1
|
Wang J, Ma B, Jiang X, Li C, Lin Z, Wang Y, Shi J, Wang G, Cui C. H 2 protects H9c2 cells from hypoxia/reoxygenation injury by inhibiting the Wnt/CX3CR1 signaling pathway. Med Gas Res 2025; 15:339-347. [PMID: 39511756 DOI: 10.4103/mgr.medgasres-d-24-00027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/26/2024] [Indexed: 11/15/2024] Open
Abstract
Myocardial ischemia‒reperfusion injury is a severe cardiovascular disease, and its treatment and prevention are crucial for improving patient prognosis and reducing the economic burden. This study aimed to explore the impact of hydrogen (H 2 ) on hypoxia/reoxygenation (H/R) injury in H9c2 cells (derived from rat embryonic heart tissue) induced by hydrogen peroxide (H 2 O 2 ) and to elucidate its underlying mechanism. An H/R injury model was established in H9c2 cells via exposure to 15 μM H 2 O 2 for 3 hours, followed by incubation in a 5% CO 2 atmosphere at 37°C for 24 hours. Then, the cells were treated with H 2 (50%) for 6, 12 or 24 hours. The results demonstrated that H9c2 cells exposed to H 2 O 2 and subjected to H/R injury presented a marked decrease in the cell survival rate, accompanied by severe morphological alterations, such as curling and wrinkling, and elevated lactate dehydrogenase levels. Notably, H 2 mitigated H/R injury induced by H 2 O 2 in a time-dependent manner, improving the morphological damage observed in H9c2 cells and decreasing lactate dehydrogenase levels. Compared with the model group, treatment with H 2 increased the activities of antioxidant enzymes, including catalase, superoxide dismutase, and glutathione peroxidase, while concurrently reducing the level of malondialdehyde, an indicator of cellular damage. Furthermore, H 2 treatment downregulated the expression of inflammatory cytokines and inflammatory-related factors, specifically interleukin-6, high-mobility group box 1, tumor necrosis factor-alpha, and Toll-like receptor 4, in H9c2 cells post-H/R injury. Furthermore, H 2 treatment resulted in a marked decrease in the expression levels of proteins associated with the Wnt/C-X3-C-motif receptor 1 signaling pathway, such as β-catenin, glycogen synthase kinase-3 beta, adenomatous polyposis coli, and Wnt and C-X3-C-motif receptor 1. This observation suggests a potential mechanism for its protective effects against H/R injury. Therefore, H 2 exerts a protective effect against H/R injury in H9c2 cells induced by H 2 O 2 , potentially by inhibiting the activated Wnt/C-X3-C-motif receptor 1 signaling pathway. This inhibition, in turn, prevents the generation of oxidative stress, inflammatory cytokines, and inflammation-associated factors.
Collapse
Affiliation(s)
- Jingsheng Wang
- Department of Pharmacy, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong Province, China
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Bin Ma
- Department of Cardiovascular Medicine, Taian City Taishan District People's Hospital, Taian, Shandong Province, China
| | - Xue Jiang
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong Province, China
| | - Chao Li
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong Province, China
| | - Zhaochen Lin
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong Province, China
| | - Yumei Wang
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong Province, China
| | - Jingfei Shi
- Shandong First Medical University, Jinan, Shandong Province, China
| | - Gang Wang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Chao Cui
- Department of Pharmacy, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong Province, China
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong Province, China
| |
Collapse
|
2
|
Mu G, Chen S, Chen X, Li Q, Lu B, Yu X. Hydrogen regulated pyroptosis through NLRP3-GSDMD pathway to improve airway mucosal oxidative stress injury induced by endotracheal tube cuff compression. Free Radic Biol Med 2024; 224:287-300. [PMID: 39216558 DOI: 10.1016/j.freeradbiomed.2024.08.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/21/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
The cuff of endotracheal tube (ETT) is an indispensable device for establishing an artificial airway, yet cuff-induced compression often causes damage to the airway mucosa. The mechanism of this damage involves mucosal compression ischemia and the oxidative stress injury following reperfusion. Currently, there is a lack of effective strategies to protect the mucosa. Hydrogen, as a natural antioxidant, has demonstrated significant potential in the prevention and treatment of oxidative stress injuries. This study aimed to determine the protective effects of hydrogen on compressed airway mucosa. We found that the damage to the airway mucosa caused by ETT cuff compression was associated with oxidative stress-induced pyroptosis of airway epithelial cells. Inhalation of hydrogen effectively reduced the levels of reactive oxygen species, significantly ameliorating changes in epithelial cell pyroptosis, and this protective effect is linked to the inhibition of the NLRP3-GSDMD pathway. Further cellular studies, involving knockdown and overexpression of NLRP3, clarified that hydrogen exerts its protective effects on the airway mucosa by inhibiting epithelial cell pyroptosis. Additionally, we observed that using hydrogen-rich saline to inflate the ETT cuff in patients under general anesthesia significantly reduced postoperative sore throat. This study confirms that hydrogen effectively enhances tolerance of airway mucosa to oxidative stress injuries, offering a potential preventive and therapeutic strategy for protecting the airway mucosa in patients undergoing endotracheal intubation.
Collapse
Affiliation(s)
- Guo Mu
- Department of Anesthesiology, Zigong Fourth People's Hospital, Zigong, Sichuan, 643000, China; Sichuan Clinical Research Center for Clinical Laboratory, Zigong Fourth People's Hospital, Zigong, Sichuan, 643000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, 643000, China
| | - Shuai Chen
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, 643000, China
| | - Xinyu Chen
- Chuanbei Medical College, Nangchong, Sichuan, 634700, China
| | - Qiang Li
- Department of Anesthesiology, Zigong Fourth People's Hospital, Zigong, Sichuan, 643000, China
| | - Bin Lu
- Department of Anesthesiology, Zigong Fourth People's Hospital, Zigong, Sichuan, 643000, China.
| | - Xuan Yu
- Department of Anesthesiology, Zigong Fourth People's Hospital, Zigong, Sichuan, 643000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, 643000, China.
| |
Collapse
|
3
|
Geng XF, Shang WY, Qi ZW, Zhang C, Li WX, Yan ZP, Fan XB, Zhang JP. The mechanism and promising therapeutic strategy of diabetic cardiomyopathy dysfunctions: Focus on pyroptosis. J Diabetes Complications 2024; 38:108848. [PMID: 39178624 DOI: 10.1016/j.jdiacomp.2024.108848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 08/26/2024]
Abstract
Diabetes is a major risk factor for cardiovascular diseases, and myocardial damage caused by hyperglycemia is the main cause of heart failure. However, there is still a lack of systematic understanding of myocardial damage caused by diabetes. At present, we believe that the cellular inflammatory damage caused by hyperglycemia is one of the causes of diabetic cardiomyopathy. Pyroptosis, as a proinflammatory form of cell death, is closely related to the occurrence and development of diabetic cardiomyopathy. Therefore, this paper focuses on the important role of inflammation in the occurrence and development of diabetic cardiomyopathy. From the perspective of pyroptosis, we summarize the pyroptosis of different types of cells in diabetic cardiomyopathy and its related signaling pathways. It also summarizes the treatment of diabetic cardiomyopathy, hoping to provide methods for the prevention and treatment of diabetic cardiomyopathy by inhibiting pyroptosis.
