1
|
Hu XM, Zheng S, Zhang Q, Wan X, Li J, Mao R, Yang R, Xiong K. PANoptosis signaling enables broad immune response in psoriasis: From pathogenesis to new therapeutic strategies. Comput Struct Biotechnol J 2024; 23:64-76. [PMID: 38125299 PMCID: PMC10730955 DOI: 10.1016/j.csbj.2023.11.049] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 11/24/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
Background Accumulating evidence suggests that regulated cell death, such as pyroptosis, apoptosis, and necroptosis, is deeply involved in the pathogenesis of psoriasis. As a newly recognized form of systematic cell death, PANoptosis is involved in a variety of inflammatory disorders through amplifying inflammatory and immune cascades, but its role in psoriasis remains elusive. Objectives To reveal the role of PANoptosis in psoriasis for a potential therapeutic strategy. Methods Multitranscriptomic analysis and experimental validation were used to identify PANoptosis signaling in psoriasis. RNA-seq and scRNA-seq analyses were performed to establish a PANoptosis-mediated immune response in psoriasis, which revealed hub genes through WGCNA and predicted disulfiram as a potential drug. The effect and mechanism of disulfiram were verified in imiquimod (IMQ)-induced psoriasis. Results Here, we found a highlighted PANoptosis signature in psoriasis patients through multitranscriptomic analysis and experimental validation. Based on this, two distinct PANoptosis patterns (non/high) were identified, which were the options for clinical classification. The high-PANoptosis-related group had a higher response rate to immune cell infiltration (such as M1 macrophages and keratinocytes). Subsequently, WGCNA showed the hub genes (e.g., S100A12, CYCS, NOD2, STAT1, HSPA4, AIM2, MAPK7), which were significantly associated with clinical phenotype, PANoptosis signature, and identified immune response in psoriasis. Finally, we explored disulfiram (DSF) as a candidate drug for psoriasis through network pharmacology, which ameliorated IMQ-mediated psoriatic symptoms through antipyroptosis-mediated inflammation and enhanced apoptotic progression. By analyzing the specific ligand-receptor interaction pairs within and between cell lineages, we speculated that DSF might exert its effects by targeting keratinocytes directly or targeting M1 macrophages to downregulate the proliferation of keratinocytes. Conclusions PANoptosis with its mediated immune cell infiltration provides a roadmap for research on the pathogenesis and therapeutic strategies of psoriasis.
Collapse
Affiliation(s)
- Xi-min Hu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, China
| | - Shengyuan Zheng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qi Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, China
| | - Xinxing Wan
- Department of Endocrinology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Rui Mao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ronghua Yang
- Department of Burn and Plastic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510000, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha 410008, China
- Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
2
|
Wen W, Cao Y, Chen P, Li J, Li W, Huang G, Zheng H, Zhu X, Zhang H, Chen Y, Huang X, Hu Y, Huang Y. A reliable strategy for establishment of an animal model of diabetic cardiomyopathy: Induction by a high-fat diet combined with single or multiple injections of low-dose streptozotocin. Life Sci 2024; 358:123161. [PMID: 39433084 DOI: 10.1016/j.lfs.2024.123161] [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: 03/07/2024] [Revised: 09/04/2024] [Accepted: 10/17/2024] [Indexed: 10/23/2024]
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) is one of the leading causes of death in patients with diabetes mellitus (DM). This study aimed to identify a reliable method for establishing an animal model of DCM for investigation of new targets and treatments. METHODS Eighty-four 4-week-old male Sprague-Dawley rats were randomly allocated to receive a normal diet or a high-fat diet (HFD) in an approximate ratio of 1:3. At 9 weeks of age, rats in the HFD group received streptozotocin (STZ) 30 mg/kg by intraperitoneal injection and rats in the control group received the same volume of buffer solution. The rodent model of DM was deemed to be successfully established when a random blood glucose measurement was >16.7 mmol/L on three consecutive occasions. If necessary, STZ was readministered. RESULTS Three of the 64 rats in the HFD group died after a second STZ injection. DM was induced in 14, 39, and 8 rats after one, two, and three injections, respectively, with cumulative success rates of 21.9 %, 82.8 %, and 95.3 %. Three months later, the rats with DM showed persistent hyperglycemia and insulin resistance and developed histopathological changes indicating cardiac hypertrophy, myocardial fibrosis, and diastolic dysfunction. The metabolic and cardiac histopathological changes were consistent regardless of whether DM was induced by one, two, or three injections of STZ. CONCLUSION An HFD combined with one or more intraperitoneal injections of low-dose STZ is a straightforward and reliable method for inducing DCM in rats. When a single dose of STZ fails to induce DM, repeated injections can be considered.
Collapse
Affiliation(s)
- Weixing Wen
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde) NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China.
| | - Yue Cao
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde) NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China.
| | - Peng Chen
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde) NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China.
| | - Jiahuan Li
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde) NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China.
| | - Weiwen Li
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde) NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China.
| | - Guolin Huang
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde) NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China.
| | - Haoxiao Zheng
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde) NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China.
| | - Xiaolin Zhu
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde) NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China.
| | - Hao Zhang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Yangxin Chen
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Xiaohui Huang
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde) NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China.
| | - Yunzhao Hu
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde) NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China.
| | - Yuli Huang
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, Australia; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation Research, Guangzhou, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde) NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, 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
|
Mitsis A, Khattab E, Myrianthefs M, Tzikas S, Kadoglou NPE, Fragakis N, Ziakas A, Kassimis G. Chemerin in the Spotlight: Revealing Its Multifaceted Role in Acute Myocardial Infarction. Biomedicines 2024; 12:2133. [PMID: 39335646 PMCID: PMC11428948 DOI: 10.3390/biomedicines12092133] [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: 08/24/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Chemerin, an adipokine known for its role in adipogenesis and inflammation, has emerged as a significant biomarker in cardiovascular diseases, including acute myocardial infarction (AMI). Recent studies have highlighted chemerin's involvement in the pathophysiological processes of coronary artery disease (CAD), where it modulates inflammatory responses, endothelial function, and vascular remodelling. Elevated levels of chemerin have been associated with adverse cardiovascular outcomes, including increased myocardial injury, left ventricular dysfunction, and heightened inflammatory states post-AMI. This manuscript aims to provide a comprehensive review of the current understanding of chemerin's role in AMI, detailing its molecular mechanisms, clinical implications, and potential as a biomarker for diagnosis and prognosis. Additionally, we explore the therapeutic prospects of targeting chemerin pathways to mitigate myocardial damage and improve clinical outcomes in AMI patients. By synthesizing the latest research findings, this review seeks to elucidate the multifaceted role of chemerin in AMI and its promise as a target for innovative therapeutic strategies.
Collapse
Affiliation(s)
- Andreas Mitsis
- Cardiology Department, Nicosia General Hospital, State Health Services Organization, Nicosia 2029, Cyprus; (E.K.); (M.M.)
| | - Elina Khattab
- Cardiology Department, Nicosia General Hospital, State Health Services Organization, Nicosia 2029, Cyprus; (E.K.); (M.M.)
| | - Michael Myrianthefs
- Cardiology Department, Nicosia General Hospital, State Health Services Organization, Nicosia 2029, Cyprus; (E.K.); (M.M.)
| | - Stergios Tzikas
- Third Department of Cardiology, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | | | - Nikolaos Fragakis
- Second Department of Cardiology, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (N.F.); (G.K.)
| | - Antonios Ziakas
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - George Kassimis
- Second Department of Cardiology, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (N.F.); (G.K.)
| |
Collapse
|
5
|
Yang Q, Chen Q, Li S, Luo J. Mesenchymal stem cells ameliorate inflammation and pyroptosis in diabetic cardiomyopathy via the miRNA-223-3p/NLRP3 pathway. Diabetol Metab Syndr 2024; 16:146. [PMID: 38956716 PMCID: PMC11221100 DOI: 10.1186/s13098-024-01389-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) stands as the primary cause of heart failure and mortality among patients with diabetes. Nevertheless, conventional treatment approaches are limited in their ability to effectively prevent myocardial tissue damage itself. Mesenchymal stem cell (MSC) therapy exhibits immense potential for treating DCM; however, the precise mechanisms involved in regulating inflammatory responses and pyroptosis processes, an emerging form of cellular death, within myocardial cells remain elusive. Hence, it is imperative to further elucidate the precise underlying mechanisms to facilitate the clinical implementation of MSC therapy. METHODS In vivo, we established a DCM mouse model by administering streptozotocin and fed the mice a high-glucose and high-fat diet, followed by MSC therapy. Cardiac function and myocardial injury were evaluated through echocardiography and histological analysis. Furthermore, the levels of inflammation and pyroptosis were assessed using ELISA, Western blotting, and qRT-PCR. In vitro experiments involved inducing H9C2 myocardial cell damage with high glucose treatment, followed by coculture with MSCs to investigate their role in modulating inflammation and pyroptosis mechanisms. RESULTS MSCs can maintain cardiac function and alleviate myocardial injury in mice with DCM. Moreover, they effectively suppress the activation of NLRP3 and reduce the release of inflammatory factors (such as IL-1β and ROS), thereby further downregulating the expression of pyroptosis-related proteins including NLRP3, Caspase-1, and GSDMD. Additionally, we experimentally validated that MSCs exert their therapeutic effects by promoting the expression of miR-223-3p in cardiac myocytes; however, this effect can be reversed by an miR-223-3p inhibitor. CONCLUSION MSCs effectively mitigate the release of inflammatory factors and cell lysis caused by pyroptosis through the regulation of the miR-223-3p/NLRP3 pathway, thereby safeguarding cardiomyocytes against damage in DCM. This mechanism establishes a novel theoretical foundation for the clinical treatment of cardiac conditions utilizing MSCs.
Collapse
Affiliation(s)
- Qu Yang
- Department of Rehabilitation Medicine, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Qi Chen
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Sihui Li
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Jun Luo
- Department of Rehabilitation Medicine, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
6
|
Zhang B, Wu H, Zhang J, Cong C, Zhang L. The study of the mechanism of non-coding RNA regulation of programmed cell death in diabetic cardiomyopathy. Mol Cell Biochem 2024; 479:1673-1696. [PMID: 38189880 DOI: 10.1007/s11010-023-04909-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/25/2023] [Indexed: 01/09/2024]
Abstract
Diabetic cardiomyopathy (DCM) represents a distinct myocardial disorder elicited by diabetes mellitus, characterized by aberrations in myocardial function and structural integrity. This pathological condition predominantly manifests in individuals with diabetes who do not have concurrent coronary artery disease or hypertension. An escalating body of scientific evidence substantiates the pivotal role of programmed cell death (PCD)-encompassing apoptosis, autophagy, pyroptosis, ferroptosis, and necroptosis-in the pathogenic progression of DCM, thereby emerging as a prospective therapeutic target. Additionally, numerous non-coding RNAs (ncRNAs) have been empirically verified to modulate the biological processes underlying programmed cell death, consequently influencing the evolution of DCM. This review systematically encapsulates prevalent types of PCD manifest in DCM as well as nascent discoveries regarding the regulatory influence of ncRNAs on programmed cell death in the pathogenesis of DCM, with the aim of furnishing novel insights for the furtherance of research in PCD-associated disorders relevant to DCM.
Collapse
Affiliation(s)
- Bingrui Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine Cardiovascular Department Cardiovascular Disease Research, Jinan, 250014, Shandong, China
| | - Hua Wu
- Tai'an Special Care Hospital Clinical Laboratory Medical Laboratory Direction, Tai'an, 271000, Shandong, China
| | - Jingwen Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine Cardiovascular Department Cardiovascular Disease Research, Jinan, 250014, Shandong, China
| | - Cong Cong
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine Cardiovascular Department Cardiovascular Disease Research, Jinan, 250014, Shandong, China
| | - Lin Zhang
- Tai'an Hospital of Chinese Medicine Cardiovascular Department Cardiovascular Disease Research, No.216, Yingxuan Street, Tai'an, 271000, Shandong, China.
| |
Collapse
|
7
|
Liu M, Zeng C, Zhang Y, Xin Y, Deng S, Hu X. Protective role of hydrogen sulfide against diabetic cardiomyopathy by inhibiting pyroptosis and myocardial fibrosis. Biomed Pharmacother 2024; 175:116613. [PMID: 38657502 DOI: 10.1016/j.biopha.2024.116613] [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: 02/12/2024] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 04/26/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) contributes significantly to the heightened mortality rate observed among diabetic patients, with myocardial fibrosis (MF) being a pivotal element in the disease's progression. Hydrogen sulfide (H2S) has been shown to mitigate MF, but the specific underlying mechanisms have yet to be thoroughly understood. A connection has been established between the evolution of DCM and the incidence of cardiomyocyte pyroptosis. Our research offers insights into H2S protective impact and its probable mode of action against DCM, analyzed through the lens of MF. In this study, a diabetic rat model was developed using intraperitoneal injections of streptozotocin (STZ), and hyperglycemia-stimulated cardiomyocytes were employed to replicate the cellular environment of DCM. There was a marked decline in the expression of cystathionine γ-lyase (CSE), a catalyst for H2S synthesis, in both the STZ-induced diabetic rats and hyperglycemia-stimulated cardiomyocytes. Experimental results in vivo indicated that H2S ameliorates MF and enhances cardiac functionality in diabetic rats by mitigating cardiomyocyte pyroptosis. In vitro assessments highlighted the induction of cardiomyocyte pyroptosis and the subsequent decline in cell viability under hyperglycemic conditions. However, the administration of sodium hydrosulfide (NaHS) curtailed cardiomyocyte pyroptosis and augmented cell viability. In contrast, propargylglycine (PAG), a CSE inhibitor, reversed the effects rendered by NaHS administration. Additional exploration indicated that the mitigating effect of H2S on cardiomyocyte pyroptosis is modulated through the ROS/NLRP3 pathway. In essence, our findings corroborate the potential of H2S in alleviating MF in diabetic subjects. This therapeutic effect is likely attributable to the regulation of cardiomyocyte pyroptosis via the ROS/NLRP3 pathway. This discovery furnishes a prospective therapeutic target for the amelioration and management of MF associated with diabetes.
