1
|
Li MR, Lu LQ, Zhang YY, Yao BF, Tang C, Dai SY, Luo XJ, Peng J. Sonic hedgehog signaling facilitates pyroptosis in mouse heart following ischemia/reperfusion via enhancing the formation of CARD10-BCL10-MALT1 complex. Eur J Pharmacol 2024; 984:177019. [PMID: 39343081 DOI: 10.1016/j.ejphar.2024.177019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/05/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Pyroptosis has been found to contribute to myocardial ischemia/reperfusion (I/R) injury, but the exact mechanisms that initiate myocardial pyroptosis are not fully elucidated. Sonic hedgehog (SHH) signaling is activated in heart suffered I/R, and intervention of SHH signaling has been demonstrated to protect heart from I/R injury. Caspase recruitment domain-containing protein 10 (CARD10)-B cell lymphoma 10 (BCL10)-mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) (CBM) complex could transduce signals from the membrane and induce inflammatory pathways in non-hematopoietic cells, which could be a downstream effector of SHH signaling pathway. This study aims to explore the role of SHH signaling in I/R-induced myocardial pyroptosis and its relationship with the CBM complex. C57BL/6J mice were subjected to 45 min-ischemia followed by 24 h-reperfusion to establish a myocardial I/R model, and H9c2 cells underwent hypoxia/reoxygenation (H/R) to mimic myocardial I/R model in vitro. Firstly, SHH signaling was significantly activated in heart suffered I/R in an autocrine- or paracrine-dependent manner via its receptor PTCH1, and inhibition of SHH signaling decreased myocardial injury via reducing caspase-11-dependent pyroptosis, concomitant with attenuating CBM complex formation. Secondly, suppression of SHH signaling decreased protein kinase C α (PKCα) level, but inhibition of PKCα attenuated CBM complex formation without impacting the protein levels of SHH and PTCH1. Finally, disruption of the CBM complex prevented MALT1 from recruiting of TRAF6, which was believed to trigger the caspase-11-dependent pyroptosis. Based on these results, we conclude that inhibition of SHH signaling suppresses pyroptosis via attenuating PKCα-mediated CARD10-BCL10-MALT1 complex formation in mouse heart suffered I/R.
Collapse
Affiliation(s)
- Ming-Rui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Li-Qun Lu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Yi-Yue Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Bi-Feng Yao
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Can Tang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Shu-Yan Dai
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
| |
Collapse
|
2
|
Ma X, Xie J, Li B, Shan H, Jia Z, Liu W, Dong Y, Han S, Jin Q. Weighted gene co-expression network analysis and single-cell sequence analysis uncover immune landscape and reveal hub genes of necroptosis in macrophages in myocardial ischaemia-reperfusion injury. Int Immunopharmacol 2024; 140:112761. [PMID: 39079349 DOI: 10.1016/j.intimp.2024.112761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/08/2024] [Accepted: 07/22/2024] [Indexed: 09/01/2024]
Abstract
Myocardial ischaemia-reperfusion injury (MIRI) caused by the treatment of acute myocardial infarction (AMI) is the primary cause of severe ventricular remodelling, heart failure (HF), and high mortality. In recent studies, research on the role of necroptosis in MIRI has focused on cardiomyocytes, but new biomarkers and immunocyte mechanisms of necroptosis are rarely studied. In the present study, weighted gene co-expression network analysis (WGCNA) algorithms were used to establish a weighted gene co-expression network, and Casp1, Hpse, Myd88, Ripk1, and Tpm3 were identified as biological markers of necroptosis using least absolute shrinkage, selection operator (LASSO) regression and support vector machine (SVM) feature selection algorithms. The role and discriminatory power of these five genes in MIRI had never been studied. Single-cell and cell-talk analyses showed that hub genes of necroptosis were focused on macrophages, which mediate the functions of monocytes, fibroblasts, haematopoietic stem cells, and cardiomyocytes, primarily through the TNF/TNFRSF1A interaction. The polarisation and functional activation of macrophages were affected by the MIF signalling network (MIF CD74/CXCR4 and MIF CD74/CD44) of other cells. The results of the immune infiltration assay showed that the five genes involved in necroptosis were significantly related to the infiltration and functional activity of M2 macrophages. TWS-119 is predicted to be a molecular drug that targets key MIRI genes. A mouse model was established to confirm the expression of five hub genes, and ventricular remodelling increased with time after ischaemia-reperfusion injury (IRI). Therefore, Casp1, Hpse, Myd88, Ripk1, and Tpm3 may be key genes regulating necroptosis and polarisation in macrophages, and causing ventricular remodelling.
