guang Liang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 311399, Zhejiang, China.
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Guosheng Fu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China.
| |
Collapse
|
18
|
Lin L, Wei J, Zhu C, Hao G, Xue J, Zhu Y, Wu R. Sema3A alleviates viral myocarditis by modulating SIRT1 to regulate cardiomyocyte mitophagy. ENVIRONMENTAL TOXICOLOGY 2023; 38:1305-1317. [PMID: 36880403 DOI: 10.1002/tox.23765] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/25/2022] [Accepted: 02/20/2023] [Indexed: 05/18/2023]
Abstract
Viral myocarditis (VMC) is a common myocardial inflammatory disease characterized by inflammatory cell infiltration and cardiomyocyte necrosis. Sema3A was reported to reduce cardiac inflammation and improve cardiac function after myocardial infarction, but its role in VMC remains to be explored. Here, a VMC mouse model was established by infection with CVB3, and Sema3A was overexpressed in vivo by intraventricular injection of an adenovirus-mediated Sema3A expression vector (Ad-Sema3A). We found that Sema3A overexpression attenuated CVB3-induced cardiac dysfunction and tissue inflammation. And Sema3A also reduced macrophage accumulation and NLRP3 inflammasome activation in the myocardium of VMC mice. In vitro, LPS was used to stimulate primary splenic macrophages to mimic the macrophage activation state in vivo. Activated macrophages were co-cultured with primary mouse cardiomyocytes to evaluate macrophage infiltration-induced cardiomyocyte damage. Ectopic expression of Sema3A in cardiomyocytes effectively protected cardiomyocytes from activated macrophage-induced inflammation, apoptosis, and ROS accumulation. Mechanistically, cardiomyocyte-expressed Sema3A mitigated macrophage infiltration-caused cardiomyocyte dysfunction by promoting cardiomyocyte mitophagy and hindering NLRP3 inflammasome activation. Furthermore, NAM (a SIRT1 inhibitor) reversed the protective effect of Sema3A against activated macrophage-induced cardiomyocyte dysfunction by suppressing cardiomyocyte mitophagy. In conclusion, Sema3A promoted cardiomyocyte mitophagy and suppressed inflammasome activation by regulating SIRT1, thereby attenuating macrophage infiltration-induced cardiomyocyte injury in VMC.
Collapse
Affiliation(s)
- Lin Lin
- Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jin Wei
- Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Canzhan Zhu
- Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Guanghua Hao
- Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiahong Xue
- Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yanhe Zhu
- Department of Medicine, School of Public Health, Institute of Endemic Diseases, Xi'an Jiaotong University, Xi'an, China
| | - Ruiyun Wu
- Department of Medicine, School of Public Health, Institute of Endemic Diseases, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
19
|
Ye S, Huang H, Han X, Luo W, Wu L, Ye Y, Gong Y, Zhao X, Huang W, Wang Y, Long X, Fu G, Liang G. Dectin-1 Acts as a Non-Classical Receptor of Ang II to Induce Cardiac Remodeling. Circ Res 2023; 132:707-722. [PMID: 36786193 DOI: 10.1161/circresaha.122.322259] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
BACKGROUND Cardiac remodeling in heart failure involves macrophage-mediated immune responses. Recent studies have shown that a PRR (pattern recognition receptor) called dectin-1, expressed on macrophages, mediates proinflammatory responses. Whether dectin-1 plays a role in pathological cardiac remodeling is unknown. Here, we identified a potential role of dectin-1 in this disease. METHODS To model aberrant cardiac remodeling, we utilized mouse models of chronic Ang II (angiotensin II) infusion. In this model, we assessed the potential role of dectin-1 through using D1KO (dectin-1 knockout) mice and bone marrow transplantation chimeric mice. We then used cellular and molecular assays to discover the underlying mechanisms of dectin-1 function. RESULTS We found that macrophage dectin-1 is elevated in mouse heart tissues following chronic Ang II administration. D1KO mice were significantly protected against Ang II-induced cardiac dysfunction, hypertrophy, fibrosis, inflammatory responses, and macrophage infiltration. Further bone marrow transplantation studies showed that dectin-1 deficiency in bone marrow-derived cells prevented Ang II-induced cardiac inflammation and dysfunction. Through detailed molecular studies, we show that Ang II binds directly to dectin-1, causing dectin-1 homodimerization and activating the downstream Syk (spleen tyrosine kinase)/NF-κB (nuclear factor kappa B) signaling pathway to induce expression of inflammatory and chemoattractant factors. Mutagenesis studies identified R184 in the C-type lectin domain to interact with Ang II. Blocking dectin-1 in macrophages suppresses Ang II-induced inflammatory mediators and subsequent intercellular cross talk with cardiomyocytes and fibroblasts. CONCLUSIONS Our study has discovered dectin-1 as a new nonclassical receptor of Ang II and a key player in cardiac remolding and dysfunction. These studies suggest that dectin-1 may be a new target for treating hypertension-related heart failure.
Collapse
Affiliation(s)
- Shiju Ye
- School of Pharmaceutical Sciences, Hangzhou Medical College, Zhejiang, China (S.Y., X.H., W.L., X.Z., G.L.).,Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.).,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.).,Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China (S.Y., W.H.)
| | - He Huang
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.).,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.)
| | - Xue Han
- School of Pharmaceutical Sciences, Hangzhou Medical College, Zhejiang, China (S.Y., X.H., W.L., X.Z., G.L.)
| | - Wu Luo
- School of Pharmaceutical Sciences, Hangzhou Medical College, Zhejiang, China (S.Y., X.H., W.L., X.Z., G.L.).,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang, China (W.L., Y.W., X.L., G.L.)
| | - Lili Wu
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.).,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.)
| | - Yang Ye
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.).,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.)
| | - Yingchao Gong
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.).,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.)
| | - Xia Zhao
- School of Pharmaceutical Sciences, Hangzhou Medical College, Zhejiang, China (S.Y., X.H., W.L., X.Z., G.L.)
| | - Weijian Huang
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China (S.Y., W.H.)
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang, China (W.L., Y.W., X.L., G.L.)
| | - Xiaohong Long
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang, China (W.L., Y.W., X.L., G.L.)
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.).,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.)
| | - Guang Liang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Zhejiang, China (S.Y., X.H., W.L., X.Z., G.L.).,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang, China (W.L., Y.W., X.L., G.L.)
| |
Collapse
|
20
|
Chen XS, Cui JR, Meng XL, Wang SH, Wei W, Gao YL, Shou ST, Liu YC, Chai YF. Angiotensin-(1-7) ameliorates sepsis-induced cardiomyopathy by alleviating inflammatory response and mitochondrial damage through the NF-κB and MAPK pathways. J Transl Med 2023; 21:2. [PMID: 36593471 PMCID: PMC9807106 DOI: 10.1186/s12967-022-03842-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/20/2022] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND There is no available viable treatment for Sepsis-Induced Cardiomyopathy (SIC), a common sepsis complication with a higher fatality risk. The septic patients showed an abnormal activation of the renin angiotensin (Ang) aldosterone system (RAAS). However, it is not known how the Ang II and Ang-(1-7) affect SIC. METHODS Peripheral plasma was collected from the Healthy Control (HC) and septic patients and Ang II and Ang-(1-7) protein concentrations were measured. The in vitro and in vivo models of SIC were developed using Lipopolysaccharide (LPS) to preliminarily explore the relationship between the SIC state, Ang II, and Ang-(1-7) levels, along with the protective function of exogenous Ang-(1-7) on SIC. RESULTS Peripheral plasma Ang II and the Ang II/Ang-(1-7) levels in SIC-affected patients were elevated compared to the levels in HC and non-SIC patients, however, the HC showed higher Ang-(1-7) levels. Furthermore, peripheral plasma Ang II, Ang II/Ang-(1-7), and Ang-(1-7) levels in SIC patients were significantly correlated with the degree of myocardial injury. Additionally, exogenous Ang-(1-7) can attenuate inflammatory response, reduce oxidative stress, maintain mitochondrial dynamics homeostasis, and alleviate mitochondrial structural and functional damage by inhibiting nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways, thus alleviating SIC. CONCLUSIONS Plasma Ang-(1-7), Ang II, and Ang II/Ang-(1-7) levels were regarded as significant SIC biomarkers. In SIC, therapeutic targeting of RAAS, for example with Ang-(1-7), may exert protective roles against myocardial damage.