Collapse
Affiliation(s)
- Xiao-Fei Geng
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Wen-Yu Shang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Zhong-Wen Qi
- Postdoctoral Research Station of China Academy of Chinese Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, PR China
| | - Chi Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Wen-Xiu Li
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Zhi-Peng Yan
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Xin-Biao Fan
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Jun-Ping Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| |
Collapse
|
4
|
Jiao A, Liu H, Wang H, Yu J, Gong L, Zhang H, Fu L. piR112710 attenuates diabetic cardiomyopathy through inhibiting Txnip/NLRP3-mediated pyroptosis in db/db mice. Cell Signal 2024; 122:111333. [PMID: 39102928 DOI: 10.1016/j.cellsig.2024.111333] [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: 02/05/2024] [Revised: 07/17/2024] [Accepted: 07/31/2024] [Indexed: 08/07/2024]
Abstract
PIWI-interacting RNAs (piRNAs) are involved in the regulation of hypertrophic cardiomyopathy, heart failure and myocardial methylation. However, their functions and the underlying molecular mechanisms in diabetic cardiomyopathy (DCM) have yet to be fully elucidated. In the present study, a pyroptosis-associated piRNA (piR112710) was identified that ameliorates cardiac remodeling through targeting the activation of inflammasomes and mitochondrial dysfunction that are mediated via the thioredoxin-interacting protein (Txnip)/NLRP3 signaling axis. Subsequently, the cardioprotective effects of piR112710 on both the myocardium from db/db mice and cardiomyocytes from neonatal mice that were incubated with a high concentration of glucose combined with palmitate were examined. piR112710 was found to significantly improve cardiac dysfunction in db/db mice, characterized by improved echocardiography, lower levels of fibrosis, attenuated expression levels of inflammatory factors and pyroptosis-associated proteins (namely, Txnip, ASC, NLRP3, caspase-1 and GSDMD-N), and enhanced myocardial mitochondrial respiratory functions. In cultured neonatal mice cardiomyocytes, piR112710 deficiency and high glucose along with palmitate treatment led to significantly upregulated expression levels of pyroptosis associated proteins and collagens, oxidative stress, mitochondrial dysfunction and increased levels of inflammatory factors. Supplementation with piR112710, however, led to a reversal of the aforementioned changes induced by high glucose and palmitate. Mechanistically, the cardioprotective effect of piR112710 appears to be dependent upon effective elimination of reactive oxygen species and inactivation of the Txnip/NLRP3 signaling axis. Taken together, the findings of the present study have revealed that the piRNA-mediated inhibitory mechanism involving the Txnip/NLRP3 axis may participate in the regulation of pyroptosis, which protects against DCM both in vivo and in vitro. piR112710 may therefore be a potential therapeutic target for the reduction of myocardial injury caused by cardiomyocyte pyroptosis in DCM.
Collapse
Affiliation(s)
- Ande Jiao
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Huaxing Liu
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Huihui Wang
- Department of Endocrinology, Qiqihar First Hospital, Qiqihar, Heilongjiang 161041, China
| | - Jiaqi Yu
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154003, China
| | - Lu Gong
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154003, China
| | - Honglian Zhang
- College of Pharmacy, Qiqihar Medical University, Qiqihar, Heilongjiang 161003, China
| | - Lu Fu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China.
| |
Collapse
|
5
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Overview of pyroptosis mechanism and in-depth analysis of cardiomyocyte pyroptosis mediated by NF-κB pathway in heart failure. Biomed Pharmacother 2024; 179:117367. [PMID: 39214011 DOI: 10.1016/j.biopha.2024.117367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
The pyroptosis of cardiomyocytes has become an essential topic in heart failure research. The abnormal accumulation of these biological factors, including angiotensin II, advanced glycation end products, and various growth factors (such as connective tissue growth factor, vascular endothelial growth factor, transforming growth factor beta, among others), activates the nuclear factor-κB (NF-κB) signaling pathway in cardiovascular diseases, ultimately leading to pyroptosis of cardiomyocytes. Therefore, exploring the underlying molecular biological mechanisms is essential for developing novel drugs and therapeutic strategies. However, our current understanding of the precise regulatory mechanism of this complex signaling pathway in cardiomyocyte pyroptosis is still limited. Given this, this study reviews the milestone discoveries in the field of pyroptosis research since 1986, analyzes in detail the similarities, differences, and interactions between pyroptosis and other cell death modes (such as apoptosis, necroptosis, autophagy, and ferroptosis), and explores the deep connection between pyroptosis and heart failure. At the same time, it depicts in detail the complete pathway of the activation, transmission, and eventual cardiomyocyte pyroptosis of the NF-κB signaling pathway in the process of heart failure. In addition, the study also systematically summarizes various therapeutic approaches that can inhibit NF-κB to reduce cardiomyocyte pyroptosis, including drugs, natural compounds, small molecule inhibitors, gene editing, and other cutting-edge technologies, aiming to provide solid scientific support and new research perspectives for the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Zeyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhihua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuai Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
6
|
Wang B, Pan S, Nie C, Zou R, Liu J, Han X, Dong L, Zhang J, Yang X, Yu M, Fan B, Hong X, Yang W. Magnesium implantation as a continuous hydrogen production generator for the treatment of myocardial infarction in rats. Sci Rep 2024; 14:10959. [PMID: 38745034 PMCID: PMC11094026 DOI: 10.1038/s41598-024-60609-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 04/25/2024] [Indexed: 05/16/2024] Open
Abstract
Molecular hydrogen is an emerging broad-spectrum antioxidant molecule that can be used to treat myocardial infarction (MI). However, with hydrogen inhalation, the concentration that can be reached within target organs is low and the duration of action is short, which makes it difficult to achieve high dose targeted delivery of hydrogen to the heart, seriously limiting the therapeutic potential of hydrogen for MI. As a result of reactions with the internal environment of the body, subcutaneous implantation of magnesium slices leads to continuous endogenous hydrogen production, leading to a higher hydrogen concentration and a longer duration of action in target organs. In this study, we propose magnesium implant-based hydrogen therapy for MI. After subcutaneous implantation of magnesium slices in the dorsum of rats, we measured hydrogen production and efficiency, and evaluated the safety of this approach. Compared with hydrogen inhalation, it significantly improved cardiac function in rats with MI. Magnesium implantation also cleared free radicals that were released as a result of mitochondrial dysfunction, as well as suppressing cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Bin Wang
- Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuang Pan
- Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chaoqun Nie
- Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | | | - Jiaren Liu
- Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xue Han
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Li Dong
- Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiawen Zhang
- Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinrui Yang
- Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mengshu Yu
- Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bowei Fan
- Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaojian Hong
- Fourth Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Wei Yang
- Fourth Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
7
|
Chang X, Wang B, Zhao Y, Deng B, Liu P, Wang Y. The role of IFI16 in regulating PANoptosis and implication in heart diseases. Cell Death Discov 2024; 10:204. [PMID: 38693141 PMCID: PMC11063201 DOI: 10.1038/s41420-024-01978-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 05/03/2024] Open
Abstract
Interferon Gamma Inducible Protein 16 (IFI16) belongs to the HIN-200 protein family and is pivotal in immunological responses. Serving as a DNA sensor, IFI16 identifies viral and aberrant DNA, triggering immune and inflammatory responses. It is implicated in diverse cellular death mechanisms, such as pyroptosis, apoptosis, and necroptosis. Notably, these processes are integral to the emergent concept of PANoptosis, which encompasses cellular demise and inflammatory pathways. Current research implies a significant regulatory role for IFI16 in PANoptosis, particularly regarding cardiac pathologies. This review delves into the complex interplay between IFI16 and PANoptosis in heart diseases, including atherosclerosis, myocardial infarction, heart failure, and diabetic cardiomyopathy. It synthesizes evidence of IFI16's impact on PANoptosis, with the intention of providing novel insights for therapeutic strategies targeting heart diseases.