Collapse
Affiliation(s)
- Xiaowen Ma
- 960th Hospital of the Joint Logistic Support Force, China
| | - Jiqing Xie
- 960th Hospital of the Joint Logistic Support Force, China
| | - Bin Li
- 960th Hospital of the Joint Logistic Support Force, China
| | - Hui Shan
- 960th Hospital of the Joint Logistic Support Force, China
| | - Zonghu Jia
- 960th Hospital of the Joint Logistic Support Force, China
| | - Wenyan Liu
- 960th Hospital of the Joint Logistic Support Force, China
| | - Yubo Dong
- 960th Hospital of the Joint Logistic Support Force, China
| | - Shufang Han
- 960th Hospital of the Joint Logistic Support Force, China.
| | - Qun Jin
- 960th Hospital of the Joint Logistic Support Force, 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
|
Li J, Zhang J, Zhong Y, Xie D, Han H, Zhang Z, Liu Y, Li S. TRPC6 regulates necroptosis in myocardial ischemia/reperfusion injury via Ca 2+/CaMKII signaling pathway. Cell Signal 2024; 122:111344. [PMID: 39134250 DOI: 10.1016/j.cellsig.2024.111344] [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: 03/17/2024] [Revised: 07/31/2024] [Accepted: 08/09/2024] [Indexed: 08/16/2024]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) frequently complicates postoperative cardiovascular disease treatment. Necroptosis, a cell death mechanism similar to apoptosis, is regulated by specific signaling pathways and plays an important role in MIRI. Receptor-interacting protein 3 (RIP3), a key protein regulating necroptosis during MIRI, directly phosphorylates calmodulin-dependent protein kinase II (CaMKII). Leading to mitochondrial permeablity transition pore (mPTP) opening and inducing necroptosis. Transient receptor potential canonical channel 6 (TRPC6) regulats Ca2+ entry, is linked to CaMKII as an important upstream effector. However, the connection between TRPC6 and MIRI necroptosis remains unclear. The study aimed to investigate the relationship between TRPC6 and MIRI necroptosis, with a specific focus on elucidating the role of TRPC6 in regulating CaMKII phosphorylation during cardiac necroptosis via Ca2+ modulation. METHODS AND RESULTS: The experiment used wild-type (WT) and TRPC6 knockout (TRPC6-/-) mice for I/R model construction, and H9c2 myocardial cell line for H/R model. After ischemia-reperfusion (I/R), TRPC6 protein levels in mice significantly increased, exacerbating myocardial injury, infarct size (IS), and cardiac function in WT mice. In contrast, TRPC6 knockout attenuated myocardial injury, IS, and improved cardiac function. The results showed a significant correlation between changes in CaMKII and TRPC6. TRPC6 knockout led to decreased intracellular calcium levels, CaMKII phosphorylation, reactive oxygen species levels, mPTP opening, and improve mitochondrial structure. CONCLUSION: I/R upregulates TRPC6, which mediates Ca2+ entry and CaMKII phosphorylation, exacerbates oxidative stress, and induces necroptosis. These findings suggest a potential therapeutic avenue for mitigating MIRI by targeting TRPC6.
Collapse
Affiliation(s)
- Junhao Li
- Department of Forensic Pathology, School of Forensic Medicine, Zunyi Medical University, Zunyi 563006, Guizhou, China
| | - Jiaji Zhang
- Department of Forensic Pathology, School of Forensic Medicine, Zunyi Medical University, Zunyi 563006, Guizhou, China
| | - Yunlong Zhong
- Department of Forensic Pathology, School of Forensic Medicine, Zunyi Medical University, Zunyi 563006, Guizhou, China
| | - Dongge Xie
- Department of Forensic Pathology, School of Forensic Medicine, Zunyi Medical University, Zunyi 563006, Guizhou, China
| | - Han Han
- Department of Forensic Pathology, School of Forensic Medicine, Zunyi Medical University, Zunyi 563006, Guizhou, China
| | - Zhongqing Zhang
- Department of Forensic Pathology, School of Forensic Medicine, Zunyi Medical University, Zunyi 563006, Guizhou, China
| | - Yong Liu
- Department of Forensic Pathology, School of Forensic Medicine, Zunyi Medical University, Zunyi 563006, Guizhou, China
| | - Shoutian Li
- Department of Forensic Pathology, School of Forensic Medicine, Zunyi Medical University, Zunyi 563006, Guizhou, China.