Collapse
Affiliation(s)
- Xin-Sen Chen
- grid.412645.00000 0004 1757 9434Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052 China
| | - Jing-Rui Cui
- grid.412645.00000 0004 1757 9434Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052 China
| | - Xiang-Long Meng
- grid.412645.00000 0004 1757 9434Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052 China
| | - Shu-Hang Wang
- grid.412645.00000 0004 1757 9434Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052 China
| | - Wei Wei
- grid.412645.00000 0004 1757 9434Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052 China
| | - Yu-Lei Gao
- grid.412645.00000 0004 1757 9434Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052 China
| | - Song-Tao Shou
- grid.412645.00000 0004 1757 9434Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052 China
| | - Yan-Cun Liu
- grid.412645.00000 0004 1757 9434Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052 China
| | - Yan-Fen Chai
- grid.412645.00000 0004 1757 9434Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052 China
| |
Collapse
|
21
|
Shen S, Wu G, Luo W, Li W, Li X, Dai C, Huang W, Liang G. Leonurine attenuates angiotensin II-induced cardiac injury and dysfunction via inhibiting MAPK and NF-κB pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 108:154519. [PMID: 36332391 DOI: 10.1016/j.phymed.2022.154519] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 10/06/2022] [Accepted: 10/20/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Hypertension is a common risk factor for heart failure, and excessive angiotensin II (Ang II) leads to hypertensive cardiac alterations such as hypertrophy, cardiac fibrosis, remodeling, and dysfunction. Leonurine is the major active alkaloid compound obtained from the traditional Chinese herbal medicine, Leonurus japonicus Houtt. The effects of leonurine on Ang II-induced hypertensive cardiac injury remain unknown. HYPOTHESIS/PURPOSE In the present study, we investigated the cardioprotective effects of leonurine in Ang II-infused mice and explored the underlying mechanisms in cardiomyocytes. METHODS Cardiac injury was induced by Ang II infusion in experimental mice with or without leonurine (at 10 or 20 mg/kg) treatment. H9c2 cells and neonatal rat primary cardiomyocytes were used to investigate the mechanisms through which leonurine exerts its protection effects. RESULTS The results showed that leonurine significantly alleviated Ang II-induced cardiac hypertrophy, fibrosis, and inflammation in both mice and cultured cardiomyocytes. Echocardiography revealed that leonurine preserved cardiac function in mice. Further investigations revealed that leonurine inhibited the activation of the mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB) pathways to reduce inflammatory response and injuries in Ang II-challenged cardiomyocytes. Inhibition of MAPKs and NF-κB in cardiomyocytes abolished the anti-inflammatory effects of leonurine. CONCLUSIONS Our study provides evidence that leonurine exerts protective effects against Ang II-induced hypertensive cardiac remodeling and dysfunction by inhibiting the MAPK and NF-κB pathways. Leonurine may be a promising agent for treating hypertensive heart failure.
Collapse
Affiliation(s)
- Siyuan Shen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Cardiology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Gaojun Wu
- Department of Cardiology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wu Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Cardiology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Weixin Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaobo Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chengyi Dai
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Cardiology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Weijian Huang
- Department of Cardiology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Cardiology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China.
| |
Collapse
|
22
|
DAMPs Released from Proinflammatory Macrophages Induce Inflammation in Cardiomyocytes via Activation of TLR4 and TNFR. Int J Mol Sci 2022; 23:ijms232415522. [PMID: 36555168 PMCID: PMC9778802 DOI: 10.3390/ijms232415522] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Cardiac dysfunction is a life-threatening complication in sepsis. Upon infection and cardiac stress, the cardiac macrophage population expands. Recruited macrophages exhibit a predominantly proinflammatory phenotype and release danger-associated molecular patterns (DAMPs) that contribute to cardiac dysfunction. However, the underlying pathomechanisms are highly complex and not fully understood. Here, we utilized an indirect macrophage-cardiomyocyte co-culture model to study the effects of proinflammatory macrophages on the activation of different cardiac receptors (TLR3, TLR4, and TNFR) and their role in cardiac inflammation and caspase-3/7 activation. The stimulation of cardiomyocytes with conditioned medium of LPS-stimulated macrophages resulted in elevated IL-6 protein concentrations and relative IL-6 and TNFα mRNA levels. Conditioned medium from LPS-stimulated macrophages also induced NFκB translocation and increased caspase-3/7 activation in cardiomyocytes. Analyzing the role of different cardiac receptors, we found that TLR4 and TNFR inhibition reduces cardiac inflammation and that the inhibition of TNFR prevents NFκB translocation into the nuclei of cardiomyocytes, induced by exposure to conditioned medium of proinflammatory macrophages. Moreover, we demonstrated that TLR3 inhibition reduces macrophage-mediated caspase-3/7 activation. Our results suggest that the immune response of macrophages under inflammatory conditions leads to the release of DAMPs, such as eRNA and cytokines, which in turn induce cardiomyocyte dysfunction. Thus, the data obtained in this study contribute to a better understanding of the pathophysiological mechanisms of cardiac dysfunction.
Collapse
|
23
|
Shen X, Tao J, Wang Z, Li G, Zhang Z, Li J, Diliar A. MiR-7015-3p Targets Nuclear Factor-Kappa-B-Inhibitor Alpha to Aggravate Hypoxia/Reoxygenation Injury in Cardiomyocytes Through the NF-κB Pathway. Int Heart J 2022; 63:881-892. [DOI: 10.1536/ihj.22-036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Xin Shen
- Department of Cardiology, People's Hospital of Xinjiang Uygur Autonomous Region
| | - Jing Tao
- Department of Cardiology, People's Hospital of Xinjiang Uygur Autonomous Region
| | - Zhao Wang
- Department of Cardiology, People's Hospital of Xinjiang Uygur Autonomous Region
| | - Guoqing Li
- Department of Cardiology, People's Hospital of Xinjiang Uygur Autonomous Region
| | - Zilong Zhang
- Department of Cardiology, People's Hospital of Xinjiang Uygur Autonomous Region
| | - Jie Li
- Department of Cardiology, People's Hospital of Xinjiang Uygur Autonomous Region
| | - Adri Diliar
- Department of Cardiology, People's Hospital of Xinjiang Uygur Autonomous Region
| |
Collapse
|
24
|
Chen XS, Wang SH, Liu CY, Gao YL, Meng XL, Wei W, Shou ST, Liu YC, Chai YF. Losartan attenuates sepsis-induced cardiomyopathy by regulating macrophage polarization via TLR4-mediated NF-κB and MAPK signaling. Pharmacol Res 2022; 185:106473. [PMID: 36182039 DOI: 10.1016/j.phrs.2022.106473] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/25/2022] [Accepted: 09/25/2022] [Indexed: 11/17/2022]
Abstract
Sepsis-induced cardiomyopathy (SIC) is a serious complication of sepsis with high mortality but no effective treatment. The renin angiotensin (Ang) aldosterone system (RAAS) is activated in patients with sepsis but it is unclear how the Ang II/Ang II type 1 receptor (AT1R) axis contributes to SIC. This study examined the link between the Ang II/AT1R axis and SIC as well as the protective effect of AT1R blockers (ARBs). The Ang II level in peripheral plasma and AT1R expression on monocytes were significantly higher in patients with SIC compared with those in non-SIC patients and healthy controls and were correlated with the degree of myocardial injury. The ARB losartan reduced the infiltration of neutrophils, monocytes, and macrophages into the heart and spleen of SIC mice. Additionally, losartan regulated macrophage polarization from the M1 to the M2 subtype via nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways, thereby maintaining the mitochondrial dynamics balance in cardiomyocytes and reducing oxidative stress and cardiomyocyte apoptosis. In conclusion, the plasma Ang II level and AT1R expression on plasma monocytes are an important biomarker in SIC. Therapeutic targeting of AT1R, for example with losartan, can potentially protect against myocardial injury in SIC.
Collapse
Affiliation(s)
- Xin-Sen Chen
- Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin 300052, China
| | - Shu-Hang Wang
- Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin 300052, China
| | - Chen-Yan Liu
- Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin 300052, China
| | - Yu-Lei Gao
- Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin 300052, China
| | - Xiang-Long Meng
- Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin 300052, China
| | - Wei Wei
- Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin 300052, China
| | - Song-Tao Shou
- Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin 300052, China
| | - Yan-Cun Liu
- Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin 300052, China.
| | - Yan-Fen Chai
- Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin 300052, China.
| |
Collapse
|
25
|
Shen XD, Zhang HS, Zhang R, Li J, Zhou ZG, Jin ZX, Wang YJ. Progress in the Clinical Assessment and Treatment of Myocardial Depression in Critically Ill Patient with Sepsis. J Inflamm Res 2022; 15:5483-5490. [PMID: 36164659 PMCID: PMC9508933 DOI: 10.2147/jir.s379905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/17/2022] [Indexed: 12/04/2022] Open
Abstract
Myocardial inhibition is the main cause of death in patients with sepsis.In recent years, methodological differences in the diagnosis, assessment, and treatment of septic myocardial depression have been observed, and how to objectively and accurately evaluate the degree of myocardial depression and the timing of treatment strategies have generally been the focus of this area of research. Based on the relevant research at home and abroad, the current review summarizes the clinical characteristics, methodological diagnosis, and symptomatic treatment of septic myocardial depression. The aim of doing so is to provide a reference for the early identification and treatment of patients with sepsis and myocardial depression.