Collapse
Affiliation(s)
- Xindi Chang
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China
| | - Bei Wang
- Department of Emergency, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China
| | - Yingli Zhao
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China
| | - Bing Deng
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China
| | - Ping Liu
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China.
| | - Yiru Wang
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China.
| |
Collapse
|
8
|
Ma YL, Xu M, Cen XF, Qiu HL, Guo YY, Tang QZ. Tectorigenin protects against cardiac fibrosis in diabetic mice heart via activating the adiponectin receptor 1-mediated AMPK pathway. Biomed Pharmacother 2024; 174:116589. [PMID: 38636400 DOI: 10.1016/j.biopha.2024.116589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/30/2024] [Accepted: 04/10/2024] [Indexed: 04/20/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is a common severe complication of diabetes that occurs independently of hypertension, coronary artery disease, and valvular cardiomyopathy, eventually leading to heart failure. Previous studies have reported that Tectorigenin (TEC) possesses extensive anti-inflammatory and anti-oxidative stress properties. In this present study, the impact of TEC on diabetic cardiomyopathy was examined. The model of DCM in mice was established with the combination of a high-fat diet and STZ treatment. Remarkably, TEC treatment significantly attenuated cardiac fibrosis and improved cardiac dysfunction. Concurrently, TEC was also found to mitigate hyperglycemia and hyperlipidemia in the DCM mouse. At the molecular level, TEC is involved in the activation of AMPK, both in vitro and in vivo, by enhancing its phosphorylation. This is achieved through the regulation of endothelial-mesenchymal transition via the AMPK/TGFβ/Smad3 pathway. Furthermore, it was demonstrated that the level of ubiquitination of the adiponectin receptor 1 (AdipoR1) protein is associated with TEC-mediated improvement of cardiac dysfunction in DCM mice. Notably the substantial reduction of myocardial fibrosis. In conclusion, TEC improves cardiac fibrosis in DCM mice by modulating the AdipoR1/AMPK signaling pathway. These findings suggest that TEC could be an effective therapeutic agent for the treatment of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Yu-Lan Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Man Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Xian-Feng Cen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Hong-Liang Qiu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Ying-Ying Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China.
| |
Collapse
|
9
|
Hu Q, Li Y, Lin Z, Zhang H, Chen H, Chao C, Zhao C. The Molecular Biological Mechanism of Hydrogen Therapy and Its Application in Spinal Cord Injury. Drug Des Devel Ther 2024; 18:1399-1414. [PMID: 38707612 PMCID: PMC11068043 DOI: 10.2147/dddt.s463177] [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: 02/15/2024] [Accepted: 04/23/2024] [Indexed: 05/07/2024] Open
Abstract
Hydrogen, which is a novel biomedical molecule, is currently the subject of extensive research involving animal experiments and in vitro cell experiments, and it is gradually being applied in clinical settings. Hydrogen has been proven to possess anti-inflammatory, selective antioxidant, and antiapoptotic effects, thus exhibiting considerable protective effects in various diseases. In recent years, several studies have provided preliminary evidence for the protective effects of hydrogen on spinal cord injury (SCI). This paper provides a comprehensive review of the potential molecular biology mechanisms of hydrogen therapy and its application in treating SCI, with an aim to better explore the medical value of hydrogen and provide new avenues for the adjuvant treatment of SCI.
Collapse
Affiliation(s)
- Quan Hu
- Department of Neurosurgery, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an City, Shandong, 271000, People’s Republic of China
| | - Yingxiao Li
- Department of Gynecology, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an City, Shandong, 271000, People’s Republic of China
| | - Zhaochen Lin
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an City, Shandong, 271000, People’s Republic of China
| | - Hao Zhang
- Department of Rehabilitation Medical Center, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an City, Shandong, 271000, People’s Republic of China
| | - Haoyue Chen
- Department of Rehabilitation Medical Center, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an City, Shandong, 271000, People’s Republic of China
| | - Cui Chao
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an City, Shandong, 271000, People’s Republic of China
| | - Chuanliang Zhao
- Department of Orthopedics, the Affiliated Taian City Central Hospital of Qingdao University, Tai’an City, Shandong, 271000, People’s Republic of China
| |
Collapse
|
10
|
Li M, Liu L, Zhang C, Deng L, Zhong Y, Liao B, Li X, Wan Y, Feng J. The latest emerging drugs for the treatment of diabetic cardiomyopathy. Expert Opin Pharmacother 2024; 25:641-654. [PMID: 38660817 DOI: 10.1080/14656566.2024.2347468] [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: 02/05/2024] [Accepted: 04/22/2024] [Indexed: 04/26/2024]
Abstract
INTRODUCTION Diabetic cardiomyopathy (DCM) is a serious complication of diabetes mellitus involving multiple pathophysiologic mechanisms. In addition to hypoglycemic agents commonly used in diabetes, metabolism-related drugs, natural plant extracts, melatonin, exosomes, and rennin-angiotensin-aldosterone system are cardioprotective in DCM. However, there is a lack of systematic summarization of drugs for DCM. AREAS COVERED In this review, the authors systematically summarize the most recent drugs used for the treatment of DCM and discusses them from the perspective of DCM pathophysiological mechanisms. EXPERT OPINION We discuss DCM drugs from the perspective of the pathophysiological mechanisms of DCM, mainly including inflammation and metabolism. As a disease with multiple pathophysiological mechanisms, the combination of drugs may be more advantageous, and we have discussed some of the current studies on the combination of drugs.
Collapse
Affiliation(s)
- Minghao Li
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Lin Liu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Chunyu Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Li Deng
- Department of Rheumatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yi Zhong
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Bin Liao
- Department of Cardiovascular Surgery, Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiuying Li
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University; Department of Pathophysiology, School of Basic Medical Science, Southwest Medical University, Luzhou, China
| | - Ying Wan
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University; Department of Pathophysiology, School of Basic Medical Science, Southwest Medical University, Luzhou, China
| | - Jian Feng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
11
|
Pang H, Huang G, Xie Z, Zhou Z. The role of regulated necrosis in diabetes and its complications. J Mol Med (Berl) 2024; 102:495-505. [PMID: 38393662 DOI: 10.1007/s00109-024-02421-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 12/21/2023] [Accepted: 01/16/2024] [Indexed: 02/25/2024]
Abstract
Morphologically, cell death can be divided into apoptosis and necrosis. Apoptosis, which is a type of regulated cell death, is well tolerated by the immune system and is responsible for hemostasis and cellular turnover under physiological conditions. In contrast, necrosis is defined as a form of passive cell death that leads to a dramatic inflammatory response (also referred to as necroinflammation) and causes organ dysfunction under pathological conditions. Recently, a novel form of cell death named regulated necrosis (such as necroptosis, pyroptosis, and ferroptosis) was discovered. Distinct from apoptosis, regulated necrosis is modulated by multiple internal or external factors, but meanwhile, it results in inflammation and immune response. Accumulating evidence has indicated that regulated necrosis is associated with multiple diseases, including diabetes. Diabetes is characterized by hyperglycemia caused by insulin deficiency and/or insulin resistance, and long-term high glucose leads to various diabetes-related complications. Here, we summarize the mechanisms of necroptosis, pyroptosis, and ferroptosis, and introduce recent advances in characterizing the associations between these three types of regulated necrosis and diabetes and its complications.
Collapse
Affiliation(s)
- Haipeng Pang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Gan Huang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Zhiguo Xie
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
12
|
Liu P, Zhang Z, Chen H, Chen Q. Pyroptosis: Mechanisms and links with diabetic cardiomyopathy. Ageing Res Rev 2024; 94:102182. [PMID: 38182080 DOI: 10.1016/j.arr.2023.102182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 12/27/2023] [Accepted: 12/30/2023] [Indexed: 01/07/2024]
Abstract
Diabetes mellitus (DM) is a chronic metabolic disease characterized by hyperglycaemia that seriously affects human health. Diabetic cardiomyopathy (DCM) is a major cardiovascular complication and one of the main causes of death in patients with DM. Although DCM attracts great attention, and new therapeutic methods are continuously developed, there is a lack of effective treatment strategies. Therefore, exploring and targeting new signalling pathways related to the evolution of DCM becomes a hotspot and difficulty in the prevention and treatment of DCM. Pyroptosis is a newly discovered regulated cell death that is heavily dependent on the formation of plasma membrane pores by members of the gasdermin protein family and is reported to be involved in the occurrence, development, and pathogenesis of DCM. In this review, we focus on the molecular mechanisms of pyroptosis, its involvement in the relevant signalling pathways of DCM, and potential pyroptosis-targeting therapeutic strategies for the treatment of DCM. Our review provides new insights into the use of pyroptosis as a useful tool for the prevention and treatment of DCM and clarifies future research directions.