Collapse
Affiliation(s)
- Maojun Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital,Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011,China
| | - Cheng Zeng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital,Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011,China
| | - Yifeng Zhang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital,Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011,China
| | - Ying Xin
- Department of Cardiovascular Medicine, The Second Xiangya Hospital,Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011,China
| | - Simin Deng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital,Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011,China
| | - Xinqun Hu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital,Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011,China.
| |
Collapse
|
8
|
Waghode P, Quadir SS, Choudhary D, Sharma S, Joshi G. Small interfering RNA (siRNA) as a potential gene silencing strategy for diabetes and associated complications: challenges and future perspectives. J Diabetes Metab Disord 2024; 23:365-383. [PMID: 38932822 PMCID: PMC11196550 DOI: 10.1007/s40200-024-01405-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/17/2024] [Indexed: 06/28/2024]
Abstract
Objective This article critically reviews the recent search on the use of Small Interfering RNA (siRNA) in the process of gene regulation that has been harnessed to silence specific genes in various cell types, including those involved in diabetes complications. Significance Diabetes, a prevalent and severe condition, poses life-threatening risks due to elevated blood glucose levels. It results from inadequate insulin production by the pancreas or ineffective insulin utilization by the body. Recent research suggests siRNA could hold promise in addressing diabetes complications. Methods In this review, we discussed several subjects, including diabetes; its function, and common treatment options. An in-depth analysis of gene silencing method for siRNA and role of siRNA in diabetes, focusing on its impact on glucose homeostasis, diabetic retinopathy, wound healing, diabetic nephropathy and peripheral neuropathy, diabetic foot ulcers, diabetic atherosclerosis, and diabetic cardiomyopathy. Result siRNA-based treatment has the potential to target specific genes without disrupting several other endogenous pathways, which decreases the risk of off-target effects. In addition, siRNA has the capability to provide long-term efficacy with a single dose which will reduce treatment options and enhance patient compliance. Conclusion In the context of diabetic complications, siRNA has been explored as a potential therapeutic tool to modulate the expression of genes involved in various processes associated with diabetes-related issues such as Diabetic Retinopathy, Neuropathy, Nephropathy, wound healing. The use of siRNA in these contexts is still largely experimental, and challenges such as delivery to specific tissues, potential off-target effects, and long-term safety need to be addressed. Additionally, the development of siRNA-based therapies for clinical use in diabetic complications is an active area of research. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-024-01405-7.
Collapse
Affiliation(s)
- Pranali Waghode
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM’s NMIMS, deemed to be University, Vile Parle West, 400056 Mumbai, Maharashtra India
| | - Sheikh Shahnawaz Quadir
- Department of Pharmaceutical Sciences, Mohanlal Sukhadia University, 313001 Udaipur, Rajasthan India
| | - Deepak Choudhary
- Department of Pharmaceutical Sciences, Mohanlal Sukhadia University, 313001 Udaipur, Rajasthan India
| | - Sanjay Sharma
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM’s NMIMS, deemed to be University, Vile Parle West, 400056 Mumbai, Maharashtra India
| | - Garima Joshi
- Department of Pharmaceutical Sciences, Mohanlal Sukhadia University, 313001 Udaipur, Rajasthan India
| |
Collapse
|
9
|
Liu Z, Chen Y, Chen Y, Zheng J, Wu W, Wang L, Wang H, Yu Y. Effect of Regulation of Chemerin/Chemokine-like Receptor 1/Stimulator of Interferon Genes Pathway on Astrocyte Recruitment to Aβ Plaques. Int J Mol Sci 2024; 25:4324. [PMID: 38673909 PMCID: PMC11049903 DOI: 10.3390/ijms25084324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/03/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Recruitment and accumulation of reactive astrocytes around senile plaques are common pathological features of Alzheimer's disease (AD), with unclear mechanisms. Chemerin, an adipokine implicated in neuroinflammation, acts through its receptor, chemokine-like receptor 1 (CMKLR1), which also functions as a receptor for amyloid β (Aβ). The impact of the chemerin/CMKLR1 axis on astrocyte migration towards Aβ plaques is unknown. Here we investigated the effect of CMKLR1 on astrocyte migration around Aβ deposition in APP/PS1 mice with Cmklr1 knockout (APP/PS1-Cmklr1-/-). CMKLR1-expressed astrocytes were upregulated in the cortices and hippocampi of 9-month-old APP/PS1 mice. Chemerin mainly co-localized with neurons, and its expression was reduced in the brains of APP/PS1 mice, compared to WT mice. CMKLR1 deficiency decreased astrocyte colocalization with Aβ plaques in APP/PS1-Cmklr1-/- mice, compared to APP/PS1 mice. Activation of the chemerin/CMKLR1 axis promoted the migration of primary cultured astrocytes and U251 cells, and reduced astrocyte clustering induced by Aβ42. Mechanistic studies revealed that chemerin/CMKLR1 activation induced STING phosphorylation. Deletion of STING attenuated the promotion of the chemerin/CMKLR1 axis relative to astrocyte migration and abolished the inhibitory effect of chemerin on Aβ42-induced astrocyte clustering. These findings suggest the involvement of the chemerin/CMKLR1/STING pathway in the regulation of astrocyte migration and recruitment to Aβ plaques/Aβ42.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yang Yu
- Engineering Research Center of Cell and Therapeutic Antibody Medicine, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (Z.L.); (Y.C.); (Y.C.); (J.Z.); (W.W.); (L.W.); (H.W.)
| |
Collapse
|
10
|
Ding P, Song Y, Yang Y, Zeng C. NLRP3 inflammasome and pyroptosis in cardiovascular diseases and exercise intervention. Front Pharmacol 2024; 15:1368835. [PMID: 38681198 PMCID: PMC11045953 DOI: 10.3389/fphar.2024.1368835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/02/2024] [Indexed: 05/01/2024] Open
Abstract
NOD-like receptor protein 3 (NLRP3) inflammasome is an intracellular sensing protein complex that possesses NACHT, leucine-rich repeat, and pyrin domain, playing a crucial role in innate immunity. Activation of the NLRP3 inflammasome leads to the production of pro-inflammatory cellular contents, such as interleukin (IL)-1β and IL-18, and induction of inflammatory cell death known as pyroptosis, thereby amplifying or sustaining inflammation. While a balanced inflammatory response is beneficial for resolving damage and promoting tissue healing, excessive activation of the NLRP3 inflammasome and pyroptosis can have harmful effects. The involvement of the NLRP3 inflammasome has been observed in various cardiovascular diseases (CVD). Indeed, the NLRP3 inflammasome and its associated pyroptosis are closely linked to key cardiovascular risk factors including hyperlipidemia, diabetes, hypertension, obesity, and hyperhomocysteinemia. Exercise compared with medicine is a highly effective measure for both preventing and treating CVD. Interestingly, emerging evidence suggests that exercise improves CVD and inhibits the activity of NLRP3 inflammasome and pyroptosis. In this review, the activation mechanisms of the NLRP3 inflammasome and its pathogenic role in CVD are critically discussed. Importantly, the purpose is to emphasize the crucial role of exercise in managing CVD by suppressing NLRP3 inflammasome activity and proposes it as the foundation for developing novel treatment strategies.
Collapse
Affiliation(s)
- Ping Ding
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuanming Song
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yang Yang
- Zhuhai People’s Hospital, Zhuhai Clinical Medical College of Jinan University, Zhuhai, China
| | - Cheng Zeng
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
11
|
Yang L, Long Y, Xiao S. Osteosarcoma-Associated Immune Genes as Potential Immunotherapy and Prognosis Biomarkers. Biochem Genet 2024; 62:798-813. [PMID: 37452172 DOI: 10.1007/s10528-023-10444-3] [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: 03/26/2023] [Accepted: 06/29/2023] [Indexed: 07/18/2023]
Abstract
Immune-modulating therapies exhibit abundant promise compared to traditional treatment for osteosarcoma. We aim to establish an immune-related prognostic signatures in osteosarcoma. We identified the differentially expressed genes in osteosarcoma compared with normal controls using the GEO dataset. The intersection with immune-related genes was considered as differentially expressed immune genes. Potential prognosis-related differentially expressed genes were first analyzed with the multifactor Cox regression and then the step function performed the iteration. The best model was finally chosen as the immunological risk score signature model. And finally, we evaluated the correlation of genes in the prognostic model with immune cells, common immune checkpoints, and immune checkpoint blockade responses. We identified 1527 significantly upregulated and 2407 significantly downregulated genes in osteosarcoma compared to normal samples. In the 258 differentially expressed immune genes, 20 genes with independent prognostic significance were included in the step function. Finally, we constructed a prognostic signature for overall survival based on five immune genes (JAG2, PLXNB1, CMKLR1, NUDT6, and PSMC4) in osteosarcoma. These five genes are closely related to immune infiltration. Osteosarcoma with high JAG2 expression or low CMKLR1 expression may be associated with better immune checkpoint blockade response. JAG2 overexpression or CMKLR1 inhibition induced sensitivity to PD-1 antibody in osteosarcoma cells. We constructed a prognosis prediction signature containing five immune-related genes (JAG2, PLXNB1, CMKLR1, NUDT6, and PSMC4) with a significant prognostic value in osteosarcoma. Significant differences in immune infiltration and immunotherapy responses were identified between groups with different levels of these genes.
Collapse
Affiliation(s)
- Li Yang
- Key Laboratory of Geriatric Diseases of Xinyang, Institute of Inspection Technology, Xinyang Vocational and Technical College, Xinyang, 464000, China
| | - Yi Long
- Department of Joint Surgery, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou, 412000, China.
| | - Shengshi Xiao
- Department of Joint Surgery, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou, 412000, China.
| |
Collapse
|
12
|
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
|
13
|
Song J, Zhang Y, Frieler RA, Andren A, Wood S, Tyrrell DJ, Sajjakulnukit P, Deng JC, Lyssiotis CA, Mortensen RM, Salmon M, Goldstein DR. Itaconate suppresses atherosclerosis by activating a Nrf2-dependent antiinflammatory response in macrophages in mice. J Clin Invest 2023; 134:e173034. [PMID: 38085578 PMCID: PMC10849764 DOI: 10.1172/jci173034] [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: 06/22/2023] [Accepted: 12/06/2023] [Indexed: 01/22/2024] Open
Abstract
Itaconate has emerged as a critical immunoregulatory metabolite. Here, we examined the therapeutic potential of itaconate in atherosclerosis. We found that both itaconate and the enzyme that synthesizes it, aconitate decarboxylase 1 (Acod1, also known as immune-responsive gene 1 [IRG1]), are upregulated during atherogenesis in mice. Deletion of Acod1 in myeloid cells exacerbated inflammation and atherosclerosis in vivo and resulted in an elevated frequency of a specific subset of M1-polarized proinflammatory macrophages in the atherosclerotic aorta. Importantly, Acod1 levels were inversely correlated with clinical occlusion in atherosclerotic human aorta specimens. Treating mice with the itaconate derivative 4-octyl itaconate attenuated inflammation and atherosclerosis induced by high cholesterol. Mechanistically, we found that the antioxidant transcription factor, nuclear factor erythroid 2-related factor 2 (Nrf2), was required for itaconate to suppress macrophage activation induced by oxidized lipids in vitro and to decrease atherosclerotic lesion areas in vivo. Overall, our work shows that itaconate suppresses atherogenesis by inducing Nrf2-dependent inhibition of proinflammatory responses in macrophages. Activation of the itaconate pathway may represent an important approach to treat atherosclerosis.