Collapse
Affiliation(s)
- Xu-Dong Shen
- Department of Critical Care Medicine, Calmette Hospital Affiliated to Kunming Medical University, Kunming, 650011, People’s Republic of China
| | - Hua-Sheng Zhang
- Department of Critical Care Medicine, Calmette Hospital Affiliated to Kunming Medical University, Kunming, 650011, People’s Republic of China
| | - Rui Zhang
- Department of Critical Care Medicine, Calmette Hospital Affiliated to Kunming Medical University, Kunming, 650011, People’s Republic of China
| | - Jun Li
- Department of Critical Care Medicine, Calmette Hospital Affiliated to Kunming Medical University, Kunming, 650011, People’s Republic of China
| | - Zhi-Gang Zhou
- Department of Critical Care Medicine, Calmette Hospital Affiliated to Kunming Medical University, Kunming, 650011, People’s Republic of China
| | - Zhi-Xian Jin
- Department of Respiratory Medicine, Calmette Hospital Affiliated to Kunming Medical University, Kunming, 650011, People’s Republic of China
| | - Yin-Jia Wang
- Department of Critical Care Medicine, Calmette Hospital Affiliated to Kunming Medical University, Kunming, 650011, People’s Republic of China
| |
Collapse
|
26
|
Csató G, Erdei N, Ványai B, Balla T, Czuriga D, Csanádi Z, Koszegi Z, Édes I, Szabó GT. Predictors of restenosis following percutaneous coronary stent implantation: The role of trimetazidine therapy. Front Cardiovasc Med 2022; 9:873899. [PMID: 35935652 PMCID: PMC9353214 DOI: 10.3389/fcvm.2022.873899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 07/04/2022] [Indexed: 11/23/2022] Open
Abstract
Aims In-stent restenosis (ISR) is an unresolved problem following percutaneous coronary intervention (PCI), having a negative impact on clinical outcome. The main goal of this study was to find new independent predictors that may influence the development of ISR. Methods and results In this retrospective analysis, 653 PCI patients were involved. All patients had coronary stent implantation and a follow-up coronary angiography. Based on the presence of ISR at follow-up, patients were divided into two groups: 221 in the ISR and 432 in the control group. When evaluating the medical therapy of patients, significantly more patients were on trimetazidine (TMZ) in the control compared to the ISR group (p = 0.039). TMZ was found to be an independent predictor of a lower degree of ISR development (p = 0.007). TMZ treatment was especially effective in bare metal stent (BMS)-implanted chronic coronary syndrome (CCS) patients with narrow coronary arteries. The inflammation marker neutrophil to lymphocyte ratio (NLR) was significantly elevated at baseline in the ISR group compared to controls. The reduction of post-PCI NLR was associated with improved efficacy of TMZ to prevent ISR development. Drug eluting stent implantation (p < 0.001) and increased stent diameter (p < 0.001) were the most important independent predictors of a lower degree of ISR development, while the use of longer stents (p = 0.005) was a major independent predictor of an increased ISR risk. Conclusion TMZ reduces the occurrence of ISR following PCI, with special effectiveness in BMS-implanted patients having CCS and narrow coronary arteries. TMZ treatment may help to lower ISR formation in countries with high BMS utilization rates.
Collapse
Affiliation(s)
- Gábor Csató
- Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Nóra Erdei
- Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Beatrix Ványai
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tímea Balla
- Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dániel Czuriga
- Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Csanádi
- Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Koszegi
- Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - István Édes
- Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Tamás Szabó
- Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- *Correspondence: Gábor Tamás Szabó
| |
Collapse
|
27
|
Cao P, Chen Q, Shi C, Wang L, Gong Z. Fusobacterium nucleatum promotes the development of acute liver failure by inhibiting the NAD + salvage metabolic pathway. Gut Pathog 2022; 14:29. [PMID: 35765030 PMCID: PMC9238040 DOI: 10.1186/s13099-022-00503-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 06/01/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Acute liver failure (ALF) patients are often accompanied by severe energy metabolism abnormalities and intestinal microecological imbalance. The intestinal mucosal barrier is severely damaged. Intestinal endotoxin can induce intestinal endotoxemia through the "Gut-Liver axis". More and more evidence shows that members of the gut microbiota, especially Fusobacterium nucleatum (F. nucleatum), are related to inflammatory bowel disease, but whether F. nucleatum is involved in the development of ALF and whether it affects the liver energy metabolism is unclear. METHODS This study first detected the abundance of F. nucleatum and its effect on ALF disease, and explored whether F. nucleatum aggravated liver inflammation in vitro and in vivo. RESULTS Our data showed that liver tissues of ALF patients contained different abundances of F. nucleatum, which were related to the degree of liver inflammation. In addition, we found that F. nucleatum infection affected the energy metabolism of the liver during the development of ALF, inhibited the synthesis pathway of nicotinamide adenine dinucleotide (NAD+)'s salvage metabolism, and promoted inflammatory damage in the liver. In terms of mechanism, F. nucleatum inhibited NAD+ and the NAD+-dependent SIRT1/AMPK signaling pathway, and promoted liver damage of ALF. CONCLUSIONS Fusobacterium nucleatum coordinates a molecular network including NAD+ and SIRT1 to control the progress of ALF. Detection and targeting of F. nucleatum and its related pathways may provide valuable insights for the treatment of ALF.
Collapse
Affiliation(s)
- Pan Cao
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 238 Jie Fang Road, Wuhan, 430060, People's Republic of China
| | - Qian Chen
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 238 Jie Fang Road, Wuhan, 430060, People's Republic of China
| | - Chunxia Shi
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 238 Jie Fang Road, Wuhan, 430060, People's Republic of China
| | - Luwen Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 238 Jie Fang Road, Wuhan, 430060, People's Republic of China
| | - Zuojiong Gong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 238 Jie Fang Road, Wuhan, 430060, People's Republic of China.
| |
Collapse
|
28
|
Ozturk A, Topcu A, Deniz E, Duman Ozturk S, Arpa M, Kutlu Yilmaz E. The protective effects of trimetazidine against ovary ischemia-reperfusion injury via the TLR4/Nf-kB signal pathway. J Biochem Mol Toxicol 2022; 36:e23114. [PMID: 35633067 DOI: 10.1002/jbt.23114] [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: 01/25/2022] [Revised: 04/12/2022] [Accepted: 05/18/2022] [Indexed: 11/12/2022]
Abstract
Late diagnosis and treatment of ovarian ischemia can lead to worsening of ischemia, irreversible damage to ovarian functions and infertility. In this process, there is no approved medical treatment that can reduce the negative effects of ischemia and contribute positively to ovarian functions during reperfusion after detorsion. Rats were randomly assigned into one of six groups of eight animals each. The groups were designed as follows: The control group, The ischemia(I) group, The Ischemia + Trimetazidine (I + TMZ) (20 mg/kg) group, and The ischemia-reperfusion group (I/R). The Ischemia-Reperfusion + Trimetazidine (I/R + TMZ) (20 mg/kg) group, and The Sham + Trimetazidine (Sham + TMZ) (20 mg/kg) group. In this study performed thiobarbituric acid reactive substances (TBARS), total thiol (-SH), interleukin 1 beta (IL-1β), interleukin 6 (IL-6), toll-like receptor 4 (TLR4), and nuclear factor-kappa B(NF-κβ). Increased oxidative stress and inflammation were as a result of ovarian I and I/R application. Trimetazidine (TMZ), was sufficient to reduce the oxidative stress and inflammation. TLR4 and NF-κβ, which were upregulated by oxidative stress and inflammation, were regressed by TMZ. TMZ should be considered as a potential therapeutic agent in addition to surgery in the clinical treatment of ovarian torsion.