Collapse
Affiliation(s)
- Pan Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, PR China
| | - Zhengdong Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, Sichuan Province, PR China; Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, Sichuan Province, PR China
| | - Huizhen Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, PR China
| | - Qiu Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, PR China.
| |
Collapse
|
13
|
Wang G, Ma TY, Huang K, Zhong JH, Lu SJ, Li JJ. Role of pyroptosis in diabetic cardiomyopathy: an updated review. Front Endocrinol (Lausanne) 2024; 14:1322907. [PMID: 38250736 PMCID: PMC10796545 DOI: 10.3389/fendo.2023.1322907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/06/2023] [Indexed: 01/23/2024] Open
Abstract
Diabetic cardiomyopathy (DCM), one of the common complications of diabetes, presents as a specific cardiomyopathy with anomalies in the structure and function of the heart. With the increasing prevalence of diabetes, DCM has a high morbidity and mortality worldwide. Recent studies have found that pyroptosis, as a programmed cell death accompanied by an inflammatory response, exacerbates the growth and genesis of DCM. These studies provide a theoretical basis for exploring the potential treatment of DCM. Therefore, this review aims to summarise the possible mechanisms by which pyroptosis promotes the development of DCM as well as the relevant studies targeting pyroptosis for the possible treatment of DCM, focusing on the molecular mechanisms of NLRP3 inflammasome-mediated pyroptosis, different cellular pyroptosis pathways associated with DCM, the effects of pyroptosis occurring in different cells on DCM, and the relevant drugs targeting NLRP3 inflammasome/pyroptosis for the treatment of DCM. This review might provide a fresh perspective and foundation for the development of therapeutic agents for DCM.
Collapse
Affiliation(s)
- Gan Wang
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, Hainan, China
| | - Tian-Yi Ma
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, Hainan, China
| | - Kang Huang
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, Hainan, China
| | - Jiang-Hua Zhong
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, Hainan, China
| | - Shi-Juan Lu
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, Hainan, China
| | - Jian-Jun Li
- State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
14
|
Quaiyoom A, Kumar R. An Overview of Diabetic Cardiomyopathy. Curr Diabetes Rev 2024; 20:e121023222139. [PMID: 37842898 DOI: 10.2174/0115733998255538231001122639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/13/2023] [Accepted: 08/17/2023] [Indexed: 10/17/2023]
Abstract
Diabetic cardiomyopathy (DCM) is a myocardial disorder that is characterised by structural and functional abnormalities of the heart muscle in the absence of hypertension, valvular heart disease, congenital heart defects, or coronary artery disease (CAD). After witnessing a particular form of cardiomyopathy in diabetic individuals, Rubler et al. came up with the moniker diabetic cardiomyopathy in 1972. Four stages of DCM are documented, and the American College of Cardiology/American Heart Association Stage and New York Heart Association Class for HF have some overlap. Diabetes is linked to several distinct forms of heart failure. Around 40% of people with heart failure with preserved ejection fraction (HFpEF) have diabetes, which is thought to be closely associated with the pathophysiology of HFpEF. Diabetes and HF are uniquely associated in a bidirectional manner. When compared to the general population without diabetes, those with diabetes have a risk of heart failure that is up to four times higher. A biomarker is a trait that is reliably measured and assessed as a predictor of healthy biological activities, pathological processes, or pharmacologic responses to a clinical treatment. Several biomarker values have been discovered to be greater in patients with diabetes than in control subjects among those who have recently developed heart failure. Myocardial fibrosis and hypertrophy are the primary characteristics of DCM, and structural alterations in the diabetic myocardium are often examined by non-invasive, reliable, and reproducible procedures. An invasive method called endomyocardial biopsy (EMB) is most often used to diagnose many cardiac illnesses.
Collapse
Affiliation(s)
- Abdul Quaiyoom
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, India
| | - Ranjeet Kumar
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, India
| |
Collapse
|
15
|
Xie Y, Zhao G, Lei X, Cui N, Wang H. Advances in the regulatory mechanisms of mTOR in necroptosis. Front Immunol 2023; 14:1297408. [PMID: 38164133 PMCID: PMC10757967 DOI: 10.3389/fimmu.2023.1297408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024] Open
Abstract
The mammalian target of rapamycin (mTOR), an evolutionarily highly conserved serine/threonine protein kinase, plays a prominent role in controlling gene expression, metabolism, and cell death. Programmed cell death (PCD) is indispensable for maintaining homeostasis by removing senescent, defective, or malignant cells. Necroptosis, a type of PCD, relies on the interplay between receptor-interacting serine-threonine kinases (RIPKs) and the membrane perforation by mixed lineage kinase domain-like protein (MLKL), which is distinguished from apoptosis. With the development of necroptosis-regulating mechanisms, the importance of mTOR in the complex network of intersecting signaling pathways that govern the process has become more evident. mTOR is directly responsible for the regulation of RIPKs. Autophagy is an indirect mechanism by which mTOR regulates the removal and interaction of RIPKs. Another necroptosis trigger is reactive oxygen species (ROS) produced by oxidative stress; mTOR regulates necroptosis by exploiting ROS. Considering the intricacy of the signal network, it is reasonable to assume that mTOR exerts a bifacial effect on necroptosis. However, additional research is necessary to elucidate the underlying mechanisms. In this review, we summarized the mechanisms underlying mTOR activation and necroptosis and highlighted the signaling pathway through which mTOR regulates necroptosis. The development of therapeutic targets for various diseases has been greatly advanced by the expanding knowledge of how mTOR regulates necroptosis.
Collapse
Affiliation(s)
- Yawen Xie
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Guoyu Zhao
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xianli Lei
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Na Cui
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Hao Wang
- Department of Critical Care Medicine, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
16
|
Liu Z, Chen Y, Mei Y, Yan M, Liang H. Gasdermin D-Mediated Pyroptosis in Diabetic Cardiomyopathy: Molecular Mechanisms and Pharmacological Implications. Molecules 2023; 28:7813. [PMID: 38067543 PMCID: PMC10708146 DOI: 10.3390/molecules28237813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/21/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is a pathophysiological condition triggered by diabetes mellitus (DM), which can lead to heart failure (HF). One of the most important cellular processes associated with DCM is the death of cardiomyocytes. Gasdermin D (GSDMD) plays a key role in mediating pyroptosis, a type of programmed cell death closely associated with inflammasome activation. Recent studies have revealed that pyroptosis is induced during hyperglycemia, which is crucial to the development of DCM. Although the effects of pyroptosis on DCM have been discussed, the relationship between DCM and GSDMD is not fully clarified. Recent studies gave us the impetus for clarifying the meaning of GSDMD in DCM. The purpose of this review is to summarize new and emerging insights, mainly discussing the structures of GSDMD and the mechanism of pore formation, activation pathways, molecular mechanisms of GSDMD-mediated pyroptosis, and the therapeutic potential of GSDMD in DCM. The implications of this review will pave the way for a new therapeutic target in DCM.