Collapse
Affiliation(s)
- Jianrui Song
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Yanling Zhang
- Department of Biochemistry and Molecular Biology, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Ryan A. Frieler
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Anthony Andren
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sherri Wood
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Daniel J. Tyrrell
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Alabama, USA
| | - Peter Sajjakulnukit
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
- University of Michigan Rogel Cancer Center
| | - Jane C. Deng
- Graduate Program in Immunology, and
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| | - Costas A. Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Richard M. Mortensen
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pharmacology
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes
| | | | - Daniel R. Goldstein
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Graduate Program in Immunology, and
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
14
|
Lin X, Qu J, Yin L, Wang R, Wang X. Aerobic exercise-induced decrease of chemerin improved glucose and lipid metabolism and fatty liver of diabetes mice through key metabolism enzymes and proteins. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159409. [PMID: 37871796 DOI: 10.1016/j.bbalip.2023.159409] [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: 07/20/2023] [Revised: 10/18/2023] [Accepted: 10/20/2023] [Indexed: 10/25/2023]
Abstract
Our previous studies have implicated an important role of adipokine chemerin in exercise-induced improvements of glycolipid metabolism and fatty liver in diabetes rat, but the underlying mechanisms remain unknown. This study first used an exogenous chemerin supplement to clarify the roles of decreased chemerin in exercised diabetes mice and possible mechanisms of glucose and lipid metabolism key enzymes and proteins [such as adipose triglyceride lipase (ATGL), lipoprotein lipase (LPL), phosphoenolpyruvate carboxykinase (PEPCK), and glucose transporter 4 (GLUT4)]. In addition, two kinds of adipose-specific chemerin knockout mice were generated to demonstrate the regulation of chemerin on glucose and lipid metabolism enzymes and proteins. We found that in diabetes mice, exercise-induced improvements of glucose and lipid metabolism and fatty liver, and exercise-induced increases of ATGL, LPL, and GLUT4 in liver, gastrocnemius and fat were reversed by exogenous chemerin. Furthermore, in chemerin knockdown mice, chemerin(-/-)∙adiponectin mice had lower body fat mass, improved blood glucose and lipid, and no fatty liver; while chemerin(-/-)∙fabp4 mice had hyperlipemia and unchanged body fat mass. Peroxisome proliferator-activated receptor γ (PPARγ), ATGL, LPL, GLUT4 and PEPCK in the liver and gastrocnemius had improve changes in chemerin(-/-)·adiponectin mice while deteriorated alterations in chemerin(-/-)·fabp4 mice, although PPARγ, ATGL, LPL, and GLUT4 increased in the fat of two kinds of chemerin(-/-) mice. CONCLUSIONS: Decreased chemerin exerts an important role in exercise-induced improvements of glucose and lipid metabolism and fatty liver in diabetes mice, which was likely to be through PPARγ mediating elevations of ATGL, LPL and GLUT4 in peripheral metabolic organs.
Collapse
Affiliation(s)
- Xiaojing Lin
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Jing Qu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Lijun Yin
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China.
| | - Xiaohui Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
15
|
Fu F, Luo H, Du Y, Chen Y, Tian K, Pan J, Li J, Wang N, Bao R, Jin H, Tong P, Ruan H, Wu C. AR/PCC herb pair inhibits osteoblast pyroptosis to alleviate diabetes-related osteoporosis by activating Nrf2/Keap1 pathway. J Cell Mol Med 2023; 27:3601-3613. [PMID: 37621124 PMCID: PMC10660633 DOI: 10.1111/jcmm.17928] [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: 05/17/2023] [Revised: 07/28/2023] [Accepted: 08/04/2023] [Indexed: 08/26/2023] Open
Abstract
Osteoporosis is a prevalent complication of diabetes, characterized by systemic metabolic impairment of bone mass and microarchitecture, particularly in the spine. Anemarrhenae Rhizoma/Phellodendri Chinensis Cortex (AR/PCC) herb pair has been extensively employed in Traditional Chinese Medicine to manage diabetes; however, its potential to ameliorate diabetic osteoporosis (DOP) has remained obscure. Herein, we explored the protective efficacy of AR/PCC herb pair against DOP using a streptozotocin (STZ)-induced rat diabetic model. Our data showed that AR/PCC could effectively reduce the elevated fasting blood glucose and reverse the osteoporotic phenotype of diabetic rats, resulting in significant improvements in vertebral trabecular area percentage, trabecular thickness and trabecular number, while reducing trabecular separation. Specifically, AR/PCC herb pair improved impaired osteogenesis, nerve ingrowth and angiogenesis. More importantly, it could mitigate the aberrant activation of osteoblast pyroptosis in the vertebral bodies of diabetic rats by reducing increased expressions of Nlrp3, Asc, Caspase1, Gsdmd and IL-1β. Mechanistically, AR/PCC activated antioxidant pathway through the upregulation of the antioxidant response protein Nrf2, while concurrently decreasing its negative feedback regulator Keap1. Collectively, our in vivo findings demonstrate that AR/PCC can inhibit osteoblast pyroptosis and alleviate STZ-induced rat DOP, suggesting its potential as a therapeutic agent for mitigating DOP.
Collapse
Affiliation(s)
- Fangda Fu
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Huan Luo
- Department of Pharmacy, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Yu Du
- The First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Yuying Chen
- The Fourth Clinical Medical College of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Kun Tian
- Department of OrthopaedicsThe First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Jin Pan
- Department of Architecture, School of ArchitectureChina Academy of ArtHangzhouChina
| | - Jian Li
- Department of OrthopaedicsHangzhou Ninth People's HospitalHangzhouChina
| | - Nani Wang
- Department of MedicineZhejiang Academy of Traditional Chinese MedicineHangzhouChina
| | - Ronghua Bao
- Hangzhou Fuyang Hospital of TCM Orthopedics and TraumatologyHangzhouChina
| | - Hongting Jin
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Peijian Tong
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Hongfeng Ruan
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Chengliang Wu
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| |
Collapse
|
16
|
You P, Chen H, Han W, Deng J. miR-200a-3p overexpression alleviates diabetic cardiomyopathy injury in mice by regulating autophagy through the FOXO3/Mst1/Sirt3/AMPK axis. PeerJ 2023; 11:e15840. [PMID: 37727684 PMCID: PMC10506579 DOI: 10.7717/peerj.15840] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/12/2023] [Indexed: 09/21/2023] Open
Abstract
Objective Hyperglycemia and insulin resistance or deficiency are characteristic features of diabetes. Diabetes is accompanied by cardiomyocyte hypertrophy, fibrosis and ventricular remodeling, and eventually heart failure. In this study, we established a diabetic cardiomyopathy (DCM) mouse model to explore the role and mechanism of miR-200a-3p in DCM. Methods We used db/db mice to simulate the animal model of DCM and the expression of miR-200a-3p was then examined by RT-qPCR. Tail vein injection of mice was done with rAAV-miR-200a-3p for 8 weeks, and cardiac function was assessed by cardiac ultrasound. The levels of myocardial tissue injury, fibrosis, inflammation, apoptosis and autophagy in mice were detected by histological staining, TUNEL and other molecular biological experiments. Results miR-200a-3p expression levels were significantly decreased in the myocardium of DCM mice. Diabetic mice developed cardiac dysfunction and presented pathological changes such as myocardial injury, myocardial interstitial fibrosis, cardiomyocyte apoptosis, autophagy, and inflammation. Overexpression of miR-200a-3p expression significantly ameliorated diabetes induced-cardiac dysfunction and myocardial injury, myocardial interstitial fibrosis, cardiomyocyte apoptosis, and inflammation, and enhanced autophagy. Mechanistically, miR-200a-3p interacted with FOXO3 to promote Mst1 expression and reduce Sirt3 and p-AMPK expression. Conclusion In type 2 diabetes, increased miR-200a-3p expression enhanced autophagy and participated in the pathogenic process of cardiomyopathy throug7 Mst1/Sirt3/AMPK axis regulation by its target gene FOXO3. This conclusion provides clues for the search of new gene targeted therapeutic approaches for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Penghua You
- Department of Cardiology, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Haichao Chen
- Department of Cardiology, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Wenqi Han
- Department of Cardiology, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Jizhao Deng
- Department of Cardiology, Shaanxi Provincial People’s Hospital, Xi’an, China
| |
Collapse
|
17
|
Yarovinsky TO, Su M, Chen C, Xiang Y, Tang WH, Hwa J. Pyroptosis in cardiovascular diseases: Pumping gasdermin on the fire. Semin Immunol 2023; 69:101809. [PMID: 37478801 PMCID: PMC10528349 DOI: 10.1016/j.smim.2023.101809] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/13/2023] [Accepted: 07/12/2023] [Indexed: 07/23/2023]
Abstract
Pyroptosis is a form of programmed cell death associated with activation of inflammasomes and inflammatory caspases, proteolytic cleavage of gasdermin proteins (forming pores in the plasma membrane), and selective release of proinflammatory mediators. Induction of pyroptosis results in amplification of inflammation, contributing to the pathogenesis of chronic cardiovascular diseases such as atherosclerosis and diabetic cardiomyopathy, and acute cardiovascular events, such as thrombosis and myocardial infarction. While engagement of pyroptosis during sepsis-induced cardiomyopathy and septic shock is expected and well documented, we are just beginning to understand pyroptosis involvement in the pathogenesis of cardiovascular diseases with less defined inflammatory components, such as atrial fibrillation. Due to the danger that pyroptosis represents to cells within the cardiovascular system and the whole organism, multiple levels of pyroptosis regulation have evolved. Those include regulation of inflammasome priming, post-translational modifications of gasdermins, and cellular mechanisms for pore removal. While pyroptosis in macrophages is well characterized as a dramatic pro-inflammatory process, pyroptosis in other cell types within the cardiovascular system displays variable pathways and consequences. Furthermore, different cells and organs engage in local and distant crosstalk and exchange of pyroptosis triggers (oxidized mitochondrial DNA), mediators (IL-1β, S100A8/A9) and antagonists (IL-9). Development of genetic tools, such as Gasdermin D knockout animals, and small molecule inhibitors of pyroptosis will not only help us fully understand the role of pyroptosis in cardiovascular diseases but may result in novel therapeutic approaches inhibiting inflammation and progression of chronic cardiovascular diseases to reduce morbidity and mortality from acute cardiovascular events.
Collapse
Affiliation(s)
- Timur O Yarovinsky
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Meiling Su
- Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, China
| | - Chaofei Chen
- Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, China
| | - Yaozu Xiang
- Shanghai East Hospital, Key Laboratory of Arrhythmias of the Ministry of Education of China, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Wai Ho Tang
- Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, China; School of Nursing and Health Studies, Hong Kong Metropolitan University, Kowloon, the Hong Kong Special Administrative Region of China
| | - John Hwa
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
18
|
Pan M. miR-22-3p ameliorates the symptoms of premature ovarian failure in mice by inhibiting CMKLR1 expression. CHINESE J PHYSIOL 2023; 66:200-208. [PMID: 37635479 DOI: 10.4103/cjop.cjop-d-23-00004] [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] [Indexed: 08/29/2023] Open
Abstract
Premature ovarian failure (POF) affects many adult women less than 40 years of age and leads to infertility. This study was aimed at exploring the improving effects of miR-22-3p on the symptoms of POF in mice by inhibiting chemokine-like receptor 1 (CMKLR1) expression. Female mice were intraperitoneally injected with cyclophosphamide to construct POF mice models. Lentiviral vectors containing miR-22-3p, short hairpin RNA (sh)-CMKLR1, and overexpression (oe)-CMKLR1, respectively, or in combination, were injected into the ovaries of both sides of POF mice. miR-22-3p and CMKLR1 expression in ovarian tissues of mice was assessed, and the targeting relationship between miR-22-3p and CMKLR1 was predicted and verified. Serum estradiol (E2), anti-Mullerian hormone, and follicle-stimulating hormone levels were assessed. Ovarian weight was weighed, and pathological changes and the number of primordial follicles, primary follicles, secondary follicles, and atresia follicles were observed. Apoptosis of ovarian tissues was determined. In ovarian tissues of POF mice, miR-22-3p expression was decreased while CMKLR1 expression was increased. miR-22-3p up-regulation or CMKLR1 down-regulation restored sex hormone levels, improved ovarian weight and the number of primordial follicles, primary follicles, and secondary follicles, and reduced the number of atresia follicle and ovarian granulosa cell apoptosis in POF mice. miR-22-3p targeted CMKLR1, and overexpressing CMKLR1 reversed the ameliorative effects of miR-22-3p overexpression on POF mice. Our research highlights that overexpressed miR-22-3p down-regulates CMKLR1 to ameliorate the symptoms of POF in mice. Therefore, the miR-22-3p/CMKLR1 axis could improve the symptoms of POF.
Collapse
Affiliation(s)
- Miaomiao Pan
- The First College of Clinical Medical Science, China Three Gorges University; Department of Gynecology, Central People's Hospital of Yichang, Yichang, China
| |
Collapse
|
19
|
Zielinski M, Chwalba A, Jastrzebski D, Ziora D. Adipokines in interstitial lung diseases. Respir Physiol Neurobiol 2023:104109. [PMID: 37393966 DOI: 10.1016/j.resp.2023.104109] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 06/20/2023] [Accepted: 06/29/2023] [Indexed: 07/04/2023]
Abstract
Interstitial lung diseases (ILD) are a heterogenic group of respiratory diseases with complex pathogenesis. A growing number of evidence suggests role of adipose tissue and it's hormones (adipokines) in pathogenesis of various disorders, including lung tissue diseases. The aim of this study was to assess the concentrations of selected adipokines and their receptors (apelin, adiponectin, chemerin, chemerin receptor - CMKLR1) in patients with IPF (idiopathic pulmonary fibrosis) and sarcoidosis in comparison to healthy controls. We found changes in adipokines concentrations in ILD. Adiponectin concentrations were higher in all respiratory diseases patients in comparison to healthy controls. Apelin concentration in ILD patients was higher then those in healthy subjects. The trend of chemerin and CMKLR1 concentrations were similar, with highest concentrations seen in sarcoidosis. The study shows a difference of adipokines concentrations between patients with ILD and healthy controls. Adipokines are a potential marker and therapeutic target in patients with IPF and sarcoidosis.