Collapse
Affiliation(s)
- Aykut Ozturk
- Department of Pharmacology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Atilla Topcu
- Department of Pharmacology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Esra Deniz
- Department of Pharmacology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Seda Duman Ozturk
- Department of Pathology, Recep Tayyip Erdogan University Education and Research Hospital, Rize, Turkey
| | - Medeni Arpa
- Department of Medical Biochemistry, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Eda Kutlu Yilmaz
- Department of Medical Biochemistry, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| |
Collapse
|
29
|
Engin S, Barut EN, Yaşar YK, Soysal AÇ, Arıcı T, Kerimoğlu G, Kadıoğlu M, Sezen SF. Trimetazidine attenuates cyclophosphamide-induced cystitis by inhibiting TLR4-mediated NFκB signaling in mice. Life Sci 2022; 301:120590. [PMID: 35504331 DOI: 10.1016/j.lfs.2022.120590] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 04/19/2022] [Accepted: 04/25/2022] [Indexed: 11/26/2022]
Abstract
AIM Cyclophosphamide (CP)-induced cystitis is a challenging clinical problem involving inflammation and dysfunction of bladder. Trimetazidine (TMZ) is an anti-anginal drug with anti-oxidant and anti-inflammatory properties. We aimed to investigate the protective effects of TMZ in CP-induced cystitis via inhibiting TLR4/NFκB signaling. MAIN METHODS Balb/c mice were administrated TMZ (10 or 20 mg/kg/day) intraperitoneally (i.p.) for 5 consecutive days before CP. On day 6, cystitis was induced by a single dose of CP (300 mg/kg, i.p.). Mesna (2-mercaptoethane sulfonate sodium; 30 mg/kg, i.p.) was administered 20 min before and at 4 and 8 h after the CP injection. After 24 h of cystitis induction, the bladders were removed for histopathological evaluation, contractility studies, biochemical analysis and western blotting. MTT assay was performed in a cancer cell line (MDA-MB-231) to evaluate the effect of TMZ on the cytotoxicity of CP. KEY FINDINGS CP-induced severe cystitis was confirmed by histological disturbances and the decrease in carbachol-evoked contractions of detrusor strips, which was partially improved by TMZ (20 mg/kg/day). SOD activity and GSH content were decreased whereas TNF-α and IL-1β levels were increased in the bladders of CP-treated mice, which were restored by TMZ or mesna. TMZ reduced the CP-induced increase in the protein expressions of caspase-3, TLR4 and phosphorylated-NFκB in bladder tissues. TMZ alone decreased the cell viability and TMZ also enhanced the cytotoxicity of CP. SIGNIFICANCE Our study provides the first preclinical evidence that TMZ attenuates CP-induced urotoxicity by enhancing anti-oxidant capacity and suppressing inflammation possibly via downregulating TLR4-mediated NFκB signaling while augmenting the cytotoxicity of CP.
Collapse
Affiliation(s)
- Seçkin Engin
- Department of Pharmacology, Faculty of Pharmacy, Karadeniz Technical University, Trabzon, Turkiye.
| | - Elif Nur Barut
- Department of Pharmacology, Faculty of Pharmacy, Karadeniz Technical University, Trabzon, Turkiye
| | - Yeşim Kaya Yaşar
- Department of Pharmacology, Faculty of Pharmacy, Karadeniz Technical University, Trabzon, Turkiye; Drug and Pharmaceutical Technology Application and Research Center, Karadeniz Technical University, Trabzon, Turkiye
| | - Aysun Çelik Soysal
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Bülent Ecevit University, Zonguldak, Turkiye
| | - Tuğba Arıcı
- Başaksehir Cam and Sakura City Hospital, İstanbul, Turkiye
| | - Gökçen Kerimoğlu
- Department of Histology and Embryology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkiye
| | - Mine Kadıoğlu
- Department of Medical Pharmacology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkiye
| | - Sena F Sezen
- Department of Pharmacology, Faculty of Pharmacy, Karadeniz Technical University, Trabzon, Turkiye; Drug and Pharmaceutical Technology Application and Research Center, Karadeniz Technical University, Trabzon, Turkiye
| |
Collapse
|
30
|
Disulfiram inhibits oxidative stress and NLRP3 inflammasome activation to prevent LPS-induced cardiac injury. Int Immunopharmacol 2022; 105:108545. [DOI: 10.1016/j.intimp.2022.108545] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 12/18/2022]
|
31
|
Tang X, Li X, Zhang D, Han W. Astragaloside-IV alleviates high glucose-induced ferroptosis in retinal pigment epithelial cells by disrupting the expression of miR-138-5p/Sirt1/Nrf2. Bioengineered 2022; 13:8240-8254. [PMID: 35302431 PMCID: PMC9162003 DOI: 10.1080/21655979.2022.2049471] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Astragaloside-IV (AS-IV) (C41H68O14) is a high-purity natural product extracted from Astragalus, which has demonstrated biological activities. However, the effect of AS-IV on retinal pigment epithelial (RPE) cells in diabetic retinopathy (DR) remains unclear. In this study, high glucose (HG) was shown to promote ARPE-19 RPE cell death, increase the contents of reactive oxygen species (ROS) and oxidized glutathione (GSSG), and enhance lipid peroxidation density of mitochondrial membrane. In contrast, AS-IV decreased glutathione (GSH) content, mitochondria size and ridge. Addition of iron death inhibitor Ferrostatin-1 (Fer-1) to RPE cells decreased cell dead rate, thus indicating that HG-induced mitochondrial damage occurred due to ferroptosis. AS-IV alleviated HG-induced RPE cell damage. Furthermore, HG decreased levels of silent information regulator 1 (Sirt1) and nuclear factor (erythroid-derived 2)-like 2 (Nrf2) in the nucleus of RPE cells; AS-IV could alleviate these effects and increased expression of glutathione peroxidase 4 (GPX4), glutamate cysteine ligase (GCLM) and glutamate cysteine ligase catalytic subunit (GCLC), which are Nrf2 downstream genes. Mechanistically, AS-IV was shown to alleviate the effects of HG by increasing mir-138-5p expression in RPE cells and promoting expression of Sirt1 and Nrf2 in the nucleus. Transfection of mir-138-5p agonist inhibited the regulatory effects of AS-IV on Sirt1 and Nrf2, accompanied by decreased GPX4, GCLM and GCLC levels, and restoration of ferroptosis-related changes. Collectively, HG increased ferroptosis rate in RPE cells. In addition, AS-IV inhibited miR-138-5p expression, subsequently increasing Sirt1/Nrf2 activity and cellular antioxidant capacity to alleviate ferroptosis, resulting decreased cell death, which potentially inhibits the DR pathological process.
Collapse
Affiliation(s)
- Xuyuan Tang
- Department of Ophthalmology, the First Affiliated Hospital of Medical College of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiuyi Li
- Department of Ophthalmology, the First Affiliated Hospital of Medical College of Zhejiang University, Hangzhou, Zhejiang, China
| | - Dongyan Zhang
- Department of Ophthalmology, the First Affiliated Hospital of Medical College of Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Han
- Department of Ophthalmology, the Second Affiliated Hospital of Medical College of Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
32
|
BRETSCHNEIDER SOLUTION AND TWO ANTIANGINAL DRUGS PROTECT PERIPHERAL TISSUE IN AN ANIMAL MODEL OF HEMORRHAGIC SHOCK. J Cardiovasc Pharmacol 2022; 79:896-903. [PMID: 35249963 DOI: 10.1097/fjc.0000000000001253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 02/01/2022] [Indexed: 11/26/2022]
|
33
|
Dai S, Ye B, Zhong L, Chen Y, Hong G, Zhao G, Su L, Lu Z. GSDMD Mediates LPS-Induced Septic Myocardial Dysfunction by Regulating ROS-dependent NLRP3 Inflammasome Activation. Front Cell Dev Biol 2021; 9:779432. [PMID: 34820388 PMCID: PMC8606561 DOI: 10.3389/fcell.2021.779432] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 10/22/2021] [Indexed: 01/06/2023] Open
Abstract
Myocardial dysfunction is a serious consequence of sepsis and contributes to high mortality. Currently, the molecular mechanism of myocardial dysfunction induced by sepsis remains unclear. In the present study, we investigated the role of gasdermin D (GSDMD) in cardiac dysfunction in septic mice and the underlying mechanism. C57BL/6 wild-type (WT) mice and age-matched Gsdmd-knockout (Gsdmd -/-) mice were intraperitoneally injected with lipopolysaccharide (LPS) (10 mg/kg) to mimic sepsis. The results showed that GSDMD-NT, the functional fragment of GSDMD, was upregulated in the heart tissue of septic WT mice induced by LPS, which was accompanied by decreased cardiac function and myocardial injury, as shown by decreased ejection fraction (EF) and fractional shortening (FS) and increased cardiac troponin I (cTnI), creatine kinase isoenzymes MB (CK-MB), and lactate dehydrogenase (LDH). Gsdmd -/- mice exhibited protection against LPS-induced myocardial dysfunction and had a higher survival rate. Gsdmd deficiency attenuated LPS-induced myocardial injury and cell death. Gsdmd deficiency prevented LPS-induced the increase of interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) in serum, as well as IL-1β and TNF-α mRNA levels in myocardium. In addition, LPS-mediated inflammatory cell infiltration into the myocardium was ameliorated and activation of NF-κB signaling pathway and the NOD-like receptor protein 3 (NLPR3) inflammasome were suppressed in Gsdmd -/- mice. Further research showed that in the myocardium of LPS-induced septic mice, GSDMD-NT enrichment in mitochondria led to mitochondrial dysfunction and reactive oxygen species (ROS) overproduction, which further regulated the activation of the NLRP3 inflammasome. In summary, our data suggest that GSDMD plays a vital role in the pathophysiology of LPS-induced myocardial dysfunction and may be a crucial target for the prevention and treatment of sepsis-induced myocardial dysfunction.