| |
Collapse
|
5
|
Kobayashi Y, Li J, Parker M, Wang J, Nagy A, Fan CPS, Runeckles K, Okumura M, Kadowaki S, Honjo O. Impact of Hemoglobin Level in Ex Vivo Heart Perfusion on Donation After Circulatory Death Hearts: A Juvenile Porcine Experimental Model. Transplantation 2024; 108:1922-1930. [PMID: 39167562 DOI: 10.1097/tp.0000000000004954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
BACKGROUND Ex vivo heart perfusion (EVHP) of donation after circulatory death (DCD) hearts has become an effective strategy in adults; however, the small circulating volume in pediatrics poses the challenge of a low-hemoglobin (Hb) perfusate. We aimed to determine the impact of perfusate Hb levels during EVHP on DCD hearts using a juvenile porcine model. METHODS Sixteen DCD piglet hearts (11-14 kg) were reperfused for 4 h in unloaded mode followed by working mode. Metabolism, cardiac function, and cell damage were compared between the low-Hb (Hb, 5.0-5.9 g/dL; n = 8) and control (Hb, 7.5-8.4 g/dL; n = 8) groups. Between-group differences were evaluated using 2-sample t -tests or Fisher's Exact tests. RESULTS During unloaded mode, the low-Hb group showed lower myocardial oxygen consumption ( P < 0.001), a higher arterial lactate level ( P = 0.001), and worse systolic ventricular function ( P < 0.001). During working mode, the low-Hb group had a lower cardiac output (mean, 71% versus 106% of normal cardiac output, P = 0.010) and a higher arterial lactate level ( P = 0.031). Adjusted cardiac troponin-I ( P = 0.112) did not differ between the groups. Morphological myocyte injury in the left ventricle was more severe in the low-Hb group ( P = 0.028). CONCLUSIONS Low-Hb perfusate with inadequate oxygen delivery induced anaerobic metabolism, resulting in suboptimal DCD heart recovery and declined cardiac function. Arranging an optimal perfusate is crucial to organ protection, and further endeavors to refine the priming volume of EVHP or the transfusion strategy are required.
Collapse
Affiliation(s)
- Yasuyuki Kobayashi
- Division of Cardiovascular Surgery, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Jing Li
- Division of Cardiovascular Surgery, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Marlee Parker
- Division of Perfusion Services, The Hospital for Sick Children, Toronto, ON, Canada
| | - Jian Wang
- Division of Perfusion Services, The Hospital for Sick Children, Toronto, ON, Canada
| | - Anita Nagy
- Division of Pathology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Chun-Po Steve Fan
- Ted Rogers Computational Program, Ted Rogers Centre for Heart Research, Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Kyle Runeckles
- Ted Rogers Computational Program, Ted Rogers Centre for Heart Research, Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Michiru Okumura
- Division of Cardiovascular Surgery, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Sachiko Kadowaki
- Division of Cardiovascular Surgery, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Osami Honjo
- Division of Cardiovascular Surgery, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
6
|
Yan X, Yang P, Yang C, Wang Y, Feng Z, Liu T, Li Y, Zhou C, Li M. Ferroptosis-Associated Extracellular Matrix Remodeling in Radiation-Induced Lung Fibrosis Progression. Dose Response 2024; 22:15593258241289829. [PMID: 39351078 PMCID: PMC11440530 DOI: 10.1177/15593258241289829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 09/05/2024] [Indexed: 10/04/2024] Open
Abstract
Background: Radiation-induced lung fibrosis (RILF) is a life-threatening complication of thoracic radiotherapy. Ferroptosis, a recently discovered type of cell death, is believed to contribute to RILF, though the associated mechanisms are unknown. This study aimed to investigate the potential mechanism of ferroptosis in RILF and examine the contribution of different cell types to ferroptosis during RILF progression. Methods: Histopathological changes in RILF lung tissue were assessed through H&E and Masson staining. IHC staining investigated ferroptosis markers (GPX4, ACSL4, NCOA4). Ferroptosis-related genes (FRG) and pathway scores were derived from RILF transcriptome microarray data. The sc-RNAseq analysis detected FRG score dynamics across cell types, validated by IF staining for PDGFR-α and ACSL4. Results: ACSL4 and NCOA4 protein levels were significantly higher and GPX4 lower in IR than control. FRG scores were positively correlated with fibrosis-related pathway scores in the RILF transcriptome data. FRG and ECM scores were concurrently upregulated in myofibroblasts. Enhanced co-staining of PDGFR-α and ACSL4 were observed in the fibrotic areas of RILF lungs. Conclusions: Our research indicated that in RILF, fibroblasts undergoing ferroptosis may release increased levels of ECM, potentially accelerating the progression of lung fibrosis. This finding presents ferroptosis as a potential therapeutic target in RILF.