Collapse
Affiliation(s)
- M Zielinski
- Department of Lung Diseases and Tuberculosis, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia in Katowice, Poland.
| | - A Chwalba
- Department of Lung Diseases and Tuberculosis, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia in Katowice, Poland
| | - D Jastrzebski
- Department of Lung Diseases and Tuberculosis, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia in Katowice, Poland
| | - D Ziora
- Department of Lung Diseases and Tuberculosis, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia in Katowice, Poland
| |
Collapse
|
20
|
Jin X, Ma Y, Liu D, Huang Y. Role of pyroptosis in the pathogenesis and treatment of diseases. MedComm (Beijing) 2023; 4:e249. [PMID: 37125240 PMCID: PMC10130418 DOI: 10.1002/mco2.249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 02/16/2023] [Accepted: 03/07/2023] [Indexed: 05/02/2023] Open
Abstract
Programmed cell death (PCD) is regarded as a pathological form of cell death with an intracellular program mediated, which plays a pivotal role in maintaining homeostasis and embryonic development. Pyroptosis is a new paradigm of PCD, which has received increasing attention due to its close association with immunity and disease. Pyroptosis is a form of inflammatory cell death mediated by gasdermin that promotes the release of proinflammatory cytokines and contents induced by inflammasome activation. Recently, increasing evidence in studies shows that pyroptosis has a crucial role in inflammatory conditions like cardiovascular diseases (CVDs), cancer, neurological diseases (NDs), and metabolic diseases (MDs), suggesting that targeting cell death is a potential intervention for the treatment of these inflammatory diseases. Based on this, the review aims to identify the molecular mechanisms and signaling pathways related to pyroptosis activation and summarizes the current insights into the complicated relationship between pyroptosis and multiple human inflammatory diseases (CVDs, cancer, NDs, and MDs). We also discuss a promising novel strategy and method for treating these inflammatory diseases by targeting pyroptosis and focus on the pyroptosis pathway application in clinics.
Collapse
Affiliation(s)
- Xiangyu Jin
- Wuxi School of MedicineJiangnan UniversityJiangsuChina
| | - Yinchu Ma
- Wuxi School of MedicineJiangnan UniversityJiangsuChina
| | - Didi Liu
- Wuxi School of MedicineJiangnan UniversityJiangsuChina
| | - Yi Huang
- Wuxi School of MedicineJiangnan UniversityJiangsuChina
| |
Collapse
|
21
|
Zhao N, Hua W, Liu Q, Wang Y, Liu Z, Jin S, Wang B, Pang Y, Qi J, Song Y. MALAT1 knockdown alleviates the pyroptosis of microglias in diabetic cerebral ischemia via regulating STAT1 mediated NLRP3 transcription. Mol Med 2023; 29:44. [PMID: 37013491 PMCID: PMC10069069 DOI: 10.1186/s10020-023-00637-2] [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: 04/12/2022] [Accepted: 03/13/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND Dysregulated long non-coding RNAs participate in the development of diabetic cerebral ischemia. This study aimed to investigate the underlying mechanism of lncRNA MALAT1 in diabetic cerebral ischemia. METHOD Middle cerebral artery occlusion (MCAO) was performed to establish diabetic cerebral I/R in vivo. TTC and neurological deficits assessment were performed to assess cerebral ischemic injury. LDH was conducted to detect cytotoxicity. RT-qPCR and western blotting assays were applied to determine mRNA and protein expression. Flow cytometry was performed to detect the pyroptosis of BV2 cells. Immunofluorescence and FISH were conducted for subcellular localization of MALAT1 and STAT1. ELISA was performed to determine cytokine release. Dual luciferase reporter, RIP, and ChIP assays were used to validate the interaction between STAT1 and MALAT1/NLRP3. Diabetes aggravated cerebral injury in vivo and in vitro. Diabetic cerebral ischemia induced inflammatory response and inflammation-induced cell pyroptosis. RESULT MALAT1 was overexpressed in diabetic cerebral ischemia models in vivo and in vitro. However, knockdown of MALAT1 suppressed inflammatory response and the pyroptosis of BV2 cells. Moreover, MALAT1 interacted with STAT1 to transcriptionally activate NLRP3. Knockdown of STAT1 significantly reversed the effects of MALAT1. Furthermore, STAT1 promotes the MALAT1 transcription. MALAT1 interacts with STAT1 to promote the pyroptosis of microglias induced by diabetic cerebral ischemia through activating NLRP3 transcription. CONCLUSION Thus, knockdown of MALAT1 may be a potential promising therapy target for diabetic cerebral ischemia.
Collapse
Affiliation(s)
- Nan Zhao
- Department of Pathology, First Clinical Hospital, Harbin Medical University, No. 23 Post Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Wei Hua
- Department of Pathology, First Clinical Hospital, Harbin Medical University, No. 23 Post Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Qi Liu
- Department of Pathology, First Clinical Hospital, Harbin Medical University, No. 23 Post Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Yueying Wang
- Department of Pathology, First Clinical Hospital, Harbin Medical University, No. 23 Post Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Zhiyi Liu
- Department of Pathology, First Clinical Hospital, Harbin Medical University, No. 23 Post Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Sinan Jin
- Department of Pathology, First Clinical Hospital, Harbin Medical University, No. 23 Post Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Benshuai Wang
- Department of Pathology, First Clinical Hospital, Harbin Medical University, No. 23 Post Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Yuxin Pang
- Department of Pathology, First Clinical Hospital, Harbin Medical University, No. 23 Post Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Jiping Qi
- Department of Pathology, First Clinical Hospital, Harbin Medical University, No. 23 Post Street, Nangang District, Harbin, 150001, Heilongjiang, China.
| | - Yuejia Song
- Department of Endocrinology, First Clinical Hospital, Harbin Medical University, No. 23 Post Street, Nangang District, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
22
|
Zhang Z, Ding Y, Li J, Su S. Up-regulation of CMKLR1 in endometriosis and its relationship with inflammatory responses. Histol Histopathol 2023; 38:329-337. [PMID: 36156768 DOI: 10.14670/hh-18-523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Inflammation plays a critical role in the pathogenesis of endometriosis. We aimed to study the proinflammatory effect of Chemerin chemokine-like receptor 1 (CMKLR1) in patients with endometriosis. Sixty patients with endometriosis and 50 healthy controls were recruited in this study for the collection of endometrial samples and peritoneal fluid. The expression levels of CMKLR1, IL-6, MCP-1, and TNF-α in peritoneal fluid and endometrial tissues were detected by ELISA, qRT-PCR, and immunohistochemical staining. Human endometrial stromal cells (HESCs) were used to measure the Chemerin-induced CMKLR1 activation and inflammatory responses. CMKLR1 level was significantly up-regulated in peritoneal fluid and endometrial tissues in patients with endometriosis. Interestingly, CMKLR1 overexpression positively correlated with pro-inflammatory cytokines and chemokine in both peritoneal fluid and ectopic endometrium. Chemerin treatment increased the expression of CMKLR1, and aggravated inflammatory responses in HESCs. CMKLR1 is up-regulated in peritoneal fluid and endometrial tissues, and promotes the inflammatory responses in of endometriosis.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Gynecology, Zibo Central Hospital, Zibo, Shandong, China
| | - Yumei Ding
- Department of Gynecology, Zibo Central Hospital, Zibo, Shandong, China
| | - Junjie Li
- Department of Anesthesiology, Zibo Central Hospital, Zibo, Shandong, China.
| | - Shan Su
- Department of Gynecology, Zibo Central Hospital, Zibo, Shandong, China
| |
Collapse
|
23
|
Yue G, An Q, Xu X, Jin Z, Ding J, Hu Y, Du Q, Xu J, Xie R. The role of Chemerin in human diseases. Cytokine 2023; 162:156089. [PMID: 36463659 DOI: 10.1016/j.cyto.2022.156089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 11/01/2022] [Accepted: 11/15/2022] [Indexed: 12/05/2022]
Abstract
Chemerin is a protein encoded by the Rarres2 gene that acts through endocrine or paracrine regulation. Chemerin can bind to its receptor, regulate insulin sensitivity and adipocyte differentiation, and thus affect glucose and lipid metabolism. There is growing evidence that it also plays an important role in diseases such as inflammation and cancer. Chemerin has been shown to play a role in the pathogenesis of inflammatory and metabolic diseases caused by leukocyte chemoattractants in a variety of organs, but its biological function remains controversial. In conclusion, the exciting findings collected over the past few years clearly indicate that targeting Chemerin signaling as a biological target will be a major research goal in the future. This article reviews the pathophysiological roles of Chemerin in various systems and diseases,and expect to provide a rationale for its role as a clinical therapeutic target.
Collapse
Affiliation(s)
- Gengyu Yue
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Qimin An
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Xiaolin Xu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Zhe Jin
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Jianhong Ding
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Yanxia Hu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Qian Du
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Jingyu Xu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China; The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi 563000, China.
| | - Rui Xie
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China; The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi 563000, China.
| |
Collapse
|
24
|
Wei Z, pinfang K, jing Z, zhuoya Y, Shaohuan Q, Chao S. Curcumin Improves Diabetic Cardiomyopathy by Inhibiting Pyroptosis through AKT/Nrf2/ARE Pathway. Mediators Inflamm 2023; 2023:3906043. [PMID: 37101595 PMCID: PMC10125772 DOI: 10.1155/2023/3906043] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 02/15/2023] [Accepted: 03/04/2023] [Indexed: 04/28/2023] Open
Abstract
This study is aimed at exploring whether curcumin can regulate the AKT pathway, promote the transfer of Nrf2 into the nucleus, and inhibit cell pyroptosis in diabetic cardiomyopathy. Diabetic rats and cardiomyocytes were treated with curcumin to study its effect on myocardial pyroptosis. Whether curcumin can promote the transfer of Nrf2 into the nucleus through AKT pathway regulation was assessed by western blotting and immunofluorescence. The Nrf2 knockout vector and ml385 were used to block the Nrf2 pathway, and the differences between the different groups in the expression of pyroptosis protein, cell activity, and incidence of apoptosis were evaluated to verify the relationship between the effect of curcumin on pyroptosis inhibition and the Nrf2 pathway. Curcumin promoted the transfer of Nrf2 into the nucleus through the AKT pathway and increased the expression of the antioxidant factors HO-1 and GCLC. These effects reduced reactive oxygen species accumulation and mitochondrial damage in diabetic myocardium and inhibited diabetes-induced pyroptosis. However, in cardiomyocytes with a blocked Nrf2 pathway, the ability of curcumin to inhibit pyroptosis was significantly reduced, and the protective effect on the cells was lost. Curcumin can reduce the accumulation of superoxide in the myocardium through AKT/Nrf2/ARE pathway activation and inhibit pyroptosis. It also has a role in diabetic cardiomyopathy treatment. This study provides new directions for evaluating the mechanism of diabetic cardiomyopathy and treating diabetic myocardium.
Collapse
Affiliation(s)
- Zhang Wei
- Department of Cardiovascular Medicine of The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui, China 233000
| | - Kang pinfang
- Department of Cardiovascular Medicine of The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui, China 233000
| | - Zhou jing
- Department of Cardiovascular Medicine of The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui, China 233000
| | - Yao zhuoya
- Department of Cardiovascular Medicine of The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui, China 233000
| | - Qian Shaohuan
- Department of Cardiovascular Medicine of The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui, China 233000
| | - Shi Chao
- Department of Cardiac Surgery of The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui, China 233000
| |
Collapse
|
25
|
Li JY, Zhao CC, Peng JF, Zhang M, Wang L, Yin G, Zhou P. The Protective Effect of Sheng Mai Yin on Diabetic Cardiomyopathy via NLRP3/Caspase-1 Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:1234434. [PMID: 36506810 PMCID: PMC9731757 DOI: 10.1155/2022/1234434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 10/30/2022] [Accepted: 11/15/2022] [Indexed: 12/02/2022]
Abstract
Sheng Mai Yin (SMY) has therapeutic effects on myocardial infarction (MI), heart failure (HF), diabetic cardiomyopathy (DCM), and myocarditis. To study whether SMY can relieve pyroptosis and play a protective role in diabetic cardiomyopathy, a molecular docking technique was used to predict the possible mechanism of SMY against DCM. Then, a DCM rat model was induced by intraperitoneal injection of streptozotocin (STZ), divided into 5 groups: the DM group (model), SMY-L group (2.7 mL/kg SMY), SMY-M group (5.4 mL/kg SMY), SMY-H group (10.8 mL/kg SMY), and Met group (120 mg/kg metformin). Rats in the CTL group (control) and DM group were given normal saline. After 8 weeks, the levels of blood glucose, lipids, and myocardial enzymes were detected according to the kit instructions. Cardiac function was detected by echocardiography. HE and Masson were used to observing the pathological changes, collagen deposition, and collagen volume fraction (CVF). The apoptosis rate of cardiomyocytes was determined by Tunel. The IL-1β level was determined by ELISA and RT-PCR. The expressions of NLRP3, caspase-1, and GSDMD were measured using RT-PCR and Western blotting. The docking results suggested that SMY may act on NLRP3 and its downstream signal pathway. The in vivo results showed that SMY could reduce blood glucose and lipid levels, improve heart function, improve histopathological changes and myocardial enzymes, and alleviate cardiomyocyte apoptosis and myocardial fibrosis. SMY inhibited the mRNA and protein expressions of NLRP3, ASC, Caspase-1, and GSDMD and IL-1β production. SMY can reduce DCM by regulating the NLRP3/caspase-1 signaling pathway, providing a new research direction for the treatment of DCM.