Collapse
Affiliation(s)
- Shanshan Dai
- The Key Laboratory of Emergency and Disaster Medicine of Wenzhou, Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bozhi Ye
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lingfeng Zhong
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yanghao Chen
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guangliang Hong
- The Key Laboratory of Emergency and Disaster Medicine of Wenzhou, Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guangju Zhao
- The Key Laboratory of Emergency and Disaster Medicine of Wenzhou, Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lan Su
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhongqiu Lu
- The Key Laboratory of Emergency and Disaster Medicine of Wenzhou, Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
34
|
Xue J, Liu J, Xu B, Yu J, Zhang A, Qin L, Liu C, Yang Y. miR-21-5p inhibits inflammation injuries in LPS-treated H9c2 cells by regulating PDCD4. Am J Transl Res 2021; 13:11450-11460. [PMID: 34786071 PMCID: PMC8581922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/31/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVES To explore the expression levels and the potential regulatory mechanism of miR-21-5p in LPS-treated H9c2 cells. METHODS The secretions of the inflammatory cytokines induced by LPS in H9c2 cells were evaluated using ELISA. We used RT-RCR and western blot to measure the relative mRNA and protein expression levels in LPS-treated H9c2 cells. CCK-8 and EdU assays showed the viability and proliferation profiles of the H9c2 cells. TUNEL assays demonstrated the apoptotic behaviors of the H9c2 cells, and a luciferase reporter analysis was used to investigate the interactions between miR-21-5p and programmed cell death protein 4 (PDCD4). RESULTS LPS induced damage to the H9c2 cells by reducing the cell viability and down-regulating miR-21-5p. On the other hand, miR-21-5p overexpression inhibited the LPS-induced inflammatory damage in the H9c2 cells. Moreover, PDCD4 was verified as a downstream target gene of miR-21-5p, and its expression was inhibited by the higher miR-21-5p content. Finally, miR-21-5p inhibited septic processes, and the PDCD4 overexpression rescued the miR-21-5p effect in the LPS-treated H9c2 cells. CONCLUSION Our findings suggest that miR-21-5p inhibits the LPS-induced progression of sepsis in H9c2 cells. Additionally, PDCD4 is a downstream target gene of miR-21-5p, and both molecules serve as potential therapeutic targets for heart sepsis patients.
Collapse
Affiliation(s)
- Jianhua Xue
- Department of Trauma Center, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Jiajia Liu
- Department of Trauma Center, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Bo Xu
- Department of Orthopaedics, Qidong Hospital of Traditional Chinese MedicineNantong, Jiangsu Province, China
| | - Junbo Yu
- Department of Trauma Center, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Aixian Zhang
- Department of General Practice Medicine, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Lili Qin
- Department of Endoscopic Center, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Chun Liu
- Department of Emergency Medicine, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Yang Yang
- Department of Trauma Center, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| |
Collapse
|
35
|
Molecular mechanism and therapeutic targeting of necrosis, apoptosis, pyroptosis, and autophagy in cardiovascular disease. Chin Med J (Engl) 2021; 134:2647-2655. [PMID: 34608069 PMCID: PMC8631411 DOI: 10.1097/cm9.0000000000001772] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
ABSTRACT Cell death occurs in various tissues and organs in the body. It is a physiological or pathological process that has different effects. It is of great significance in maintaining the morphological function of cells and clearing abnormal cells. Pyroptosis, apoptosis, and necrosis are all modes of cell death that have been studied extensively by many experts and scholars, including studies on their effects on the liver, kidney, the heart, other organs, and even the whole body. The heart, as the most important organ of the body, should be a particular focus. This review summarizes the mechanisms underlying the various cell death modes and the relationship between the various mechanisms and heart diseases. The current research status for heart therapy is discussed from the perspective of pathogenesis.
Collapse
|
36
|
Han X, Ding C, Sang X, Peng M, Yang Q, Ning Y, Lv Q, Shan Q, Hao M, Wang K, Wu X, Zhang H, Cao G. Targeting Sirtuin1 to treat aging-related tissue fibrosis: From prevention to therapy. Pharmacol Ther 2021; 229:107983. [PMID: 34480962 DOI: 10.1016/j.pharmthera.2021.107983] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 08/18/2021] [Accepted: 08/18/2021] [Indexed: 12/19/2022]
Abstract
Fibrosis, which is characterized by excessive extracellular matrix (ECM) deposition, is a wound-healing response to organ injury and may promote cancer and failure in various organs, such as the heart, liver, lung, and kidney. Aging associated with oxidative stress and inflammation exacerbates cellular dysfunction, tissue failure, and body function disorders, all of which are closely related to fibrosis. Sirtuin-1 (SIRT1) is a class III histone deacetylase that regulates growth, transcription, aging, and metabolism in various organs. This protein is downregulated in organ injury and fibrosis associated with aging. Its expression and distribution change with age in different organs and play critical roles in tissue oxidative stress and inflammation. This review first described the background on fibrosis and regulatory functions of SIRT1. Second, we summarized the relationships of SIRT1 with other proteins and its protective action during fibrosis in the heart, liver, lung and kidney. Third, the activation of SIRT1 in therapies of tissue fibrosis, especially in liver fibrosis and aging-related tissue injury, was analyzed. In conclusion, SIRT1 targeting may be a new therapeutic strategy in fibrosis.
Collapse
Affiliation(s)
- Xin Han
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chuan Ding
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - XiaNan Sang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - MengYun Peng
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiao Yang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Ning
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiang Lv
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - QiYuan Shan
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Min Hao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - KuiLong Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xin Wu
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hongyan Zhang
- Cancer Hospital of The University of Chinese Academy of Sciences, Hangzhou, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
37
|
Luo YY, Yang ZQ, Lin XF, Zhao FL, Tu HT, Wang LJ, Wen MY, Xian SX. Knockdown of lncRNA PVT1 attenuated macrophage M1 polarization and relieved sepsis induced myocardial injury via miR-29a/HMGB1 axis. Cytokine 2021; 143:155509. [PMID: 33840587 DOI: 10.1016/j.cyto.2021.155509] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/02/2021] [Accepted: 03/19/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND LncRNA PVT1 was reported to be elevated in septic myocardial tissue. The underlying mechanism by which PVT1 aggravated sepsis induced myocardial injury needs further investigation. METHODS Mice was subjected to LPS injection to mimic in vivo sepsis model. HE staining was applied to observe tissue injury. Cardiac function of mice was determined by echocardiography. Bone marrow derived macrophage (BMDM) was used to confirm the regulatory effect of PVT1 in macrophage polarization. Western blotting or qRT-PCR were performed to evaluate protein or mRNA levels, respectively. ELISA was conducted to determine cytokine levels. Interaction between PVT1 and miR-29a, miR-29a and HMGB1 were accessed by dual luciferase assay. RESULTS Expression of PVT1 was elevated in myocardial tissue and heart infiltrating macrophages of sepsis mice. PVT1 knockdown alleviated LPS induced myocardial injury and attenuated M1 macrophage polarization. The mechanic study suggested that PVT1 targeted miR-29a, thus elevated expression of HMGB1, which was repressed by miR-29a targeting. The effect of PVT1 on M1 macrophage polarization was dependent on targeting miR-29a. CONCLUSION PVT1 promoted M1 polarization and aggravated LPS induced myocardial injury via miR-29a/HMGB1 axis.