Collapse
Affiliation(s)
- Zhou Liu
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; (Z.L.); (Y.C.); (Y.M.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou 510006, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China
| | - Yifan Chen
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; (Z.L.); (Y.C.); (Y.M.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou 510006, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China
| | - Yu Mei
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; (Z.L.); (Y.C.); (Y.M.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou 510006, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China
| | - Meiling Yan
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; (Z.L.); (Y.C.); (Y.M.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou 510006, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China
| | - Haihai Liang
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; (Z.L.); (Y.C.); (Y.M.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou 510006, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China
| |
Collapse
|
17
|
Nie C, A R, Wang J, Pan S, Zou R, Wang B, Xi S, Hong X, Zhou M, Wang H, Yu M, Wu L, Sun X, Yang W. Controlled Release of Hydrogen-Carrying Perfluorocarbons for Ischemia Myocardium-Targeting 19 F MRI-Guided Reperfusion Injury Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304178. [PMID: 37596718 PMCID: PMC10582447 DOI: 10.1002/advs.202304178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/20/2023] [Indexed: 08/20/2023]
Abstract
Hydrogen gas is recently proven to have anti-oxidative and anti-inflammation effects on ischemia-reperfusion injury. However, the efficacy of hydrogen therapy is limited by the efficiency of hydrogen storage, targeted delivery, and controlled release. In this study, H2 -PFOB nanoemulsions (NEs) is developed with high hydrogen loading capacity for targeted ischemic myocardium precision therapy. The hydrogen-carrying capacity of H2 -PFOB NEs is determined by gas chromatography and microelectrode methods. Positive uptake of H2 -PFOB NEs in ischemia-reperfusion myocardium and the influence of hydrogen on 19 F-MR signal are quantitatively visualized using a 9.4T MR imaging system. The biological therapeutic effects of H2 -PFOB NEs are examined on a myocardial ischemia-reperfusion injury mouse model. The results illustrated that the developed H2 -PFOB NEs can efficaciously achieve specific infiltration into ischemic myocardium and exhibit excellent antioxidant and anti-inflammatory properties on myocardial ischemia-reperfusion injury, which can be dynamically visualized by 19 F-MR imaging system. Moreover, hydrogen burst release induced by low-intensity focused ultrasound (LIFU) irradiation further promotes the therapeutic effect of H2 -PFOB NEs with a favorable biosafety profile. In this study, the potential therapeutic effects of H2 -PFOB NEs is fully unfolded, which may hold great potential for future hydrogen-based precision therapeutic applications tailored to ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Chaoqun Nie
- Department of CardiologyThe Fourth Hospital of Harbin Medical University150000HarbinP. R. China
| | - Rong A
- Department of Nuclear MedicineThe Fourth Hospital of Harbin Medical University150000HarbinP. R. China
- NHC Key Laboratory of Molecular Probe and Targeted TheranosticsMolecular Imaging Research Center (MIRC) of Harbin Medical University150000HarbinP. R. China
| | - Jing Wang
- Department of Nuclear MedicineThe Fourth Hospital of Harbin Medical University150000HarbinP. R. China
- NHC Key Laboratory of Molecular Probe and Targeted TheranosticsMolecular Imaging Research Center (MIRC) of Harbin Medical University150000HarbinP. R. China
| | - Shuang Pan
- Department of CardiologyThe Fourth Hospital of Harbin Medical University150000HarbinP. R. China
| | - Rentong Zou
- Department of CardiologyThe Fourth Hospital of Harbin Medical University150000HarbinP. R. China
| | - Bin Wang
- Department of CardiologyThe Fourth Hospital of Harbin Medical University150000HarbinP. R. China
| | - Shuiqing Xi
- Department of CardiologyThe Fourth Hospital of Harbin Medical University150000HarbinP. R. China
| | - Xiaojian Hong
- Department of CardiologyThe Fourth Hospital of Harbin Medical University150000HarbinP. R. China
| | - Meifang Zhou
- Department of Nuclear MedicineThe Fourth Hospital of Harbin Medical University150000HarbinP. R. China
- NHC Key Laboratory of Molecular Probe and Targeted TheranosticsMolecular Imaging Research Center (MIRC) of Harbin Medical University150000HarbinP. R. China
| | - Haoyu Wang
- Department of Nuclear MedicineThe Fourth Hospital of Harbin Medical University150000HarbinP. R. China
- NHC Key Laboratory of Molecular Probe and Targeted TheranosticsMolecular Imaging Research Center (MIRC) of Harbin Medical University150000HarbinP. R. China
| | - Mengshu Yu
- Department of CardiologyThe Fourth Hospital of Harbin Medical University150000HarbinP. R. China
| | - Lina Wu
- Department of Nuclear MedicineThe Fourth Hospital of Harbin Medical University150000HarbinP. R. China
- NHC Key Laboratory of Molecular Probe and Targeted TheranosticsMolecular Imaging Research Center (MIRC) of Harbin Medical University150000HarbinP. R. China
| | - Xilin Sun
- Department of Nuclear MedicineThe Fourth Hospital of Harbin Medical University150000HarbinP. R. China
- NHC Key Laboratory of Molecular Probe and Targeted TheranosticsMolecular Imaging Research Center (MIRC) of Harbin Medical University150000HarbinP. R. China
| | - Wei Yang
- Department of CardiologyThe Fourth Hospital of Harbin Medical University150000HarbinP. R. China
| |
Collapse
|
18
|
Li X, Su X, Xia F, Qiu J, Zhang J, Wu H, Xie X, Xu M. Bibliometric and visual analysis of diabetes mellitus and pyroptosis from 2011 to 2022. Eur J Med Res 2023; 28:235. [PMID: 37443131 DOI: 10.1186/s40001-023-01175-7] [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: 02/01/2023] [Accepted: 06/14/2023] [Indexed: 07/15/2023] Open
Abstract
OBJECTIVE To visualize and analyze the published literature on diabetes mellitus and pyroptosis based on a bibliometric approach, so as to provide a comprehensive picture of the hot research directions and dynamic progress in this field. METHODS This study was based on the web of science core collection database to conduct a comprehensive search of the published literature in the field of diabetes mellitus and Pyroptosis from January 1985 to August 2022, including the published research literature in this field, as well as a visual analysis of the number of citations, year of publication, journal, author, research institution, country, and research topic. RESULTS A total of 139 literature on research related to diabetes mellitus and cellular scorch from 2011 to 2022 were retrieved, with a total of 3009 citations and a maximum of 255 citations for a single article, which had a first author Schmid-Burgk, JL The first author of this article is from Germany; among 20 publishing countries, China leads with 100 articles; among 222 publishing institutions, Harbin Medical University leads with 18 articles and 184 citations; among 980 authors, Chen, X from China tops the list of high-impact authors with 5 articles and 29 citations. Among the 98 journals, "CELL DEATH DISEASE" ranked first in both volume and high-impact journals with 4 articles and 29 citations. Among 349 keywords, "pyroptosis" ranked first with a cumulative frequency of 65 times. The cluster analysis was divided into three categories, chronic complications of diabetes mellitus and pyroptosis (67 articles), diabetes mellitus and pyroptosis (60 articles), and diabetes mellitus combined with other diseases and pyroptosis (12 articles), and the number of articles related to diabetes mellitus and its chronic complications increased rapidly from 2019, among which, diabetic cardiomyopathy (27 articles) had the highest number of articles. CONCLUSIONS Based on a comprehensive analysis of published literature in the field of diabetes mellitus and pyroptosis from 2011 to 2022, this study achieved a visual analysis of studies with significant and outstanding contributions to the field, thus framing a picture showing the development and changes in the field. At the same time, this study provides research information and direction for clinicians and investigators to conduct diabetes mellitus and pyroptosis-related research in the future.
Collapse
Affiliation(s)
- Xiaodong Li
- The First Affiliated Hospital of Guizhou University of Chinese Medicine, Guiyang, 550000, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Xiaojuan Su
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Fenglin Xia
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Jing Qiu
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Jiaqi Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Haiyan Wu
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Xuejun Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Mingchao Xu
- Traditional Chinese Medicine Hospital of Meishan, Meishan, 620010, China.
| |
Collapse
|
19
|
Samidurai A, Saravanan M, Ockaili R, Kraskauskas D, Lau SYV, Kodali V, Ramasamy S, Bhoopathi K, Nair M, Roh SK, Kukreja RC, Das A. Single-Dose Treatment with Rapamycin Preserves Post-Ischemic Cardiac Function through Attenuation of Fibrosis and Inflammation in Diabetic Rabbit. Int J Mol Sci 2023; 24:8998. [PMID: 37240345 PMCID: PMC10218967 DOI: 10.3390/ijms24108998] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/09/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Robust activation of mTOR (mammalian target of rapamycin) signaling in diabetes exacerbates myocardial injury following lethal ischemia due to accelerated cardiomyocyte death with cardiac remodeling and inflammatory responses. We examined the effect of rapamycin (RAPA, mTOR inhibitor) on cardiac remodeling and inflammation following myocardial ischemia/reperfusion (I/R) injury in diabetic rabbits. Diabetic rabbits (DM) were subjected to 45 min of ischemia and 10 days of reperfusion by inflating/deflating a previously implanted hydraulic balloon occluder. RAPA (0.25 mg/kg, i.v.) or DMSO (vehicle) was infused 5 min before the onset of reperfusion. Post-I/R left ventricular (LV) function was assessed by echocardiography and fibrosis was evaluated by picrosirius red staining. Treatment with RAPA preserved LV ejection fraction and reduced fibrosis. Immunoblot and real-time PCR revealed that RAPA treatment inhibited several fibrosis markers (TGF-β, Galectin-3, MYH, p-SMAD). Furthermore, immunofluorescence staining revealed the attenuation of post-I/R NLRP3-inflammasome formation with RAPA treatment as shown by reduced aggregation of apoptosis speck-like protein with a caspase recruitment domain and active-form of caspase-1 in cardiomyocytes. In conclusion, our study suggests that acute reperfusion therapy with RAPA may be a viable strategy to preserve cardiac function with the alleviation of adverse post-infarct myocardial remodeling and inflammation in diabetic patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Rakesh C. Kukreja
- Division of Cardiology, Pauley Heart Center, Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (A.S.)