Collapse
Affiliation(s)
- Xinyu Yan
- Zhongshan City People’s Hospital, Xinxiang Medical University, Xinxiang, China
- Department of Radiation Oncology, Zhongshan City People’s Hospital, Zhongshan, China
| | - Peixuan Yang
- Zhongshan City People’s Hospital, Xinxiang Medical University, Xinxiang, China
- Department of Radiation Oncology, Zhongshan City People’s Hospital, Zhongshan, China
| | - Chen Yang
- Zhongshan City People’s Hospital, Xinxiang Medical University, Xinxiang, China
- Department of Radiation Oncology, Zhongshan City People’s Hospital, Zhongshan, China
| | - Yinghui Wang
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, NMPA Key Laboratory for Safety Evaluation of Cosmetics, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zhijun Feng
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, NMPA Key Laboratory for Safety Evaluation of Cosmetics, School of Public Health, Southern Medical University, Guangzhou, China
| | - Ting Liu
- Department of Radiation Oncology, Zhongshan City People’s Hospital, Zhongshan, China
| | - Yani Li
- Department of Radiation Oncology, Zhongshan City People’s Hospital, Zhongshan, China
| | - Cheng Zhou
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Minying Li
- Department of Radiation Oncology, Zhongshan City People’s Hospital, Zhongshan, China
| |
Collapse
|
7
|
Peng F, Liao M, Jin W, Liu W, Li Z, Fan Z, Zou L, Chen S, Zhu L, Zhao Q, Zhan G, Ouyang L, Peng C, Han B, Zhang J, Fu L. 2-APQC, a small-molecule activator of Sirtuin-3 (SIRT3), alleviates myocardial hypertrophy and fibrosis by regulating mitochondrial homeostasis. Signal Transduct Target Ther 2024; 9:133. [PMID: 38744811 PMCID: PMC11094072 DOI: 10.1038/s41392-024-01816-1] [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: 08/15/2023] [Revised: 02/20/2024] [Accepted: 03/25/2024] [Indexed: 05/16/2024] Open
Abstract
Sirtuin 3 (SIRT3) is well known as a conserved nicotinamide adenine dinucleotide+ (NAD+)-dependent deacetylase located in the mitochondria that may regulate oxidative stress, catabolism and ATP production. Accumulating evidence has recently revealed that SIRT3 plays its critical roles in cardiac fibrosis, myocardial fibrosis and even heart failure (HF), through its deacetylation modifications. Accordingly, discovery of SIRT3 activators and elucidating their underlying mechanisms of HF should be urgently needed. Herein, we identified a new small-molecule activator of SIRT3 (named 2-APQC) by the structure-based drug designing strategy. 2-APQC was shown to alleviate isoproterenol (ISO)-induced cardiac hypertrophy and myocardial fibrosis in vitro and in vivo rat models. Importantly, in SIRT3 knockout mice, 2-APQC could not relieve HF, suggesting that 2-APQC is dependent on SIRT3 for its protective role. Mechanically, 2-APQC was found to inhibit the mammalian target of rapamycin (mTOR)-p70 ribosomal protein S6 kinase (p70S6K), c-jun N-terminal kinase (JNK) and transforming growth factor-β (TGF-β)/ small mother against decapentaplegic 3 (Smad3) pathways to improve ISO-induced cardiac hypertrophy and myocardial fibrosis. Based upon RNA-seq analyses, we demonstrated that SIRT3-pyrroline-5-carboxylate reductase 1 (PYCR1) axis was closely assoiated with HF. By activating PYCR1, 2-APQC was shown to enhance mitochondrial proline metabolism, inhibited reactive oxygen species (ROS)-p38 mitogen activated protein kinase (p38MAPK) pathway and thereby protecting against ISO-induced mitochondrialoxidative damage. Moreover, activation of SIRT3 by 2-APQC could facilitate AMP-activated protein kinase (AMPK)-Parkin axis to inhibit ISO-induced necrosis. Together, our results demonstrate that 2-APQC is a targeted SIRT3 activator that alleviates myocardial hypertrophy and fibrosis by regulating mitochondrial homeostasis, which may provide a new clue on exploiting a promising drug candidate for the future HF therapeutics.