Collapse
Affiliation(s)
- Yuan-Yuan Luo
- Department of Intensive Care Unit, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong Province, PR China
| | - Zhong-Qi Yang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong Province, PR China
| | - Xin-Feng Lin
- Department of Intensive Care Unit, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong Province, PR China
| | - Feng-Li Zhao
- Department of Intensive Care Unit, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong Province, PR China
| | - Hai-Tao Tu
- Department of Nephrology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong Province, PR China
| | - Ling-Jun Wang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong Province, PR China.
| | - Min-Yong Wen
- Department of Intensive Care Unit, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong Province, PR China.
| | - Shao-Xiang Xian
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong Province, PR China.
| |
Collapse
|
38
|
Ye S, Lin K, Wu G, Xu MJ, Shan P, Huang W, Wang Y, Liang G. Toll-like receptor 2 signaling deficiency in cardiac cells ameliorates Ang II-induced cardiac inflammation and remodeling. Transl Res 2021; 233:62-76. [PMID: 33652137 DOI: 10.1016/j.trsl.2021.02.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/05/2021] [Accepted: 02/23/2021] [Indexed: 11/28/2022]
Abstract
Activation of the innate immune system represents a vital step in inflammation during cardiac remodeling induced by the angiotensin II (Ang II). This study aimed to explore the role of Toll-like receptors 2 (TLR2) in Ang II-induced cardiac remodeling. We investigated the effect of TLR2 deficiency on Ang II-induced cardiac remodeling by utilizing TLR2 knockout mice, bone marrow transplantation models, and H9C2 cells. Though TLR2 deficiency had no effect on body weight change, cardiac Ang II content and blood pressure, it significantly ameliorated cardiac hypertrophy, fibrosis and inflammation, as well as improved heart function. Further bone marrow transplantation studies showed that TLR2-deficiency in cardiac cells but not bone marrow-derived cells prevented Ang II-induced cardiac remodeling and cardiac dysfunction. The underlying mechanism may involve increased TLR2-MyD88 interaction. Further in vitro studies in Ang II-treated H9C2 cells showed that TLR2 knockdown by siRNA significantly decreased Ang II-induced cell hypertrophy, fibrosis and inflammation. Moreover, Ang II significantly increased TLR2-MyD88 interaction in H9C2 cells in a TLR4-independent manner. TLR2 deficiency in cardiac cells prevents Ang II-induced cardiac remodeling, inflammation and dysfunction through reducing the formation of TLR2-MyD88 complexes. Inhibition of TLR2 pathway may be a therapeutic strategy of hypertensive heart failure.
Collapse
Affiliation(s)
- Shiju Ye
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ke Lin
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gaojun Wu
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ming-Jiang Xu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Peiren Shan
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weijian Huang
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhuji Biomedicine Institute, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhuji, Zhejiang, China.
| | - Guang Liang
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhuji Biomedicine Institute, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhuji, Zhejiang, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
39
|
Xie L, Wu Y, Zhou C, Tan Z, Xu H, Chen G, Chen H, Huang G, Fan H, Gao L, Liu B, Zhou Y. Piceatannol protects against sepsis-induced myocardial dysfunction via direct inhibition of JAK2. Int Immunopharmacol 2021; 96:107639. [PMID: 34162128 DOI: 10.1016/j.intimp.2021.107639] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 03/24/2021] [Accepted: 03/31/2021] [Indexed: 11/26/2022]
Abstract
Sepsis-induced myocardial dysfunction (SIMD) represents one of the serious complications secondary to sepsis, which is a leading cause of the high mortality rate among septic cases. Subsequent cardiomyocyte apoptosis, together with the uncontrolled inflammatory response, has been suggested to be closely related to SIMD. Piceatannol (PIC) is verified with potent anti-apoptotic and anti-inflammatory effects, but its function and molecular mechanism in SIMD remain unknown so far. This study aimed to explore the potential role and mechanism of action of PIC in resisting SIMD. The interaction of PIC with JAK2 proteins was evaluated by molecular docking, molecular dynamics (MD) simulation and surface plasmon resonance imaging (SPRi). The cecal ligation and puncture-induced septicemia mice and the LPS-stimulated H9C2 cardiomyocytes were prepared as the models in vivo and in vitro, separately. Molecular docking showed that JAK2-PIC complex had the -8.279 kcal/mol binding energy. MD simulations showed that JAK2-PIC binding was stable. SPRi analysis also showed that PIC has a strong binding affinity to JAK2. PIC treatment significantly ameliorated the cardiac function, attenuated the sepsis-induced myocardial loss, and suppressed the myocardial inflammatory responses both in vivo and in vitro. Further detection revealed that PIC inhibited the activation of the JAK2/STAT3 signaling, which was tightly associated with apoptosis and inflammation. Importantly, pre-incubation with a JAK2 inhibitor (AG490) partially blocked the cardioprotective effects of PIC. Collectively, the findings demonstrated that PIC restored the impaired cardiac function by attenuating the sepsis-induced apoptosis and inflammation via suppressing the JAK2/STAT3 pathway both in septic mice and H9C2 cardiomyocytes.
Collapse
Affiliation(s)
- Lingpeng Xie
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Yuting Wu
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China; Department of Traditional Chinese Medicine, Binzhou Medical University Hospital, Binzhou 256603, China
| | - Chuying Zhou
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Zhangbin Tan
- Department of Traditional Chinese Medicine (Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, State Key Laboratory of Respiratory Disease), the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510260, China
| | - Honglin Xu
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Guanghong Chen
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Hongmei Chen
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Guiqiong Huang
- Department of Internal Medicine, Huizhou Hospital of Guangzhou University of Traditional Chinese Medicine, Huizhou 516000, China
| | - Huijie Fan
- TCM Health Construction Department of Yangjiang People's Hospital, Guangdong Province, Yangjiang 529500, China
| | - Lei Gao
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China.
| | - Bin Liu
- Department of Traditional Chinese Medicine (Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, State Key Laboratory of Respiratory Disease), the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510260, China.
| | - Yingchun Zhou
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
40
|
Xiao W, Oldham WM, Priolo C, Pandey AK, Loscalzo J. Immunometabolic Endothelial Phenotypes: Integrating Inflammation and Glucose Metabolism. Circ Res 2021; 129:9-29. [PMID: 33890812 PMCID: PMC8221540 DOI: 10.1161/circresaha.120.318805] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 12/20/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Wusheng Xiao
- Division of Cardiovascular Medicine (W.X., A.K.P., J.L.), Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - William M. Oldham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine (W.M.O., C.P.), Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Carmen Priolo
- Division of Pulmonary and Critical Care Medicine, Department of Medicine (W.M.O., C.P.), Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Arvind K. Pandey
- Division of Cardiovascular Medicine (W.X., A.K.P., J.L.), Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine (W.X., A.K.P., J.L.), Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
41
|
Luo XY, Zhong Z, Chong AG, Zhang WW, Wu XD. Function and Mechanism of Trimetazidine in Myocardial Infarction-Induced Myocardial Energy Metabolism Disorder Through the SIRT1-AMPK Pathway. Front Physiol 2021; 12:645041. [PMID: 34220528 PMCID: PMC8248253 DOI: 10.3389/fphys.2021.645041] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/14/2021] [Indexed: 02/04/2023] Open
Abstract
Myocardial energy metabolism (MEM) is an important factor of myocardial injury. Trimetazidine (TMZ) provides protection against myocardial ischemia/reperfusion injury. The current study set out to evaluate the effect and mechanism of TMZ on MEM disorder induced by myocardial infarction (MI). Firstly, a MI mouse model was established by coronary artery ligation, which was then treated with different concentrations of TMZ (5, 10, and 20 mg kg-1 day-1). The results suggested that TMZ reduced the heart/weight ratio in a concentration-dependent manner. TMZ also reduced the levels of Bax and cleaved caspase-3 and promoted Bcl-2 expression. In addition, TMZ augmented adenosine triphosphate (ATP) production and superoxide dismutase (SOD) activity induced by MI and decreased the levels of lipid peroxide (LPO), free fatty acids (FFA), and nitric oxide (NO) in a concentration-dependent manner (all P < 0.05). Furthermore, an H2O2-induced cell injury model was established and treated with different concentrations of TMZ (1, 5, and 10 μM). The results showed that SIRT1 overexpression promoted ATP production and reactive oxygen species (ROS) activity and reduced the levels of LPO, FFA, and NO in H9C2 cardiomyocytes treated with H2O2 and TMZ. Silencing SIRT1 suppressed ATP production and ROS activity and increased the levels of LPO, FFA, and NO (all P < 0.05). TMZ activated the SIRT1-AMPK pathway by increasing SIRT1 expression and AMPK phosphorylation. In conclusion, TMZ inhibited MI-induced myocardial apoptosis and MEM disorder by activating the SIRT1-AMPK pathway.