| | - Anindita Das
- Division of Cardiology, Pauley Heart Center, Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (A.S.)
| |
Collapse
|
20
|
Xie F, Song Y, Yi Y, Jiang X, Ma S, Ma C, Li J, Zhanghuang Z, Liu M, Zhao P, Ma X. Therapeutic Potential of Molecular Hydrogen in Metabolic Diseases from Bench to Bedside. Pharmaceuticals (Basel) 2023; 16:ph16040541. [PMID: 37111299 PMCID: PMC10141176 DOI: 10.3390/ph16040541] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/26/2023] [Accepted: 03/29/2023] [Indexed: 04/29/2023] Open
Abstract
Oxidative stress and chronic inflammation have been implicated in the pathophysiology of metabolic diseases, including diabetes mellitus (DM), metabolic syndrome (MS), fatty liver (FL), atherosclerosis (AS), and obesity. Molecular hydrogen (H2) has long been considered a physiologically inert gas. In the last two decades, accumulating evidence from pre-clinical and clinical studies has indicated that H2 may act as an antioxidant to exert therapeutic and preventive effects on various disorders, including metabolic diseases. However, the mechanisms underlying the action of H2 remain unclear. The purpose of this review was to (1) provide an overview of the current research on the potential effects of H2 on metabolic diseases; (2) discuss the possible mechanisms underlying these effects, including the canonical anti-oxidative, anti-inflammatory, and anti-apoptotic effects, as well as suppression of ER stress, activation of autophagy, improvement of mitochondrial function, regulation of gut microbiota, and other possible mechanisms. The potential target molecules of H2 will also be discussed. With more high-quality clinical trials and in-depth mechanism research, it is believed that H2 will eventually be applied to clinical practice in the future, to benefit more patients with metabolic disease.
Collapse
Affiliation(s)
- Fei Xie
- Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
- Beijing Molecular Hydrogen Research Center, Beijing 100124, China
| | - Yifei Song
- Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
- Beijing Molecular Hydrogen Research Center, Beijing 100124, China
| | - Yang Yi
- Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
- Beijing Molecular Hydrogen Research Center, Beijing 100124, China
| | - Xue Jiang
- Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
- Beijing Molecular Hydrogen Research Center, Beijing 100124, China
| | - Shiwen Ma
- Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
- Beijing Molecular Hydrogen Research Center, Beijing 100124, China
| | - Chen Ma
- Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
- Beijing Molecular Hydrogen Research Center, Beijing 100124, China
| | - Junyu Li
- Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
- Beijing Molecular Hydrogen Research Center, Beijing 100124, China
| | - Ziyi Zhanghuang
- Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
- Beijing Molecular Hydrogen Research Center, Beijing 100124, China
| | - Mengyu Liu
- Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
- Beijing Molecular Hydrogen Research Center, Beijing 100124, China
| | - Pengxiang Zhao
- Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
- Beijing Molecular Hydrogen Research Center, Beijing 100124, China
| | - Xuemei Ma
- Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
- Beijing Molecular Hydrogen Research Center, Beijing 100124, China
| |
Collapse
|
21
|
Dhar A, Venkadakrishnan J, Roy U, Vedam S, Lalwani N, Ramos KS, Pandita TK, Bhat A. A comprehensive review of the novel therapeutic targets for the treatment of diabetic cardiomyopathy. Ther Adv Cardiovasc Dis 2023; 17:17539447231210170. [PMID: 38069578 PMCID: PMC10710750 DOI: 10.1177/17539447231210170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 10/09/2023] [Indexed: 12/18/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is characterized by structural and functional abnormalities in the myocardium affecting people with diabetes. Treatment of DCM focuses on glucose control, blood pressure management, lipid-lowering, and lifestyle changes. Due to limited therapeutic options, DCM remains a significant cause of morbidity and mortality in patients with diabetes, thus emphasizing the need to develop new therapeutic strategies. Ongoing research is aimed at understanding the underlying molecular mechanism(s) involved in the development and progression of DCM, including oxidative stress, inflammation, and metabolic dysregulation. The goal is to develope innovative pharmaceutical therapeutics, offering significant improvements in the clinical management of DCM. Some of these approaches include the effective targeting of impaired insulin signaling, cardiac stiffness, glucotoxicity, lipotoxicity, inflammation, oxidative stress, cardiac hypertrophy, and fibrosis. This review focuses on the latest developments in understanding the underlying causes of DCM and the therapeutic landscape of DCM treatment.
Collapse
Affiliation(s)
- Arti Dhar
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Hyderabad, Telangana, India
| | | | - Utsa Roy
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Hyderabad, Telangana, India
| | - Sahithi Vedam
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Hyderabad, Telangana, India
| | - Nikita Lalwani
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Hyderabad, Telangana, India
| | - Kenneth S. Ramos
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, TX 77030, USA
| | - Tej K. Pandita
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, TX 77030, USA
| | - Audesh Bhat
- Centre for Molecular Biology, Central University of Jammu, Samba, Jammu and Kashmir (UT) 184311, India
| |
Collapse
|
22
|
Zhang T, Wang Y, Yao W, Chen Y, Zhang D, Gao Y, Jin S, Li L, Yang S, Wu Y. Metformin antagonizes nickel-refining fumes-induced cell pyroptosis via Nrf2/GOLPH3 pathway in vitro and in vivo. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 247:114233. [PMID: 36334342 DOI: 10.1016/j.ecoenv.2022.114233] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/20/2022] [Accepted: 10/23/2022] [Indexed: 05/16/2023]
Abstract
Nickel compounds, an international carcinogen in the industrial environment, increased the risk of lung inflammation even lung cancer in Ni refinery workers. Metformin has displayed the intense anti-inflammation and anti-cancer properties through regulating pyroptosis. This study was designed to explore whether Nickel-refining fumes (NiRF) can induce cell pyroptosis and how AMPK/CREB/Nrf2 mediated the protection afforded by metformin against Ni particles-induced lung impairment. Our results represented that Ni fumes exposure evoked pyroptosis via GOLPH3 and induced oxidative stress, while, metformin treatment alleviated Ni particles-mediated above changes. Moreover, nuclear factor erythroid 2-related factor 2 (Nrf2) involved in the protection of metformin, and the deficiency of Nrf2 attenuated the beneficial protection. We also determined that Nrf2 was a downstream molecule of AMPK/CREB pathway. Furthermore, male C57BL/6 mice were administered with Ni at a dose of 2 mg/kg by non-exposed endotracheal instillation and metformin (100, 200 and 300 mg/kg) via oral gavage for 4 weeks. The results indicated that NiRF promoted GOLPH3 and pyroptosis by stimulating NLRP3, caspase-1, N-GSDMD, IL-18 and IL-1β expression. However, various doses of metformin reduced GOLPH3 and the above protein levels of pyroptosis, also improved AMPK/CREB/Nrf2 expression. In summary, we found that metformin suppressed NiRF-connected GOLPH3-prompted pyroptosis via AMPK/CREB/Nrf2 signaling pathway to confer pulmonary protection.