Collapse
Affiliation(s)
- Jing-Ya Li
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Chun-Chun Zhao
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Jian-Fei Peng
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Meng Zhang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Liang Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Gang Yin
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
- The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Peng Zhou
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| |
Collapse
|
26
|
Mesenchymal Stem Cell Transplantation Increases Antioxidant Protein Expression and Ameliorates GP91/ROS/Inflammasome Signals in Diabetic Cardiomyopathy. J Cardiovasc Dev Dis 2022; 9:jcdd9110381. [DOI: 10.3390/jcdd9110381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/29/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
Background: Cardiomyopathy is one of the complications associated with diabetes. Due to its high prevalence, diabetic cardiomyopathy has become an urgent issue for diabetic patients. Various pathological signals are related to diabetic cardiomyopathy progress, including inflammasome. Mesenchymal stem cell transplantation is full of potential for the treatment of diabetic cardiomyopathy because of stem cell cardiac regenerative capability. This study investigates whether mesenchymal stem cell transplantation shows therapeutic effects on diabetic cardiomyopathy through inflammasome signaling regulation. Methods: Wistar male rats were divided into three groups including Sham, T1DM (rats with type 1 diabetes) and T1DM + WJSC (T1DM rats receiving 1 × 106 stem cells per rat). Results: Compared to the Sham, experimental results indicated that several pathological conditions can be observed in heart tissues with T1DM, including structural change, fibrosis, oxidative stress elevation and inflammasome related protein expression. All of these pathological conditions were significantly improved in T1DM rats receiving mesenchymal stem cell transplantation (T1DM + WJSC). Furthermore, the experimental findings suggest that mesenchymal stem cell transplantation exerted antioxidant protein expression in diabetic heart tissues, resulting in a decrease in oxidative stress and inflammasome signaling blockage. Conclusion: These findings imply that mesenchymal stem cell transplantation shows therapeutic effects on diabetic cardiomyopathy through inflammasome regulation induced by oxidative stress.
Collapse
|
27
|
Xi Y, Chen D, Dong Z, Zhang J, Lam H, He J, Du K, Chen C, Guo J, Xiao J. Multi-omics insights into potential mechanism of SGLT2 inhibitors cardiovascular benefit in diabetic cardiomyopathy. Front Cardiovasc Med 2022; 9:999254. [PMID: 36277768 PMCID: PMC9579694 DOI: 10.3389/fcvm.2022.999254] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Background Metabolic and energy disorders are considered central to the etiology of diabetic cardiomyopathy (DCM). Sodium-glucose cotransporter-2 inhibitors (SGLT2i) can effectively reduce the risk of cardiovascular death and heart failure in patients with DCM. However, the underlying mechanism has not been elucidated. Methods We established a DCM rat model followed by treatment with empagliflozin (EMPA) for 12 weeks. Echocardiography, blood tests, histopathology, and transmission electron microscopy (TEM) were used to evaluate the phenotypic characteristics of the rats. The proteomics and metabolomics of the myocardium in the rat model were performed to identify the potential targets and signaling pathways associated with the cardiovascular benefit of SGLT2i. Results The diabetic rat showed pronounced DCM characterized by mitochondrial pleomorphic, impaired lipid metabolism, myocardial fibrosis, and associated diastolic and systolic functional impairments in the heart. To some extent, these changes were ameliorated after treatment with EMPA. A total of 43 proteins and 34 metabolites were identified as targets in the myocardium of diabetic rats treated with EMPA. The KEGG analysis showed that arachidonic acid is associated with the maximum number of related pathways and may be a potential target of EMPA treatment. Fatty acid (FA) metabolism was enhanced in diabetic hearts, and the perturbation of biosynthesis of unsaturated FAs and arachidonic acid metabolism was a potential enabler for the cardiovascular benefit of EMPA. Conclusion SGLT2i ameliorated lipid accumulation and mitochondrial damage in the myocardium of diabetic rats. The metabolomic and proteomic data revealed the potential targets and signaling pathways associated with the cardiovascular benefit of SGLT2i, which provides a valuable resource for the mechanism of SGLT2i.
Collapse
Affiliation(s)
- Yangbo Xi
- The First Clinical Medical College, Jinan University, Guangzhou, China,Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Dongping Chen
- Central Laboratory, Binhaiwan Central Hospital of Dongguan, The Dongguan Affiliated Hospital of Jinan University, Dongguan, China
| | - Zhihui Dong
- Central Laboratory, Binhaiwan Central Hospital of Dongguan, The Dongguan Affiliated Hospital of Jinan University, Dongguan, China
| | - Jinhua Zhang
- The First Clinical Medical College, Jinan University, Guangzhou, China
| | - Hingcheung Lam
- The First Clinical Medical College, Jinan University, Guangzhou, China
| | - Jiading He
- The First Clinical Medical College, Jinan University, Guangzhou, China
| | - Keyi Du
- The First Clinical Medical College, Jinan University, Guangzhou, China
| | - Can Chen
- Department of Pathology, Binhaiwan Central Hospital of Dongguan, The Dongguan Affiliated Hospital of Jinan University, Dongguan, China
| | - Jun Guo
- The First Clinical Medical College, Jinan University, Guangzhou, China,Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, China,*Correspondence: Jun Guo,
| | - Jianmin Xiao
- The First Clinical Medical College, Jinan University, Guangzhou, China,Central Laboratory, Binhaiwan Central Hospital of Dongguan, The Dongguan Affiliated Hospital of Jinan University, Dongguan, China,Department of Cardiology, Binhaiwan Central Hospital of Dongguan, The Dongguan Affiliated Hospital of Jinan University, Dongguan, China,Jianmin Xiao,
| |
Collapse
|
28
|
Huang Y, Li X, Luo G, Wang J, Li R, Zhou C, Wan T, Yang F. Pyroptosis as a candidate therapeutic target for Alzheimer’s disease. Front Aging Neurosci 2022; 14:996646. [PMID: 36185484 PMCID: PMC9520296 DOI: 10.3389/fnagi.2022.996646] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/12/2022] [Indexed: 11/22/2022] Open
Abstract
Pyroptosis is a form of cell death mediated by inflammasomes and gasdermins, and the relevance of pyroptosis to neurodegenerative diseases is currently receiving increasing attention. Alzheimer’s disease (AD) is a chronic progressive neurodegenerative disease that is closely associated with neuroinflammation. Its main pathological features include β-amyloid (Aβ) deposition, Tau protein hyperphosphorylation and neuronal loss. Aβ, tau-induced microglia pyroptosis and polarization leading to neuroinflammation play an important role in the pathogenesis of AD. Studying the pathogenesis and treatment of AD based on cellular pyroptosis has become a new direction in AD research. In this paper, we review the research progress of pyroptosis and will focus on the pathogenic roles of pyroptosis in AD and the role of targeted inhibition of inflammasome-dependent pyroptosis in AD treatment. These results deepen our understanding of the pathogenesis of AD and provide ideas for the development of new drugs based on the regulation of pyroptosis in AD patients.
Collapse
Affiliation(s)
- Yuehua Huang
- Department of Reproductive Medicine, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
- Reproductive Medicine, Guangxi Medical and Health Key Discipline Construction Project of the Affiliated Hospital of Youjiang Medical University for Nationalities, Baise ,Guangxi, China
| | - Xiaoyu Li
- Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Guifei Luo
- Department of Reproductive Medicine, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
- Reproductive Medicine, Guangxi Medical and Health Key Discipline Construction Project of the Affiliated Hospital of Youjiang Medical University for Nationalities, Baise ,Guangxi, China
| | - Junli Wang
- Reproductive Medicine, Guangxi Medical and Health Key Discipline Construction Project of the Affiliated Hospital of Youjiang Medical University for Nationalities, Baise ,Guangxi, China
- Industrial College of Biomedicine and Health Industry, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Ranhui Li
- Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Chuyi Zhou
- Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Teng Wan
- Hengyang Medical College, University of South China, Hengyang, Hunan, China
- Teng Wan,
| | - Fenglian Yang
- Industrial College of Biomedicine and Health Industry, Youjiang Medical University for Nationalities, Baise, Guangxi, China
- *Correspondence: Fenglian Yang,
| |
Collapse
|
29
|
The Role of NLRP3 Inflammasome in Diabetic Cardiomyopathy and Its Therapeutic Implications. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3790721. [PMID: 36111168 PMCID: PMC9470324 DOI: 10.1155/2022/3790721] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022]
Abstract
Diabetic cardiomyopathy (DCM) is a serious complication of diabetes mellitus (DM). However, the precise molecular mechanisms remain largely unclear, and it is still a challenging disease to diagnose and treat. The nucleotide-binding oligomerization domain and leucine-rich repeat pyrin 3 domain (NLRP3) inflammasome is a critical part of the innate immune system in the host to defend against endogenous danger and pathogenic microbial infections. Dysregulated NLRP3 inflammasome activation results in the overproduction of cytokines, primarily IL-1β and IL-18, and eventually, inflammatory cell death-pyroptosis. A series of studies have indicated that NLRP3 inflammasome activation participates in the development of DCM, and that corresponding interventions could mitigate disease progression. Accordingly, this narrative review is aimed at briefly summarizing the cell-specific role of the NLRP3 inflammasome in DCM and provides novel insights into developing DCM therapeutic strategies targeting the NLRP3 inflammasome.
Collapse
|
30
|
Habimana O, Modupe Salami O, Peng J, Yi GH. Therapeutic Implications of Targeting Pyroptosis in Cardiac-related Etiology of Heart Failure. Biochem Pharmacol 2022; 204:115235. [PMID: 36044938 DOI: 10.1016/j.bcp.2022.115235] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/26/2022]
Abstract
Heart failure remains a considerable clinical and public health problem, it is the dominant cause of death from cardiovascular diseases, besides, cardiovascular diseases are one of the leading causes of death worldwide. The survival of patients with heart failure continues to be low with 45-60% reported deaths within five years. Apoptosis, necrosis, autophagy, and pyroptosis mediate cardiac cell death. Acute cell death is the hallmark pathogenesis of heart failure and other cardiac pathologies. Inhibition of pyroptosis, autophagy, apoptosis, or necrosis reduces cardiac damage and improves cardiac function in cardiovascular diseases. Pyroptosis is a form of inflammatory deliberate cell death that is characterized by the activation of inflammasomes such as NOD-like receptors (NLR), absent in melanoma 2 (AIM2), interferon-inducible protein 16 (IFI-16), and their downstream effector cytokines: Interleukin IL-1β and IL-18 leading to cell death. Recent studies have shown that pyroptosis is also the dominant cell death process in cardiomyocytes, cardiac fibroblasts, endothelial cells, and immune cells. It plays a crucial role in the pathogenesis of cardiac diseases that contribute to heart failure. This review intends to summarize the therapeutic implications targeting pyroptosis in the main cardiac pathologies preceding heart failure.
Collapse
Affiliation(s)
- Olive Habimana
- International College, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China
| | | | - Jinfu Peng
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China; Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China
| | - Guang-Hui Yi
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China; Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China.
| |
Collapse
|
31
|
The Chemerin/CMKLR1 Axis Is Involved in the Recruitment of Microglia to Aβ Deposition through p38 MAPK Pathway. Int J Mol Sci 2022; 23:ijms23169041. [PMID: 36012305 PMCID: PMC9409288 DOI: 10.3390/ijms23169041] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/03/2022] [Accepted: 08/09/2022] [Indexed: 01/12/2023] Open
Abstract
The accumulation of microglia around senile plaques is one of the pathological features of Alzheimer's disease (AD). Chemerin is an adipokine with immune-modulating properties. Our previous study showed that chemokine-like receptor 1 (CMKLR1), the receptor for chemerin, is also a functional receptor of Aβ. However, it remains unclear whether and how the chemerin/CMKLR1 axis affects the migration of microglia. The impact of CMKLR1 on microglial activation and recruitment toward Aβ deposits was examined in APP/PS1 mice mated with CMKLR1 knockout (CMKLR1-/-) mice. CMKLR1 deficiency reduced the number of microglia around Aβ deposits in aged APP/PS1-CMKLR1-/- mice compared with APP/PS1 mice. Chemerin expression was significantly decreased in the hippocampus and cortex of aged APP/PS1 mice compared with WT mice. In vitro assays demonstrated that activation of the chemerin/CMKLR1 axis promoted the migration of primary cultures of microglia and murine microglial N9 cells. Mechanistic studies found that chemerin/CMKLR1 induced polarization and protrusion formation of microglia by promoting the remodeling of actin filaments and microtubules, and Golgi apparatus reorientation. The inhibition of p38 MAPK attenuated the promotion of the chemerin/CMKLR1 axis on microglial migration and polarization. In addition, chemerin inhibited Aβ-induced microglial clustering. The inhibition of p38 MAPK alleviated the suppressive effect of chemerin on Aβ-induced microglial aggregation. Our data indicate that the chemerin/CMKLR1 axis is involved in the migration and recruitment of microglia to senile plaques via the p38 MAPK pathway. Modulation of the chemerin/CMKLR1 axis is a potential new strategy for AD therapy.
Collapse
|
32
|
Geng X, Li Z, Yang Y. Emerging Role of Epitranscriptomics in Diabetes Mellitus and Its Complications. Front Endocrinol (Lausanne) 2022; 13:907060. [PMID: 35692393 PMCID: PMC9184717 DOI: 10.3389/fendo.2022.907060] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/14/2022] [Indexed: 01/13/2023] Open
Abstract
Diabetes mellitus (DM) and its related complications are among the leading causes of disability and mortality worldwide. Substantial studies have explored epigenetic regulation that is involved in the modifications of DNA and proteins, but RNA modifications in diabetes are still poorly investigated. In recent years, posttranscriptional epigenetic modification of RNA (the so-called 'epitranscriptome') has emerged as an interesting field of research. Numerous modifications, mainly N6 -methyladenosine (m6A), have been identified in nearly all types of RNAs and have been demonstrated to have an indispensable effect in a variety of human diseases, such as cancer, obesity, and diabetes. Therefore, it is particularly important to understand the molecular basis of RNA modifications, which might provide a new perspective for the pathogenesis of diabetes mellitus and the discovery of new therapeutic targets. In this review, we aim to summarize the recent progress in the epitranscriptomics involved in diabetes and diabetes-related complications. We hope to provide some insights for enriching the understanding of the epitranscriptomic regulatory mechanisms of this disease as well as the development of novel therapeutic targets for future clinical benefit.