Collapse
Affiliation(s)
- Xiu-Ying Luo
- Department of Cardiology, The Second Affiliated Hospital (Jiande Branch), Zhejiang University School of Medicine, Hangzhou, China
| | - Ze Zhong
- Department of Cardiology, The Second Affiliated Hospital (Jiande Branch), Zhejiang University School of Medicine, Hangzhou, China
| | - Ai-Guo Chong
- Department of Cardiology, The Second Affiliated Hospital (Jiande Branch), Zhejiang University School of Medicine, Hangzhou, China
| | - Wei-Wei Zhang
- Department of Cardiology, The Second Affiliated Hospital (Jiande Branch), Zhejiang University School of Medicine, Hangzhou, China
| | - Xin-Dong Wu
- Department of Cardiology, The Second Affiliated Hospital (Jiande Branch), Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
42
|
Eid BG, El-Shitany NAEA, Neamatallah T. Trimetazidine improved adriamycin-induced cardiomyopathy by downregulating TNF-α, BAX, and VEGF immunoexpression via an antioxidant mechanism. ENVIRONMENTAL TOXICOLOGY 2021; 36:1217-1225. [PMID: 33704910 DOI: 10.1002/tox.23120] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 02/19/2021] [Indexed: 06/12/2023]
Abstract
Few studies have reported a prophylactic effect of the anti-ischemic trimetazidine (TRI) against cardiac toxicity caused by adriamycin (ADR). However, the mechanism of action of TRI remained incomplete. The cardioprotective mechanism(s) of TRI against ADR-induced cardiotoxicity was investigated in this study. Cardiotoxicity was induced in three groups of Wistar rats by injecting a single dose of ADR (10 mg/kg, i.p.). TRI was administered in two doses regimen, low (L) (2.5 mg/kg, i.p.) and high (H) (10 mg/kg, i.p.). The results of the study showed that both TRI L and H doses improved cardiac enzymes and pathology, while only the TRI H dose improved the electrocardiogram. Both TRI L and H doses decreased malondialdehyde and increased reduced glutathione and superoxide dismutase. Only TRI H dose increased glutathione peroxidase and catalase. Both TRI L and H doses decreased interleukin-1 beta and tumor necrosis factor-alpha (TNF-α). Both TRI L and H doses downregulated TNF-α, BAX, and vascular endothelial growth factor cardiac protein expression. The data obtained in this study provided evidence that TRI opposed ADR-induced cardiotoxicity. The mechanism could be due to improved antioxidant levels as well as inhibition of inflammation and programmed cell death.
Collapse
Affiliation(s)
- Basma G Eid
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nagla Abd El-Aziz El-Shitany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Thikryat Neamatallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
43
|
Hohensinner PJ, Lenz M, Haider P, Mayer J, Richter M, Kaun C, Goederle L, Brekalo M, Salzmann M, Sharma S, Fischer MB, Stojkovic S, Ramsmayer D, Hengstenberg C, Podesser BK, Huber K, Binder CJ, Wojta J, Speidl WS. Pharmacological inhibition of fatty acid oxidation reduces atherosclerosis progression by suppression of macrophage NLRP3 inflammasome activation. Biochem Pharmacol 2021; 190:114634. [PMID: 34058186 DOI: 10.1016/j.bcp.2021.114634] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Inflammation is a key process during atherosclerotic lesion development and propagation. Recent evidence showed clearly that especially the inhibition of interleukin (IL)-1β reduced atherosclerotic adverse events in human patients. Fatty acid oxidation (FAO) was previously demonstrated to interact with the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) pathway which is required for mature IL-1β secretion. To understand possible anti-inflammatory properties of FAO inhibition, we tested the effect of pharmacological FAO inhibition using the inhibitor for long-chain 3-ketoacyl coenzyme A thiolase trimetazidine on atherosclerotic plaque development and inflammation. EXPERIMENTAL APPROACH The effect of FAO inhibition was determined in LDL-R-/- male mice on a C57/BL6 background. In vitro effects of trimetazidine treatment were analyzed in human umbilical vein endothelial cells and human monocyte derived macrophages. KEY RESULTS We were able to demonstrate that inhibition of FAO reduced atherosclerotic plaque growth. We did not find direct anti-inflammatory properties of trimetazidine in endothelial cells or macrophages in vitro. However, we found that the activation of the NLRP3 system and the secretion of IL-1β were significantly reduced in macrophages after FAO inhibition. These results were confirmed in atherosclerotic lesions of mice treated with trimetazidine as they showed a significant reduction of IL-1β and cleaved caspase-1 in the atherosclerotic lesion as well as of IL-1β and IL-18 in the circulation. CONCLUSION Overall, we therefore suggest that the main mechanism of reducing inflammation of trimetazidine and FAO inhibition is the reduction of the NLRP-3 activation leading to reduced levels of the proinflammatory cytokine IL-1β.
Collapse
Affiliation(s)
- Philipp J Hohensinner
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria; Center for Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Max Lenz
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria
| | - Patrick Haider
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria
| | - Julia Mayer
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
| | - Manuela Richter
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
| | - Christoph Kaun
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria
| | - Laura Goederle
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Mira Brekalo
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria
| | - Manuel Salzmann
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria
| | - Smriti Sharma
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria
| | - Michael B Fischer
- Clinic for Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria; Department for Health Science and Biomedicine, Danube University Krems, Krems, Austria
| | - Stefan Stojkovic
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria
| | - Daniel Ramsmayer
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria
| | - Christian Hengstenberg
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria
| | - Bruno K Podesser
- Center for Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Kurt Huber
- 3(rd) Medical Department, Wilhelminenhospital, Vienna, Austria; Sigmund Freud University, Medical Faculty, Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Johann Wojta
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria; Core Facilities, Medical University of Vienna, Vienna, Austria.
| | - Walter S Speidl
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
44
|
Medvegy M, Simonyi G. Supplementary Therapeutic Possibilities to Alleviate Myocardial Damage Due to Microvascular Dysfunction in Coronavirus Disease 2019 (COVID-19). Cardiol Ther 2021; 10:1-7. [PMID: 33811310 PMCID: PMC8018688 DOI: 10.1007/s40119-021-00216-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Indexed: 11/28/2022] Open
Abstract
Myocardial damage with a consequent rise in cardio-specific troponin level is a frequent phenomenon in severe cases of coronavirus disease 2019 (COVID-19). Its causes are capillary endothelial cell dysfunction, associated carditis, low oxygenization, and increased sympathetic tone, which all worsen myocardial stiffness and microvascular dysfunction (MD). They lead to severe myocardial dysfunction, arrhythmia, acute congestive heart failure, and a significant rise in death cases. During COVID-19, no specific cardiological treatment is started. As adjuvant therapy, anxiolytics in COVID-19 are widely used, but not in all anxious patients who had been infected with coronavirus. Anxiolytics can be useful to moderate MD and immunosuppressive effect of anxiety. The favorable effects of trimetazidine (TMZ) and Coenzyme Q10 (CoQ10) in the treatment of myocardial ischemia and heart failure had previously been proven, and also their anti-inflammatory effects had been suspected; however, they have not yet been used in COVID-19 cases. TMZ promotes glucose-mediated ATP production, which requires less oxygen, which explains its advantageous cardiac effects. Since it lowers serum and myocardial tissue proinflammatory cytokine levels and inhibits myocardial macrophage infiltration, it was suspected that TMZ might represent a novel therapeutic agent to prevent and treat sepsis-induced myocardial dysfunction. CoQ10 plays an important role in cellular ATP production; however, its concentration is decreased in cardiovascular diseases and in influenza patients. Due to its anti-inflammatory effect, CoQ10 has been suspected to have a key therapeutic target in influenza infection. We suggest considering these medicines to alleviate myocardial damage and inflammation in COVID-19.
Collapse
Affiliation(s)
- Mihály Medvegy
- Cardiology Department, Flor Ferenc Hospital, Kistarcsa, Hungary.
| | - Gábor Simonyi
- Cardiology Department, St Imre Hospital, Budapest, Hungary
| |
Collapse
|
45
|
Xingyue L, Shuang L, Qiang W, Jinjuan F, Yongjian Y. Chrysin Ameliorates Sepsis-Induced Cardiac Dysfunction Through Upregulating Nfr2/Heme Oxygenase 1 Pathway. J Cardiovasc Pharmacol 2021; 77:491-500. [PMID: 33818552 DOI: 10.1097/fjc.0000000000000989] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 12/30/2020] [Indexed: 12/15/2022]
Abstract
ABSTRACT The incidence of myocardial dysfunction caused by sepsis is high, and the mortality of patients with sepsis can be significantly increased. During sepsis, oxidative stress and inflammation can lead to severe organ dysfunction. Flavone chrysin is one of the indispensable biological active ingredients for different fruits and vegetables and has antioxidant and anti-inflammatory properties. However, it is not clear whether chrysin is an effective treatment for heart dysfunction caused by sepsis. We found that it had protective effects against the harmful effects caused by LPS, manifested in improved survival, normalized cardiac function, improved partial pathological scores of myocardial tissue, and remission of apoptosis, as well as reduced oxidative stress and inflammation. Mechanism studies have found that chrysin is an important antioxidant protein, a key regulator of heme oxygenase 1 (HO-1). We found that HO-1 levels were increased after LPS intervention, and chrysin further increased HO-1 levels, along with the addition of Nrf2, a regulator of antioxidant proteins. Pretreatment with PD98059, an extracellular signal-regulated kinase-specific inhibitor, blocked chrysin-mediated phosphorylation of Nrf2 and the nuclear translocation of Nrf2. The protective effect of chrysin on sepsis-induced cardiac dysfunction was blocked by ZnPP, which is a HO-1 blocker. Chrysin increased antioxidant activity and reduced markers of oxidative stress (SOD and MDA) and inflammation (MPO and IL-1β), all of which were blocked by ZnPP. This indicates that HO-1 is the upstream molecule regulating the protective effect of chrysin. Thus, by upregulation of HO-1, chrysin protects against LPS-induced cardiac dysfunction and inflammation by inhibiting oxidative stress.