Collapse
Affiliation(s)
- Tong Zhang
- Department of Occupational Health, College of Public Health, Harbin Medical University, Harbin, Heilongjiang Province 150086, PR China
| | - Yue Wang
- Department of Occupational Health, College of Public Health, Harbin Medical University, Harbin, Heilongjiang Province 150086, PR China
| | - Wenxue Yao
- Department of Occupational Health, College of Public Health, Harbin Medical University, Harbin, Heilongjiang Province 150086, PR China
| | - Yangyang Chen
- Department of Occupational Health, College of Public Health, Harbin Medical University, Harbin, Heilongjiang Province 150086, PR China
| | - Dan Zhang
- Department of Occupational Health, College of Public Health, Harbin Medical University, Harbin, Heilongjiang Province 150086, PR China
| | - Ying Gao
- Department of Occupational Health, College of Public Health, Harbin Medical University, Harbin, Heilongjiang Province 150086, PR China
| | - Shuo Jin
- Department of Occupational Health, College of Public Health, Harbin Medical University, Harbin, Heilongjiang Province 150086, PR China
| | - Lina Li
- Department of Occupational Health, College of Public Health, Harbin Medical University, Harbin, Heilongjiang Province 150086, PR China
| | - Shikuan Yang
- Department of Occupational Health, College of Public Health, Harbin Medical University, Harbin, Heilongjiang Province 150086, PR China
| | - Yonghui Wu
- Department of Occupational Health, College of Public Health, Harbin Medical University, Harbin, Heilongjiang Province 150086, PR China.
| |
Collapse
|
23
|
Sun H, Song K, Zhou Y, Ding JF, Tu B, Yang JJ, Sha JM, Zhao JY, Zhang Y, Tao H. MTHFR epigenetic derepression protects against diabetes cardiac fibrosis. Free Radic Biol Med 2022; 193:330-341. [PMID: 36279972 DOI: 10.1016/j.freeradbiomed.2022.10.304] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/17/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Diabetes cardiac fibrosis is associated with altered DNA methylation of fibrogenic genes; however, the underlying mechanisms remain unclear. OBJECTIVES In this study, we investigate the critical role of DNA methylation aberration-associated suppression of MTHFR in diabetes cardiac fibrosis, and the protective effects of folate on diabetes cardiac fibrosis, using cultured cells, animal models, and clinical samples. METHODS AND RESULTS Herein, we report that DNA methylation repression of MTHFR, critically involved in diabetes cardiac fibrosis, mediates the significant protective effects of folate in a mouse model of diabetes cardiac fibrosis induced by STZ. Heart MTHFR expression was markedly suppressed in diabetes cardiac fibrosis patients and mice, accompanied by increased DNMT3A and MTHFR promoter methylation. Knockdown of DNMT3A demethylated MTHFR promoter, recovered the MTHFR loss, and alleviated the diabetes cardiac fibrosis pathology and cardiac fibroblasts pyroptosis. Mechanistically, DNMT3A epigenetically repressed MTHFR expression via methylation of the promoter. Interestingly, folate supplementation can rescue the effect of MTHFR loss in diabetes cardiac fibrosis, suggesting that inactivation of MTHFR through epigenetics is a critical mediator of diabetes cardiac fibrosis. CONCLUSIONS The current study identifies that MTHFR repression due to aberrant DNMT3A elevation and subsequent MTHFR promoter hypermethylation is likely an important epigenetic feature of diabetes cardiac fibrosis, and folate supplementation protects against diabetes cardiac fibrosis.
Collapse
Affiliation(s)
- He Sun
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China
| | - Kai Song
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China
| | - Yang Zhou
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China
| | - Ji-Fei Ding
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China; Department of Cardiothoracic Surgery, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, Jiangsu Province, PR China
| | - Bin Tu
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China
| | - Jing-Jing Yang
- Department of Clinical Pharmacy, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China
| | - Ji-Ming Sha
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China
| | - Jian-Yuan Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China.
| | - Ye Zhang
- Department of Anesthesiology, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China.
| | - Hui Tao
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China; Department of Anesthesiology, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China.
| |
Collapse
|
24
|
Liu X, Yang B, Tan YF, Feng JG, Jia J, Yang CJ, Chen Y, Wang MH, Zhou J. The role of AMPK-Sirt1-autophagy pathway in the intestinal protection process by propofol against regional ischemia/reperfusion injury in rats. Int Immunopharmacol 2022; 111:109114. [PMID: 35933747 DOI: 10.1016/j.intimp.2022.109114] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/21/2022] [Accepted: 07/28/2022] [Indexed: 11/05/2022]
Abstract
Intestinal ischemia/reperfusion (II/R) is a clinical event associated with high morbidity and mortality. AMP-activated protein kinase (AMPK), a central cellular energy sensor, is associated with oxidative stress and inflammation. However, whether the AMPK is involved in the II/R-induced intestinal injury and the underlying mechanism is yet to be elucidated. Propofol has a protective effect on organs; yet, its specific mechanism of action remains unclear. This study explored the role of the AMPK-Sirt1-autophagy pathway in intestinal injury, and whether propofol could reduce intestinal injury and investigated the mechanisms in a rat model of II/R injury as well as a cell model (IEC-6 cells) of hypoxia/reoxygenation (H/R). Propofol, AMPK agonist (AICAR) and AMPK inhibitor (Compound C) were then administered, respectively. The histopathological changes, cell viability and apoptosis were detected. Furthermore, the levels of proinflammatory factors, the activities of oxidative stress, diamine oxidase, and signaling pathway were also analyzed. The results demonstrated that the AMPK-Sirt1-autophagy pathway of intestine was activated after II/R or H/R. Propofol could further activate the pathway, which reduced intestinal injury, inhibited apoptosis, reversed inflammation and oxidative stress, and improved the 24-hour survival rate in II/R rats in vivo, and attenuated H/R-induced IEC-6 cell injury, oxidative stress, and apoptosis in vitro, as fine as changes in AICAR treatment. Compound C abrogated the protective effect of propofol on II/R and H/R-induced injury. These results suggested a crucial effect of AMPK on the mechanism of intestinal injury and might provide a new insight into the mechanism of propofol reducing II/R injury.
Collapse
Affiliation(s)
- Xiao Liu
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China; Laboratory of Anesthesiology, Southwest Medical University, Luzhou, PR China
| | - Bo Yang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China; Laboratory of Anesthesiology, Southwest Medical University, Luzhou, PR China
| | - Ya-Fang Tan
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China; Laboratory of Anesthesiology, Southwest Medical University, Luzhou, PR China
| | - Jian-Guo Feng
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China; Laboratory of Anesthesiology, Southwest Medical University, Luzhou, PR China
| | - Jing Jia
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China; Laboratory of Anesthesiology, Southwest Medical University, Luzhou, PR China
| | - Cheng-Jie Yang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China; Laboratory of Anesthesiology, Southwest Medical University, Luzhou, PR China
| | - Ye Chen
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China; Laboratory of Anesthesiology, Southwest Medical University, Luzhou, PR China
| | - Mao-Hua Wang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China; Laboratory of Anesthesiology, Southwest Medical University, Luzhou, PR China.
| | - Jun Zhou
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China; Laboratory of Anesthesiology, Southwest Medical University, Luzhou, PR China.
| |
Collapse
|
25
|
Zhang ZT, He WJ, Deng SM, Xu SH, Zeng X, Qian ZM, Chen ZQ, Wang SM, Tang D. Trilobatin alleviates non-alcoholic fatty liver disease in high-fat diet plus streptozotocin-induced diabetic mice by suppressing NLRP3 inflammasome activation. Eur J Pharmacol 2022; 933:175291. [PMID: 36150533 DOI: 10.1016/j.ejphar.2022.175291] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/09/2022]
Abstract
Diabetes mellitus (DM) is a factor with great risk in the course of non-alcoholic fatty liver disease (NAFLD) due to its high glucotoxicity and lipotoxicity. Trilobatin, a glycosylated dihydrochalcone derived from the leaves of the Chinese sweet tea Lithocarpus polystachyus Rehd, is reported to possess various pharmacological activities. Nevertheless, it is still unclear regarding if trilobatin can alleviate liver injury in diabetic mice with NAFLD and its mechanism. Our aim was to investigative the protective effects of trilobatin against DM with NAFLD and its mechanism of action. A DM mice model was established by high-fat diet (HFD) feeding with streptozocin (STZ) injections, and treated with trilobatin for 10 weeks. The biochemical results showed that trilobatin restored glucose metabolic disorder and liver function in diabetic mice. The histopathological evaluation revealed that trilobatin improved liver injury by alleviating lipid accumulation and liver fibrosis. Mechanistically, trilobatin decreased expression of NLRP3, p65 NF-κB, cleaved-Caspase-1 and N-GSDMD, as well as the release of IL-18 and IL-1β, leading to a alleviation of inflammation and pyroptosis. Taken together, we determined for the first time found that trilobatin could prevent liver injury in diabetic mice with NAFLD by suppressing NLRP3 inflammasome activation to reduce inflammation and pyroptosis.