Collapse
Affiliation(s)
- Xinqian Geng
- Department of Endocrinology, The Affiliated Hospital of Yunnan University and the Second People’s Hospital of Yunnan Province, Kunming, China
| | - Zheng Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ying Yang
- Department of Endocrinology, The Affiliated Hospital of Yunnan University and the Second People’s Hospital of Yunnan Province, Kunming, China
| |
Collapse
|
33
|
Zhang Q, Xiao Z, Lee CL, Duan YG, Fan X, Yeung WSB, Chiu PCN, Zhang JV. The Regulatory Roles of Chemerin-Chemokine-Like Receptor 1 Axis in Placental Development and Vascular Remodeling During Early Pregnancy. Front Cell Dev Biol 2022; 10:883636. [PMID: 35656551 PMCID: PMC9152263 DOI: 10.3389/fcell.2022.883636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Chemerin is an adipokine that regulates metabolism in pregnancy. An elevation of serum chemerin level is associated with pregnancy complications. Consistently, we demonstrated that the chemerin expression was increased in placenta of preeclamptic patients at deliveries. The G protein-coupled receptor chemokine-like receptor 1 (CMKLR1) mediates the actions of chemerin. The functions of the chemerin-CMKLR1 axis in maintaining pregnancy are still unknown. In this study, we demonstrated that CMKLR1 was expressed in the decidual natural killer (dNK) cells and chorionic villi of human. Chemerin suppressed the proliferation of the dNK cells in vitro. Specific antagonist of CMKLR1, α-Neta abolished the suppressive effect of spent medium from chemerin-treated dNK cells culture on extravillous trophoblast invasion. Activation of the chemerin-CMKLR1 axis promoted fusion and differentiation of human cytotrophoblast to syncytiotrophoblast in vitro. We generated Cmklr1 knockout mice and showed that the Cmklr1 deficiency negatively affected pregnancy outcome in terms of number of implantation sites, litter size and fetal weight at birth. Histologically, the Cmklr1 deficiency impaired formation of the syncytiotrophoblast layer II, induced enlargement of the maternal lacunae in the labyrinth, increased the diameter of the spiral arteries and increased trophoblast invasion in the decidua. The Cmklr1 deficient placenta also displayed an increased number of dNK cells and serum IL-15 level. In summary, the chemerin-CMKLR1 axis regulated placental development and spiral artery remodeling in early pregnancy.
Collapse
Affiliation(s)
- Qingqing Zhang
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Obstetrics and Gynecology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Zhonglin Xiao
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Cheuk-Lun Lee
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Obstetrics and Gynecology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yong-Gang Duan
- Department of Obstetrics and Gynecology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xiujun Fan
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - William S. B. Yeung
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Obstetrics and Gynecology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Philip C. N. Chiu
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Obstetrics and Gynecology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- *Correspondence: Philip C. N. Chiu, ; Jian V. Zhang,
| | - Jian V. Zhang
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Key Laboratory of Metabolic Health, Shenzhen, China
- *Correspondence: Philip C. N. Chiu, ; Jian V. Zhang,
| |
Collapse
|
34
|
Xi Y, Chen D, Dong Z, Lam H, He J, Du K, Chen C, Guo J, Xiao J. RNA Sequencing of Cardiac in a Rat Model Uncovers Potential Target LncRNA of Diabetic Cardiomyopathy. Front Genet 2022; 13:848364. [PMID: 35495145 PMCID: PMC9044075 DOI: 10.3389/fgene.2022.848364] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/18/2022] [Indexed: 12/16/2022] Open
Abstract
Background: Diabetic cardiomyopathy (DCM) is one of the major causes of heart failure in diabetic patients; however, its pathogenesis remains unclear. Long non-coding RNAs (lncRNAs) are involved in the development of various cardiovascular diseases, but little is known in DCM. Objective: The present study was conducted to investigate the altered expression signature of lncRNAs and mRNAs by RNA-sequencing and uncovers the potential targets of DCM. Methods: A DCM rat model was established, and the genome-wide expression profile of cardiac lncRNAs and mRNAs was investigated in the rat model with and without DCM by RNA-sequencing. Bioinformatics analysis included the co-expression, competitive endogenous RNA (ceRNA) network, and functional enrichment analysis of deregulated lncRNAs and mRNAs. Results: A total of 355 lncRNA transcripts and 828 mRNA transcripts were aberrantly expressed. The ceRNA network showed that lncRNA XR_351927.3, ENSRNOT00000089581, XR_597359.2, XR_591602.2, and XR_001842089.1 are associated with the greatest number of differentially expressed mRNAs and AURKB, MELK, and CDK1 may be the potential regulatory targets of these lncRNAs. Functional analysis showed that these five lncRNAs are closely associated with fibration, cell proliferation, and energy metabolism of cardiac myocytes, indicating that these core lncRNAs have high significance in DCM. Conclusions: The present study profiled the DCM-specific lncRNAs and mRNAs, constructed the lncRNA-related ceRNA regulatory network, and identified the potential prognostic biomarkers, which provided new insights into the pathogenesis of DCM.
Collapse
Affiliation(s)
- Yangbo Xi
- Department of The First Clinical Medical College, Jinan University, Guangzhou, China
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Dongping Chen
- Central Laboratory, The Dongguan Affiliated Hospital of Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Zhihui Dong
- Central Laboratory, The Dongguan Affiliated Hospital of Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Hingcheung Lam
- Department of The First Clinical Medical College, Jinan University, Guangzhou, China
| | - Jiading He
- Department of The First Clinical Medical College, Jinan University, Guangzhou, China
| | - Keyi Du
- Department of The First Clinical Medical College, Jinan University, Guangzhou, China
| | - Can Chen
- Department of Pathology, The Dongguan Affiliated Hospital of Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Jun Guo
- Department of The First Clinical Medical College, Jinan University, Guangzhou, China
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- *Correspondence: Jun Guo, ; Jianmin Xiao,
| | - Jianmin Xiao
- Department of The First Clinical Medical College, Jinan University, Guangzhou, China
- Central Laboratory, The Dongguan Affiliated Hospital of Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan, China
- Department of Cardiology, The Dongguan Affiliated Hospital of Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan, China
- *Correspondence: Jun Guo, ; Jianmin Xiao,
| |
Collapse
|
35
|
Agbaedeng TA, Zacharia AL, Iroga PE, Rathnasekara VM, Munawar DA, Bursill C, Noubiap JJ. Associations between adipokines and atrial fibrillation: A systematic review and meta-analysis. Nutr Metab Cardiovasc Dis 2022; 32:853-862. [PMID: 35227548 DOI: 10.1016/j.numecd.2022.01.019] [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: 09/24/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 11/24/2022]
Abstract
AIMS Although overweight and obesity are associated with increased risk of atrial fibrillation (AF), the underlying mechanisms are not well characterised. Recent data suggest that this link may be partly due to abnormal adipose tissue-derived cytokines or adipokines. However, this relationship is not well clarified. To evaluate the association between adipokines and AF in a systematic review and meta-analysis. DATA SYNTHESIS PubMed, Embase, and Web of Science Core Collection were searched from inception through 1st March 2021. Studies were included if they reported any adipokine and AF, with their quality assessed using the Newcastle-Ottawa scale. Data were independently abstracted, with unadjusted and multivariable adjusted estimates pooled in a random-effects meta-analysis. Data are presented for overall prevalent or incident AF and AF subtypes (paroxysmal, persistent, or non-paroxysmal AF). A total of 34 studies, with 31,479 patients, were included. The following adipokines were significantly associated with AF in the pooled univariate data - apelin (risk ratio for prevalent AF: 0.05 [0.00-0.50], p = 0.01; recurrent AF: 0.21 [0.11-0.42], p < 0.01) and resistin (incident AF: 2.05 [1.02-4.1], p = 0.04; prevalent AF: 2.62 [1.78-3.85], p < 0.01). Pooled analysis of multivariable adjusted effect size estimates showed adiponectin as the sole independent predictor of AF incidence (1.14 [1.02-1.27], p = 0.02). Moreover, adiponectin was associated with non-paroxysmal AF (persistent AF: 1.45 [1.08-1.94, p = 0.01; non-paroxysmal versus paroxysmal AF: 3.14 [1.87-5.27, p < 0.01). CONCLUSIONS Adipokines, principally adiponectin, apelin, and resistin, are associated with the risk of atrial fibrillation. However, the association is not seen after multivariate adjustment, likely reflecting the lack of statistical power. Future research should investigate these relationships in larger prospective cohorts and how they can refine AF monitoring strategies. PROSPERO ID CRD42020208879.
Collapse
Affiliation(s)
- Thomas A Agbaedeng
- Adelaide Medical School, The University of Adelaide, Adelaide, Australia.
| | | | - Peter E Iroga
- Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| | | | - Dian A Munawar
- Lyell McEwin Hospital, The University of Adelaide, Adelaide, Australia; Department of Cardiology and Vascular Medicine, University of Indonesia, Jakarta, Indonesia
| | - Christina Bursill
- Adelaide Medical School, The University of Adelaide, Adelaide, Australia; Vascular Research Centre, Lifelong Health Theme, SAHMRI, Adelaide, Australia
| | - Jean Jacques Noubiap
- Adelaide Medical School, The University of Adelaide, Adelaide, Australia; Centre for Heart Rhythm Disorders, The University of Adelaide, Adelaide, Australia
| |
Collapse
|
36
|
Peng L, Chen Y, Li Y, Feng P, Zheng Y, Dong Y, Yang Y, Wang R, Li A, Yan J, Shang F, Tang P, Chen D, Gao Y, Huang W. Chemerin Regulates the Proliferation and Migration of Pulmonary Arterial Smooth Muscle Cells via the ERK1/2 Signaling Pathway. Front Pharmacol 2022; 13:767705. [PMID: 35370637 PMCID: PMC8971604 DOI: 10.3389/fphar.2022.767705] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is an incurable disease with high mortality. Chemerin has been found to be associated with pulmonary hypertension (PH). However, the specific role of chemerin in mediating PH development remains unclear. This study aimed to elucidate the regulatory effects and the underlying mechanism of chemerin on PH and to investigate the expression levels of chemerin protein in plasma in PAH patients. In vivo, two animal models of PH were established in rats by monocrotaline (MCT) injection and hypoxia. We found that the expression levels of chemerin and its receptor, chemokine-like receptor 1 (CMKLR1), were significantly upregulated in the lungs of PH rats. Primary cultured pulmonary arterial smooth muscle cells [(PASMCs) (isolated from pulmonary arteries of normal healthy rats)] were exposed to hypoxia or treated with recombinant human chemerin, we found that CMKLR1 expression was upregulated in PASMCs in response to hypoxia or chemerin stimulation, whereas the exogenous chemerin significantly promoted the migration and proliferation of PASMCs. Notably, the regulatory effects of chemerin on PASMCs were blunted by PD98059 (a selective ERK1/2 inhibitor). Using enzyme linked immunosorbent assay (ELISA), we found that the protein level of chemerin was also markedly increased in plasma from idiopathic pulmonary arterial hypertension (IPAH) patients compared to that from healthy controls. Moreover, the diagnostic value of chemerin expression in IPAH patients was determined through receiver operating characteristic (ROC) curve analysis and the result revealed that area under ROC curve (AUC) for plasma chemerin was 0.949. Taken together, these results suggest that chemerin exacerbates PH progression by promoting the proliferation and migration of PASMCs via the ERK1/2 signaling pathway, and chemerin is associated with pulmonary hypertension.
Collapse
Affiliation(s)
- Linqian Peng
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Yunwei Chen
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Yan Li
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Panpan Feng
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Zheng
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Yongjie Dong
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Yunjing Yang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ruiyu Wang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Ailing Li
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianghong Yan
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Feifei Shang
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Ping Tang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dewei Chen
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
| | - Yuqi Gao
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
| | - Wei Huang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
37
|
Wei Y, Yang L, Pandeya A, Cui J, Zhang Y, Li Z. Pyroptosis-Induced Inflammation and Tissue Damage. J Mol Biol 2022; 434:167301. [PMID: 34653436 PMCID: PMC8844146 DOI: 10.1016/j.jmb.2021.167301] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/23/2021] [Accepted: 10/05/2021] [Indexed: 02/07/2023]
Abstract
Programmed cell deaths are pathways involving cells playing an active role in their own destruction. Depending on the signaling system of the process, programmed cell death can be divided into two categories, pro-inflammatory and non-inflammatory. Pyroptosis is a pro-inflammatory form of programmed cell death. Upon cell death, a plethora of cytokines are released and trigger a cascade of responses from the neighboring cells. The pyroptosis process is a double-edged sword, could be both beneficial and detrimental in various inflammatory disorders and disease conditions. A physiological outcome of these responses is tissue damage, and sometimes death of the host. In this review, we focus on the inflammatory response triggered by pyroptosis, and resulting tissue damage in selected organs.