Collapse
Affiliation(s)
- Li Xingyue
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, PR China
| | - Li Shuang
- Department of Cardiovascular Medicine, The General Hospital of Western Theater Command PLA, Chengdu, PR China ; and
| | - Wang Qiang
- Department of Cardiovascular Medicine, The General Hospital of Western Theater Command PLA, Chengdu, PR China ; and
| | - Fu Jinjuan
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, PR China
- Department of Cardiology, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, PR China
| | - Yang Yongjian
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, PR China
- Department of Cardiovascular Medicine, The General Hospital of Western Theater Command PLA, Chengdu, PR China ; and
| |
Collapse
|
46
|
Akdemir O, Tatar BE, Gökhan A, Şirin C, Çavuşoğlu T, Erbaş O, Uyanıkgil Y, Çetin EÖ, Zhang F, Lineaweaver W. Preventive effect of trimetazidine against ischemia-reperfusion injury in rat epigastric island flaps: an experimental study. EUROPEAN JOURNAL OF PLASTIC SURGERY 2021; 44:177-188. [DOI: 10.1007/s00238-020-01757-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/20/2020] [Indexed: 10/23/2022]
|
47
|
Qian J, Luo W, Dai C, Wang J, Guan X, Zou C, Chattipakorn N, Wu G, Huang W, Liang G. Myeloid differentiation protein 2 mediates angiotensin II-induced inflammation and mesenchymal transition in vascular endothelium. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166043. [PMID: 33338595 DOI: 10.1016/j.bbadis.2020.166043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/21/2020] [Accepted: 12/04/2020] [Indexed: 11/28/2022]
Abstract
Angiotensin II (Ang II)-induced vascular inflammation and injury entails endothelial to mesenchymal transition (EndMT). Recent studies have shown that Ang II engages toll-like receptor 4 (TLR4) in the vasculature to mediate adverse effects. Here, we aimed to investigate whether myeloid differentiation protein 2 (MD2), an extracellular molecule indispensable for TLR4 activation, mediates Ang II-induced vascular injury and EndMT. We utilized MD2 knockout mice and wildtype mice treated with a specific MD2 inhibitor to decipher its role in aortas of Ang II-challenged mice. To confirm our results and to provide mechanistic insights, we exposed cultured endothelial cells to Ang II, with or without MD2 silencing. We show that Ang II causes deleterious remodeling and EndMT in aortas of mice within two weeks. These Ang II effects were largely absent in MD2 knockout mice and in wildtype mice treated with a MD2 inhibitor. MD2 silencing in cultured endothelial cells confirmed the essential role of MD2 in Ang II-induced inflammatory factor induction, and EndMT-associated phenotypic change. We also found that Ang II-MD2-EndMT axis involves the activation of nuclear factor-κB. Our studies highlight an essential role of MD2 in Ang II-induced vascular inflammation and EndMT contributing to vascular injury. These results also imply that MD2 may be targeted to dampen inflammatory cardiovascular and EndMT-associated diseases.
Collapse
Affiliation(s)
- Jinfu Qian
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wu Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chengyi Dai
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jun Wang
- Affiliated Cangnan Hospital, Wenzhou Medical University, Cangnan, Zhejiang 325800, China
| | - Xinfu Guan
- Affiliated Cangnan Hospital, Wenzhou Medical University, Cangnan, Zhejiang 325800, China
| | - Chunpeng Zou
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Gaojun Wu
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Weijian Huang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Affiliated Cangnan Hospital, Wenzhou Medical University, Cangnan, Zhejiang 325800, China; Zhuji Biomedicine Institute, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhuji, Zhejiang 311800, China.
| |
Collapse
|
48
|
Fan J, Li H, Xie R, Zhang X, Nie X, Shi X, Zhan J, Yin Z, Zhao Y, Dai B, Yuan S, Wen Z, Chen C, Wang DW. LncRNA ZNF593-AS Alleviates Contractile Dysfunction in Dilated Cardiomyopathy. Circ Res 2021; 128:1708-1723. [PMID: 33550812 DOI: 10.1161/circresaha.120.318437] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Jiahui Fan
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Huaping Li
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Rong Xie
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Xudong Zhang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Xiang Nie
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Xiaolu Shi
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China (X.S.)
| | - Jiabing Zhan
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Zhongwei Yin
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Yanru Zhao
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Beibei Dai
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Shuai Yuan
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Zheng Wen
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Dao Wen Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| |
Collapse
|
49
|
Shu H, Peng Y, Hang W, Zhou N, Wang DW. Trimetazidine in Heart Failure. Front Pharmacol 2021; 11:569132. [PMID: 33597865 PMCID: PMC7883591 DOI: 10.3389/fphar.2020.569132] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 10/07/2020] [Indexed: 12/13/2022] Open
Abstract
Heart failure is a systemic syndrome caused by multiple pathological factors. Current treatments do not have satisfactory outcomes. Several basic studies have revealed the protective effect of trimetazidine on the heart, not only by metabolism modulation but also by relieving myocardial apoptosis, fibrosis, autophagy, and inflammation. Clinical studies have consistently indicated that trimetazidine acts as an adjunct to conventional treatments and improves the symptoms of heart failure. This review summarizes the basic pathological changes in the myocardium, with an emphasis on the alteration of cardiac metabolism in the development of heart failure. The clinical application of trimetazidine in heart failure and the mechanism of its protective effects on the myocardium are carefully discussed, as well as its main adverse effects. The intention of this review is to highlight this treatment as an effective alternative against heart failure and provide additional perspectives for future studies.
Collapse
Affiliation(s)
- Hongyang Shu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Yizhong Peng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weijian Hang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Ning Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
50
|
Gunne S, Heinicke U, Parnham MJ, Laux V, Zacharowski K, von Knethen A. Nrf2-A Molecular Target for Sepsis Patients in Critical Care. Biomolecules 2020; 10:biom10121688. [PMID: 33348637 PMCID: PMC7766194 DOI: 10.3390/biom10121688] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/13/2020] [Accepted: 12/15/2020] [Indexed: 12/24/2022] Open
Abstract
The transcription factor NF-E2 p45-related factor 2 (Nrf2) is an established master regulator of the anti-oxidative and detoxifying cellular response. Thus, a role in inflammatory diseases associated with the generation of large amounts of reactive oxygen species (ROS) seems obvious. In line with this, data obtained in cell culture experiments and preclinical settings have shown that Nrf2 is important in regulating target genes that are necessary to ensure cellular redox balance. Additionally, Nrf2 is involved in the induction of phase II drug metabolizing enzymes, which are important both in degrading and converting drugs into active forms, and into putative carcinogens. Therefore, Nrf2 has also been implicated in tumorigenesis. This must be kept in mind when new therapy approaches are planned for the treatment of sepsis. Therefore, this review highlights the function of Nrf2 in sepsis with a special focus on the translation of rodent-based results into sepsis patients in the intensive care unit (ICU).
Collapse
Affiliation(s)
- Sandra Gunne
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany; (S.G.); (M.J.P.); (V.L.)
| | - Ulrike Heinicke
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany; (U.H.); (K.Z.)
| | - Michael J. Parnham
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany; (S.G.); (M.J.P.); (V.L.)
| | - Volker Laux
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany; (S.G.); (M.J.P.); (V.L.)
| | - Kai Zacharowski
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany; (U.H.); (K.Z.)
| | - Andreas von Knethen
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany; (S.G.); (M.J.P.); (V.L.)
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany; (U.H.); (K.Z.)
- Correspondence: ; Tel.: +49-69-6301-87824
| |
Collapse
|