Collapse
Affiliation(s)
- Zhi-Tong Zhang
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering & Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Wen-Jiao He
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering & Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Si-Min Deng
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering & Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Shu-Hong Xu
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering & Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xia Zeng
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering & Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | | | - Zhi-Quan Chen
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, 530021, China
| | - Shu-Mei Wang
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering & Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Dan Tang
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering & Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
26
|
7-Hydroxyflavone Alleviates Myocardial Ischemia/Reperfusion Injury in Rats by Regulating Inflammation. Molecules 2022; 27:molecules27175371. [PMID: 36080137 PMCID: PMC9458087 DOI: 10.3390/molecules27175371] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 11/18/2022] Open
Abstract
Inflammation is the primary pathological process of myocardial ischemia/reperfusion injury (MI/RI). 7-Hydroxyflavone (HF), a natural flavonoid with a variety of bioactivities, plays a crucial role in various biological processes. However, its cardioprotective effects and the underlying mechanisms of MI/RI have not been investigated. This study aimed to explore whether pretreatment with HF could attenuate MI/RI-induced inflammation in rats and investigate its potential mechanisms. The results showed that pretreatment with HF could significantly improve the anatomic data and electrocardiograph parameters, reduce the myocardial infarct size, decrease markers of myocardial injury (aspartate transaminase, creatine kinase, lactate dehydrogenase, and cardiac troponin I), inhibit inflammatory cytokines (IL-1β, IL-6, and TNF-α), suppress oxidative stress, and recover the architecture of the cardiomyocytes. The cardioprotective effect of HF was connected with the regulation of the MAPK/NF-κB signaling pathway. What is more, molecular docking was carried out to prove that HF could be stably combined with p38, ERK1/2, JNK, and NF-κB. In summary, this is a novel study demonstrating the cardioprotective effects of HF against MI/RI in vivo. Consequently, these results demonstrate that HF can be considered a promising potential therapy for MI/RI.
Collapse
|
27
|
Li Y, Gao Y, Li G. Preclinical multi-target strategies for myocardial ischemia-reperfusion injury. Front Cardiovasc Med 2022; 9:967115. [PMID: 36072870 PMCID: PMC9444048 DOI: 10.3389/fcvm.2022.967115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
Despite promising breakthroughs in diagnosing and treating acute coronary syndromes, cardiovascular disease’s high global mortality rate remains indisputable. Nearly half of these patients died of ischemic heart disease. Primary percutaneous coronary intervention (PCI) and coronary artery bypass grafting can rapidly restore interrupted blood flow and become the most effective method for salvaging viable myocardium. However, restoring blood flow could increase the risk of other complications and myocardial cell death attributed to myocardial ischemia-reperfusion injury (IRI). How to reduce the damage of blood reperfusion to ischemic myocardium has become an urgent problem to be solved. In preclinical experiments, many treatments have substantial cardioprotective effects against myocardial IRI. However, the transition from these cardioprotective therapies to clinically beneficial therapies for patients with acute myocardial infarction remains elusive. The reasons for the failure of the clinical translation may be multi-faceted, and three points are summarized here: (1) Our understanding of the complex pathophysiological mechanisms of myocardial IRI is far from enough, and the classification of specific therapeutic targets is not rigorous, and not clear enough; (2) Most of the clinical patients have comorbidities, and single cardioprotective strategies including ischemia regulation strategies cannot exert their due cardioprotective effects under conditions of hyperglycemia, hypertension, hyperlipidemia, and aging; (3) Most preclinical experimental results are based on adult, healthy animal models. However, most clinical patients had comorbidities and received multiple drug treatments before reperfusion therapy. In 2019, COST Action proposed a multi-target drug combination initiative for prospective myocardial IRI; the optimal cardioprotective strategy may be a combination of additive or synergistic multi-target therapy, which we support. By establishing more reasonable preclinical models, screening multi-target drug combinations more in line with clinical practice will benefit the translation of clinical treatment strategies.
Collapse
|
28
|
Jia Y, Li D, Yu J, Jiang W, Liao X, Zhao Q. Potential diabetic cardiomyopathy therapies targeting pyroptosis: A mini review. Front Cardiovasc Med 2022; 9:985020. [PMID: 36061533 PMCID: PMC9433721 DOI: 10.3389/fcvm.2022.985020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022] Open
Abstract
Pyroptosis is primarily considered a pro-inflammatory class of caspase-1- and gasdermin D (GSDMD)-dependent programmed cell death. Inflammasome activation promotes the maturation and release of interleukin (IL)-1β and IL-18, cleavage of GSDMD, and development of pyroptosis. Recent studies have reported that NLRP3 inflammasome activation-mediated pyroptosis aggravates the formation and development of diabetes cardiomyopathy (DCM). These studies provide theoretical mechanisms for exploring a novel approach to treat DCM-associated cardiac dysfunction. Accordingly, this review aims to summarize studies that investigated possible DCM therapies targeting pyroptosis and elucidate the molecular mechanisms underlying NLRP3 inflammasome-mediated pyroptosis, and its potential association with the pathogenesis of DCM. This review may serve as a basis for the development of potential pharmacological agents as novel and effective treatments for managing and treating DCM.
Collapse
Affiliation(s)
- Yu Jia
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Dongze Li
- Department of Emergency Medicine and National Clinical Research Center for Geriatrics, Disaster Medicine Center, West China Hospital, Sichuan University West China School of Medicine, Chengdu, China
| | - Jing Yu
- Department of Emergency Medicine and National Clinical Research Center for Geriatrics, Disaster Medicine Center, West China Hospital, Sichuan University West China School of Medicine, Chengdu, China
| | - Wenli Jiang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Xiaoyang Liao
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Zhao
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Qian Zhao,
| |
Collapse
|
29
|
Current knowledge of pyroptosis in heart diseases. J Mol Cell Cardiol 2022; 171:81-89. [PMID: 35868567 DOI: 10.1016/j.yjmcc.2022.07.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/22/2022] [Accepted: 07/13/2022] [Indexed: 10/17/2022]
Abstract
Pyroptosis is a form of pro-inflammatory, necrotic cell death mediated by proteins of the gasdermin family. Various heart diseases, including myocardial ischemia/reperfusion injury, myocardial infarction, and heart failure, involve cardiomyocyte and non-myocyte pyroptosis. Cardiomyocyte pyroptosis also causes the release of pro-inflammatory cytokines. Recent studies have confirmed that pyroptosis is predominantly triggered by both the canonical and non-canonical inflammasome pathways, which independently facilitate caspase-1 or caspase-11/4/5 activation and gasdermin D (GSDMD) cleavage. Cardiac fibroblast and myeloid cell pyroptosis also contributes to the pathogenesis and development of heart diseases. This review summarizes the recent studies on pyroptosis in heart diseases and discusses the associated therapeutic targets.
Collapse
|
30
|
Li X, Xiao GY, Guo T, Song YJ, Li QM. Potential therapeutic role of pyroptosis mediated by the NLRP3 inflammasome in type 2 diabetes and its complications. Front Endocrinol (Lausanne) 2022; 13:986565. [PMID: 36387904 PMCID: PMC9646639 DOI: 10.3389/fendo.2022.986565] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/13/2022] [Indexed: 11/25/2022] Open
Abstract
As a new way of programmed cell death, pyroptosis plays a vital role in many diseases. In recent years, the relationship between pyroptosis and type 2 diabetes (T2D) has received increasing attention. Although the current treatment options for T2D are abundant, the occurrence and development of T2D appear to continue, and the poor prognosis and high mortality of patients with T2D remain a considerable burden in the global health system. Numerous studies have shown that pyroptosis mediated by the NLRP3 inflammasome can affect the progression of T2D and its complications; targeting the NLRP3 inflammasome has potential therapeutic effects. In this review, we described the molecular mechanism of pyroptosis more comprehensively, discussed the most updated progress of pyroptosis mediated by NLRP3 inflammasome in T2D and its complications, and listed some drugs and agents with potential anti-pyroptosis effects. Based on the available evidence, exploring more mechanisms of the NLRP3 inflammasome pathway may bring more options and benefits for preventing and treating T2D and drug development.
Collapse
|