Collapse
Affiliation(s)
- Yinan Wei
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY, USA.
| | - Ling Yang
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY, USA
| | - Ankit Pandeya
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY, USA
| | - Jian Cui
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY, USA
| | - Yan Zhang
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY, USA.,Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou,China
| | - Zhenyu Li
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
38
|
Kian M, Hosseini E, Abdizadeh T, Langaee T, Khajouei A, Ghasemi S. Molecular docking and mouse modeling suggest CMKLR1 and INSR as targets for improving PCOS phenotypes by minocycline. EXCLI JOURNAL 2022; 21:400-414. [PMID: 35368462 PMCID: PMC8971357 DOI: 10.17179/excli2021-4534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 02/09/2022] [Indexed: 11/10/2022]
Abstract
Polycystic ovary syndrome (PCOS) is the most common cause of women's infertility. Some inflammatory pathways play a pivotal role in the pathogenesis of PCOS. This study aimed to investigate the possible beneficial effects of minocycline on chemokine-like receptor 1 (CMKLR1) and Insulin Receptor (INSR) in a PCOS model. A molecular docking study was implemented using Molecular Operating Environment (MOE) software. The PCOS was induced in NMRI mice (mean body weight 14.47±0.23) by 28 days estradiol valerate injection (2 mg/kg/day). The mice were then divided into six groups (n=8 per group, mean body weight 17.77± 0.26): control (received normal saline), PCOS model, control for minocycline, minocycline treated PCOS (50 mg/kg), letrozole treated PCOS (0.5 mg/kg), and metformin-treated PCOS (300 mg/kg). Serum FSH, LH, estradiol (E2), and testosterone were detected by ELISA. The ovarian tissues were stained by hematoxylin and eosin. The CMKLR1 and INSR expression levels were determined by Real-time-PCR. The molecular docking studies showed scores of -10.92 and -9.30 kcal/mol, respectively, for minocycline with CMKLR1 and INSR. Estradiol valerate treatment led to a significant increase in E2, graffian follicle, and decrease in corpus luteum (CL) numbers (P<0.05), while minocycline treatment improved these PCOS features. The minocycline treatment significantly decreased the CMKLR1 expression and increased the INSR expression (P<0.05) while the CMKLR1 expression was increased in PCOS model. Minocycline may improve ovulation in PCOS model by returning E2 to a normal level and increasing CL number (ovulation signs). These beneficial outcomes may be related to the changes in CMKLR1 and INSR gene expression involved in glucose metabolism and inflammation.
Collapse
Affiliation(s)
- Mahdie Kian
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Elham Hosseini
- Department of Obstetrics and Gynecology, IVF Clinic, Mousavi Hospital, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Tooba Abdizadeh
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Taimour Langaee
- Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Azadeh Khajouei
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sorayya Ghasemi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran,*To whom correspondence should be addressed: Sorayya Ghasemi, Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran; Tel: 09131856090, 03833331471, E-mail: ,
| |
Collapse
|
39
|
Lu Y, Lu Y, Meng J, Wang Z. Pyroptosis and Its Regulation in Diabetic Cardiomyopathy. Front Physiol 2022; 12:791848. [PMID: 35145423 PMCID: PMC8822267 DOI: 10.3389/fphys.2021.791848] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/10/2021] [Indexed: 12/17/2022] Open
Abstract
Diabetic cardiomyopathy (DbCM) is a prevalent disease, characterized by contractile dysfunction and left ventricular hypertrophy. Patients with DbCM have high morbidity and mortality worldwide. Recent studies have identified that pyroptosis, a kind of cell death, could be induced by hyperglycemia involved in the formation of DbCM. This review summarizes the regulatory mechanisms of pyroptosis in DbCM, including NOD-like receptor3, AIM2 inflammasome, long non-coding RNAs, microRNAs, circular RNA, autophagy, and some drugs.
Collapse
Affiliation(s)
- Yafang Lu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Yaqiong Lu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Jun Meng
- Functional Department, The First Affiliated Hospital, University of South China, Hengyang, China
- *Correspondence: Jun Meng,
| | - Zuo Wang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
- Zuo Wang,
| |
Collapse
|
40
|
Muñoz-Córdova F, Hernández-Fuentes C, Lopez-Crisosto C, Troncoso MF, Calle X, Guerrero-Moncayo A, Gabrielli L, Chiong M, Castro PF, Lavandero S. Novel Insights Into the Pathogenesis of Diabetic Cardiomyopathy and Pharmacological Strategies. Front Cardiovasc Med 2022; 8:707336. [PMID: 35004869 PMCID: PMC8734937 DOI: 10.3389/fcvm.2021.707336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 11/29/2021] [Indexed: 12/17/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is a severe complication of diabetes developed mainly in poorly controlled patients. In DCM, several clinical manifestations as well as cellular and molecular mechanisms contribute to its phenotype. The production of reactive oxygen species (ROS), chronic low-grade inflammation, mitochondrial dysfunction, autophagic flux inhibition, altered metabolism, dysfunctional insulin signaling, cardiomyocyte hypertrophy, cardiac fibrosis, and increased myocardial cell death are described as the cardinal features involved in the genesis and development of DCM. However, many of these features can be associated with broader cellular processes such as inflammatory signaling, mitochondrial alterations, and autophagic flux inhibition. In this review, these mechanisms are critically discussed, highlighting the latest evidence and their contribution to the pathogenesis of DCM and their potential as pharmacological targets.
Collapse
Affiliation(s)
- Felipe Muñoz-Córdova
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santiago, Chile
| | - Carolina Hernández-Fuentes
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santiago, Chile
| | - Camila Lopez-Crisosto
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santiago, Chile.,Division of Cardiovascular Diseases, Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), Pontifical Catholic University of Chile, Santiago, Chile
| | - Mayarling F Troncoso
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santiago, Chile.,Department of Medical Technology, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Ximena Calle
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santiago, Chile
| | - Alejandra Guerrero-Moncayo
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santiago, Chile
| | - Luigi Gabrielli
- Division of Cardiovascular Diseases, Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), Pontifical Catholic University of Chile, Santiago, Chile
| | - Mario Chiong
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santiago, Chile
| | - Pablo F Castro
- Division of Cardiovascular Diseases, Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), Pontifical Catholic University of Chile, Santiago, Chile.,Corporación Centro de Estudios Científicos de las Enfermedades Crónicas (CECEC), University of Chile, Santiago, Chile
| | - Sergio Lavandero
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santiago, Chile.,Corporación Centro de Estudios Científicos de las Enfermedades Crónicas (CECEC), University of Chile, Santiago, Chile.,Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
41
|
Wan L, Bai X, Zhou Q, Chen C, Wang H, Liu T, Xue J, Wei C, Xie L. The advanced glycation end-products (AGEs)/ROS/NLRP3 inflammasome axis contributes to delayed diabetic corneal wound healing and nerve regeneration. Int J Biol Sci 2022; 18:809-825. [PMID: 35002527 PMCID: PMC8741862 DOI: 10.7150/ijbs.63219] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 11/27/2021] [Indexed: 12/31/2022] Open
Abstract
Diabetic keratopathy (DK) is an important diabetic complication at the ocular surface. Chronic low-grade inflammation mediated by the NLRP3 inflammasome promotes pathogenesis of diabetes and its complications. However, the effect of the NLRP3 inflammasome on DK pathogenesis remains elusive. Wild-type (WT) and Nlrp3 knockout (KO) C57 mice were used to establish a type I diabetes model by intraperitoneal injection of streptozotocin. The effect of the NLRP3 inflammasome on diabetic corneal wound healing and never regeneration was examined by a corneal epithelial abrasion model. Western blot, immunofluorescence staining, enzyme-linked immunosorbent assay (ELISA) and pharmacological treatment were performed to investigate the regulatory mechanism of advanced glycation end products (AGEs) on NLRP3 inflammasome activation and corneal wound healing in vivo. The cultured mouse corneal epithelial cells (TKE2) were used to evaluate the effect and mechanism of AGEs on NLRP3 inflammasome activation in vitro. We revealed that NLRP3 inflammasome-mediated inflammation and pyroptosis contributed to DK pathogenesis. Under physiological conditions, the NLRP3 inflammasome was required for corneal wound healing and nerve regeneration. However, under a diabetic scenario, sustained activation of the NLRP3 inflammasome resulted in postponed corneal wound healing and impaired nerve regeneration. Mechanistically, the accumulated AGEs promoted hyperactivation of the NLRP3 inflammasome through ROS production. Moreover, genetically and pharmacologically blocking the AGEs/ROS/NLRP3 inflammasome axis significantly expedited diabetic corneal epithelial wound closure and nerve regeneration. Our results revealed that AGEs-induced hyperactivation of the NLRP3 inflammasome resulted in delayed diabetic corneal wound healing and impaired nerve regeneration, which further highlighted the NLRP3 inflammasome as a promising target for DK treatment.
Collapse
Affiliation(s)
- Luqin Wan
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266071, China
| | - Xiaofei Bai
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Qingjun Zhou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Chen Chen
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Huifeng Wang
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266071, China
| | - Ting Liu
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Junfa Xue
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Chao Wei
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Lixin Xie
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| |
Collapse
|
42
|
Rao Y, Gai X, Xiong J, Le Y, Sun Y. Transient Receptor Potential Cation Channel Subfamily V Member 4 Mediates Pyroptosis in Chronic Obstructive Pulmonary Disease. Front Physiol 2022; 12:783891. [PMID: 35002766 PMCID: PMC8740047 DOI: 10.3389/fphys.2021.783891] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
TRPV4, a calcium permeable cation selective channel, was found to be involved in chronic obstructive pulmonary disease (COPD) through releasing ATP and IL-1β. Pyroptosis, a newly discovered pro-inflammatory cell death, was induced by cigarette smoke (CS) in airway epithelial cells (AECs). More recent studies indicated that blocking Ca2+ influx effectively inhibited pyroptosis. Therefore, we asked whether TRPV4 mediated CS-induced pyroptosis of AECs and hence participated in the pathogenesis of COPD. We found that pyroptosis and TRPV4 were upregulated in AECs from patients with COPD and long-term CS-exposed mice. Moreover, pharmacological inhibition or knockdown of TRPV4 function alleviated CS extract (CSE)-induced pyroptosis by inhibiting NACHT, LRP, PYD domains-containing protein 3 (NLRP3) inflammasome/activated caspase-1/gasdermin D pathway, decreasing the number of PI positive cells and lactate dehydrogenase (LDH) release, decreasing the expression of pro- inflammatory interleukin gene (IL)-1β, IL-8, and IL-18 expression, as well as increasing anti-inflammatory gene expression [NAD(P)H quinone dehydrogenase 1 (NQO1), superoxide dismutase 2 (mitochondrial) (MNSOD), and catalase, (CAT)]. Moreover, pharmacological inhibition or knockdown of TRPV4 function significantly relieved CSE-induced mitochondrial damage including decreased mitochondrial membrane potential, mitochondrial fusion protein (OPA1, MFN2) expression, and increased mitochondrial fission protein (DRP1, MFF) expression. Taken together, these findings indicate that TRPV4 mediates AEC pyroptosis via NLRP3/caspase-1/GSDMD pathway in COPD.
Collapse
Affiliation(s)
- Yafei Rao
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Xiaoyan Gai
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Jing Xiong
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Yanqing Le
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Yongchang Sun
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| |
Collapse
|
43
|
Meng L, Lin H, Huang X, Weng J, Peng F, Wu S. METTL14 suppresses pyroptosis and diabetic cardiomyopathy by downregulating TINCR lncRNA. Cell Death Dis 2022; 13:38. [PMID: 35013106 PMCID: PMC8748685 DOI: 10.1038/s41419-021-04484-z] [Citation(s) in RCA: 88] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 12/07/2021] [Accepted: 12/17/2021] [Indexed: 12/14/2022]
Abstract
N6-methyladenosine (m6A) is one of the most important epigenetic regulation of RNAs, such as lncRNAs. However, the underlying regulatory mechanism of m6A in diabetic cardiomyopathy (DCM) is very limited. In this study, we sought to define the role of METTL14-mediated m6A modification in pyroptosis and DCM progression. DCM rat model was established and qRT-PCR, western blot, and immunohistochemistry (IHC) were used to detect the expression of METTL14 and TINCR. Gain-and-loss functional experiments were performed to define the role of METTL14-TINCR-NLRP3 axis in pyroptosis and DCM. RNA pulldown and RNA immunoprecipitation (RIP) assays were carried out to verify the underlying interaction. Our results showed that pyroptosis was tightly involved in DCM progression. METTL14 was downregulated in cardiomyocytes and hear tissues of DCM rat tissues. Functionally, METTL14 suppressed pyroptosis and DCM via downregulating lncRNA TINCR, which further decreased the expression of key pyroptosis-related protein, NLRP3. Mechanistically, METTL14 increased m6A methylation level of TINCR gene, resulting in its downregulation. Moreover, the m6A reader protein YTHDF2 was essential for m6A methylation and mediated the degradation of TINCR. Finally, TINCR positively regulated NLRP3 by increasing its mRNA stability. To conclude, our work revealed the novel role of METTL14-mediated m6A methylation and lncRNA regulation in pyroptosis and DCM, which could help extend our understanding the epigenetic regulation of pyroptosis in DCM progression.
Collapse
Affiliation(s)
- Liping Meng
- Department of Cardiology, Shaoxing People's Hospital(Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang, China
| | - Hui Lin
- Department of Cardiology, Shaoxing People's Hospital(Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang, China
| | - Xingxiao Huang
- Department of Cardiology, Shaoxing People's Hospital(Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang, China
| | - Jingfan Weng
- Department of Cardiology, Shaoxing People's Hospital(Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang, China
| | - Fang Peng
- Department of Cardiology, Shaoxing People's Hospital(Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang, China.
| | - Shengjie Wu
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
44
|
Wang H, Luo J, Li A, Su X, Fang C, Xie L, Wu Y, Wen F, Liu Y, Wang T, Zhong Y, Ma L. Proteomic and phosphorylated proteomic landscape of injured lung in juvenile septic rats with therapeutic application of umbilical cord mesenchymal stem cells. Front Immunol 2022; 13:1034821. [PMID: 36341346 PMCID: PMC9635340 DOI: 10.3389/fimmu.2022.1034821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/10/2022] [Indexed: 02/05/2023] Open
Abstract
Acute lung injury (ALI) is the most common complication of sepsis. Intravenous injection of HUMSCs can regulate the level of circulating endothelial cytokines and alleviate lung injury in juvenile septic rats. In this study, we performed proteomic and phosphorylated proteomic analysis of lung tissue of juvenile septic rats after Human Umbilical Cord Mesenchymal Stem Cells (HUMSCs) intervention for the first time, and screened the potential proteins and pathways of HUMSCs for therapeutic effect. The 4D proteome quantitative technique was used to quantitatively analyze the lung tissues of septic rats 24 hours (3 biological samples) and 24 hours after HUMSCs intervention (3 biological samples). A total of 213 proteins were identified as differentially expressed proteins, and 971 phosphorylation sites changed significantly. Based on the public database, we analyzed the functional enrichment of these proteins and phosphorylated proteins. In addition, Tenascin-C may be the key differential protein and ECM receptor interaction pathway may be the main signal pathway by using various algorithms to analyze the protein-protein interaction network. Phosphorylation analysis showed that tight junction pathway was closely related to immune inflammatory reaction, and EGFR interacted most, which may be the key differential phosphorylated protein. Finally, 123 conserved motifs of serine phosphorylation site (pS) and 17 conserved motifs of threonine (pT) phosphorylation sites were identified by motif analysis of phosphorylation sites. Results from proteomics and phosphorylated proteomics, the potential new therapeutic targets of HUMSCs in alleviating lung injury in juvenile septic rats were revealed.
Collapse
Affiliation(s)
- Hongwu Wang
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Department of Hematology and Oncology, Shenzhen Children's Hospital of China Medical University, Shenzhen, China
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, China
| | - Junlin Luo
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Aijia Li
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Xing Su
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Chuiqin Fang
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Lichun Xie
- Department of Hematology and Oncology, Shenzhen Children's Hospital of China Medical University, Shenzhen, China
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, China
- Department of Pediatrics, The Third Affiliated Hospital of Guangzhou Medical University (The Women and Children’s Medical Hospital of Guangzhou Medical University), Guangzhou, China
| | - Yi Wu
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Feiqiu Wen
- Department of Hematology and Oncology, Shenzhen Children's Hospital of China Medical University, Shenzhen, China
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, China
- Department of Hematology and Oncology, Shenzhen Public Service Platform of Molecular Medicine in Pediatric Hematology and Oncology, Shenzhen, China
| | - Yufeng Liu
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tianyou Wang
- Department of Hematology and Oncology, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Yong Zhong
- Department of Pediatrics, The Southeast General Hospital of Dongguan, Dongguan, China
| | - Lian Ma
- Department of Hematology and Oncology, Shenzhen Children's Hospital of China Medical University, Shenzhen, China
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, China
- Department of Pediatrics, The Third Affiliated Hospital of Guangzhou Medical University (The Women and Children’s Medical Hospital of Guangzhou Medical University), Guangzhou, China
- Department of Hematology and Oncology, Shenzhen Public Service Platform of Molecular Medicine in Pediatric Hematology and Oncology, Shenzhen, China
| |
Collapse
|
45
|
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: 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: 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
|
46
|
Wei J, Zhao Y, Liang H, Du W, Wang L. Preliminary evidence for the presence of multiple forms of cell death in diabetes cardiomyopathy. Acta Pharm Sin B 2022; 12:1-17. [PMID: 35127369 PMCID: PMC8799881 DOI: 10.1016/j.apsb.2021.08.026] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/25/2021] [Accepted: 07/30/2021] [Indexed: 12/11/2022] Open
Abstract
Diabetic mellitus (DM) is a common degenerative chronic metabolic disease often accompanied by severe cardiovascular complications (DCCs) as major causes of death in diabetic patients with diabetic cardiomyopathy (DCM) as the most common DCC. The metabolic disturbance in DCM generates the conditions/substrates and inducers/triggers and activates the signaling molecules and death executioners leading to cardiomyocyte death which accelerates the development of DCM and the degeneration of DCM to heart failure. Various forms of programmed active cell death including apoptosis, pyroptosis, autophagic cell death, autosis, necroptosis, ferroptosis and entosis have been identified and characterized in many types of cardiac disease. Evidence has also been obtained for the presence of multiple forms of cell death in DCM. Most importantly, published animal experiments have demonstrated that suppression of cardiomyocyte death of any forms yields tremendous protective effects on DCM. Herein, we provide the most updated data on the subject of cell death in DCM, critical analysis of published results focusing on the pathophysiological roles of cell death, and pertinent perspectives of future studies.
Collapse
Affiliation(s)
- Jinjing Wei
- Department of Endocrinology, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Yongting Zhao
- Department of Endocrinology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Haihai Liang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Weijie Du
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Lihong Wang
- Department of Endocrinology, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| |
Collapse
|
47
|
Ma S, Bi W, Liu X, Li S, Qiu Y, Huang C, Lv R, Yin Q. Single-Cell Sequencing Analysis of the db/db Mouse Hippocampus Reveals Cell-Type-Specific Insights Into the Pathobiology of Diabetes-Associated Cognitive Dysfunction. Front Endocrinol (Lausanne) 2022; 13:891039. [PMID: 35721719 PMCID: PMC9200615 DOI: 10.3389/fendo.2022.891039] [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: 03/07/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetes-associated cognitive decline (DCD), is one of the complications of diabetes, which is characterized by a series of neurophysiological and pathological abnormalities. However, the exact pathogenesis of DCD is still unknown. Single-cell RNA sequencing (scRNA-seq) could discover unusual subpopulations, explore functional heterogeneity and identify signaling pathways and potential markers. The aim of this research was to provide deeper opinion into molecular and cellular changes underlying DCD, identify different cellular types of the diabetic mice hippocampus at single-cell level, and elucidate the factors mediating the pathogenesis of DCD. To elucidate cell specific gene expression changes in the hippocampus of diabetic encephalopathy. Single-cell RNA sequencing of hippocampus from db/m and db/db mice was carried out. Subclustering analysis was performed to further describe microglial cell subpopulations. Interestingly using immunohistochemistry, these findings were confirmed at the protein level. Single cell analysis yielded transcriptome data for 14621 hippocampal cells and defined 11 different cell types. Analysis of differentially expressed genes in the microglia compartments indicated that infection- and immune system process- associated terms, oxidative stress and inflammation play vital roles in the progression of DCD. Compared with db/m mouse, experiments at the protein level supported the activation of microglia, increased expression of inflammatory factors and oxidative stress damage in the hippocampus of db/db mouse. In addition, a major finding of our research was the subpopulation of microglia that express genes related to pro-inflammatory disease-associated microglia (DAM). Our research reveals pathological alterations of inflammation and oxidative stress mediated hippocampal damage in the db/db mice, and may provide potential diagnostic biomarkers and therapeutic interventions for DCD.
Collapse
Affiliation(s)
- Shizhan Ma
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Wenkai Bi
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xueying Liu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shangbin Li
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yaxin Qiu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Chengcheng Huang
- Clinical Education Administration, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Renjun Lv
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Renjun Lv, ; Qingqing Yin,
| | - Qingqing Yin
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Renjun Lv, ; Qingqing Yin,
| |
Collapse
|
48
|
Bai Y, Mu Q, Bao X, Zuo J, Fang X, Hua J, Zhang D, Jiang G, Li P, Gao S, Zhao D. Targeting NLRP3 Inflammasome in the Treatment Of Diabetes and Diabetic Complications: Role of Natural Compounds from Herbal Medicine. Aging Dis 2021; 12:1587-1604. [PMID: 34631209 PMCID: PMC8460305 DOI: 10.14336/ad.2021.0318] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 03/18/2021] [Indexed: 12/11/2022] Open
Abstract
Diabetes, a common metabolic disease with various complications, is becoming a serious global health pandemic. So far there are many approaches in the management of diabetes; however, it still remains irreversible due to its complicated pathogenesis. Recent studies have revealed that nucleotide-binding and oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome plays a vital role in the progression of diabetes and many of its complications, making it a promising therapeutic target in pharmaceutical design. Natural derived herbal medicine, known for its utilization of natural products such as herbs or its bioactive ingredients, is shown to be able to ameliorate hyperglycemia-associated symptoms and to postpone the progression of diabetic complications due to its anti-inflammatory and anti-oxidative properties. In this review, we summarized the role of NLRP3 inflammasome in diabetes and several diabetic complications, as well as 31 active compounds that exert therapeutic effect on diabetic complications via inhibiting NLRP3 inflammasome. Improving our understanding of these promising candidates from natural compounds in herbal medicine targeting NLRP3 inflammasome inspires us the relationship between inflammation and metabolic disorders, and also sheds light on searching potential agents or therapies in the treatment of diabetes and diabetic complications.
Collapse
Affiliation(s)
- Ying Bai
- 1College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qianqian Mu
- 2Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xueli Bao
- 3Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jiacheng Zuo
- 1College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xin Fang
- 3Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Hua
- 3Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Dongwei Zhang
- 1College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Guangjian Jiang
- 1College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ping Li
- 3Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Sihua Gao
- 1College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Dandan Zhao
- 1College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
49
|
Yanan Y, Yi J, Xiaojing L, Jing Q, Xiaohui W. Adipo-specific chemerin knockout alters the metabolomic profile of adipose tissue under normal and high-fat diet conditions: Application of an untargeted liquid chromatography-tandem mass spectrometry metabolomics method. Biomed Chromatogr 2021; 35:e5220. [PMID: 34323295 DOI: 10.1002/bmc.5220] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 06/17/2021] [Accepted: 07/25/2021] [Indexed: 12/17/2022]
Abstract
To explore the metabolic effect of chemerin, adipose-specific chemerin knockout (adipo-chemerin-/- ) male mice were established and fed with 5-week normal diet (ND) or high-fat diet (HFD), and then the glycolipid metabolism index was measured and epididymal adipose tissue metabolomics detected using untargeted LC-tandem mass spectrometry (LC-MS/MS). Under HFD, adipo-chemerin-/- mice showed improved glycolipid metabolism (decreased total cholesterol, low-density lipoprotein-cholesterol, insulin and Homeostasis Model Assessment of Insulin Resistance) compared with flox (control) mice. Furthermore, orthogonal partial least squares-discriminant analysis score plots identified separation of metabolites between adipo-chemerin-/- mice and flox mice fed ND and HFD. Under HFD, 28 metabolites were significantly enhanced in adipo-chemerin-/- mice, and pathway enrichment analysis suggested strong relationship of the differential metabolites with arginine and proline metabolism, phenylalanine metabolism, and phenylalanine, tyrosine and tryptophan biosynthesis, which were directly or indirectly related to lipid metabolism, inflammation and oxidative stress. Under ND, taurine was increased in adipo-chemerin-/- mice, resulting in taurine and hypotaurine metabolism and primary bile acid biosynthesis. In conclusion, the improved effect of chemerin knockdown on the glycolipid metabolism of HFD-feeding male mice might be associated with the increases in differential metabolites and metabolic pathways involved in lipid metabolism, inflammation and oxidative stress, which provided insights into the mechanism of chemerin from a metabolomics aspect.
Collapse
Affiliation(s)
- Yang Yanan
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jia Yi
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Lin Xiaojing
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Qu Jing
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Wang Xiaohui
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
50
|
Ji N, Qi Z, Wang Y, Yang X, Yan Z, Li M, Ge Q, Zhang J. Pyroptosis: A New Regulating Mechanism in Cardiovascular Disease. J Inflamm Res 2021; 14:2647-2666. [PMID: 34188515 PMCID: PMC8235951 DOI: 10.2147/jir.s308177] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 06/02/2021] [Indexed: 12/17/2022] Open
Abstract
Pyroptosis is a kind of pro-inflammatory cell death. Compared with autophagy and apoptosis, pyroptosis has unique characteristics in morphology and mechanism. Specifically, pyroptosis is a kind of cell lysis mediated by the Gasdermin family, releases inflammatory cytokines IL-1β and IL-18. There are three different forms of mechanism, which are caspase-1-mediated, caspase-4/5/11-mediated and caspase-3-mediated. A large number of studies have proved that pyroptosis is closely related to cardiovascular disease. This paper reviewed the recent progress in the related research on pyroptosis and myocardial infarction, ischemia-reperfusion, atherosclerosis, diabetic cardiomyopathy, arrhythmia, heart failure hypertension and Kawasaki disease. Therefore, we believe that pyroptosis may be a new therapeutic target in the cardiovascular field.
Collapse
Affiliation(s)
- Nan Ji
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, People's Republic of China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, People's Republic of China
| | - Zhongwen Qi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, People's Republic of China
| | - Yueyao Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, People's Republic of China
| | - Xiaoya Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, People's Republic of China
| | - Zhipeng Yan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, People's Republic of China
| | - Meng Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, People's Republic of China
| | - Qihui Ge
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, People's Republic of China
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, People's Republic of China
| |
Collapse
|