Collapse
Affiliation(s)
- Fu Peng
- West China School of Pharmacy and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Minru Liao
- West China School of Pharmacy and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenke Jin
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Wei Liu
- West China School of Pharmacy and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zixiang Li
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Zhichao Fan
- West China School of Pharmacy and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ling Zou
- West China School of Pharmacy and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Siwei Chen
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Lingjuan Zhu
- School of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Qian Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Gu Zhan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Liang Ouyang
- West China School of Pharmacy and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Jin Zhang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, China.
| | - Leilei Fu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
8
|
Zhang BS, Zhang XM, Ito M, Yajima S, Yoshida K, Ohno M, Nishi E, Wang H, Li SY, Kubota M, Yoshida Y, Matsutani T, Mine S, Machida T, Takemoto M, Yamagata H, Hayashi A, Yokote K, Kobayashi Y, Takizawa H, Kuroda H, Shimada H, Iwadate Y, Hiwasa T. JMJD6 Autoantibodies as a Potential Biomarker for Inflammation-Related Diseases. Int J Mol Sci 2024; 25:4935. [PMID: 38732153 PMCID: PMC11084951 DOI: 10.3390/ijms25094935] [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: 03/15/2024] [Revised: 04/15/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Inflammation is closely associated with cerebrovascular diseases, cardiovascular diseases, diabetes, and cancers, and it is accompanied by the development of autoantibodies in the early stage of inflammation-related diseases. Hence, it is meaningful to discover novel antibody biomarkers targeting inflammation-related diseases. In this study, Jumonji C-domain-containing 6 (JMJD6) was identified by the serological identification of antigens through recombinant cDNA expression cloning. In particular, JMJD6 is an antigen recognized in serum IgG from patients with unstable angina pectoris (a cardiovascular disease). Then, the serum antibody levels were examined using an amplified luminescent proximity homogeneous assay-linked immunosorbent assay and a purified recombinant JMJD6 protein as an antigen. We observed elevated levels of serum anti-JMJD6 antibodies (s-JMJD6-Abs) in patients with inflammation-related diseases such as ischemic stroke, acute myocardial infarction (AMI), diabetes mellitus (DM), and cancers (including esophageal cancer, EC; gastric cancer; lung cancer; and mammary cancer), compared with the levels in healthy donors. The s-JMJD6-Ab levels were closely associated with some inflammation indicators, such as C-reactive protein and intima-media thickness (an atherosclerosis index). A better postoperative survival status of patients with EC was observed in the JMJD6-Ab-positive group than in the negative group. An immunohistochemical analysis showed that JMJD6 was highly expressed in the inflamed mucosa of esophageal tissues, esophageal carcinoma tissues, and atherosclerotic plaques. Hence, JMJD6 autoantibodies may reflect inflammation, thereby serving as a potential biomarker for diagnosing specific inflammation-related diseases, including stroke, AMI, DM, and cancers, and for prediction of the prognosis in patients with EC.
Collapse
Affiliation(s)
- Bo-Shi Zhang
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (B.-S.Z.)
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Xiao-Meng Zhang
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Masaaki Ito
- Department of Clinical Oncology, Graduate School of Medicine, Toho University, Tokyo 143-8541, Japan (H.S.)
| | - Satoshi Yajima
- Department of Gastroenterological Surgery, Graduate School of Medicine, Toho University, Tokyo 143-8541, Japan
| | - Kimihiko Yoshida
- Department of Gastroenterological Surgery, Graduate School of Medicine, Toho University, Tokyo 143-8541, Japan
| | - Mikiko Ohno
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
- Department of Pharmacology, Shiga University of Medical Science, Otsu 520-2192, Japan
| | - Eiichiro Nishi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
- Department of Pharmacology, Shiga University of Medical Science, Otsu 520-2192, Japan
| | - Hao Wang
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Shu-Yang Li
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (B.-S.Z.)
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Masaaki Kubota
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (B.-S.Z.)
- Comprehensive Stroke Center, Chiba University Hospital, Chiba 260-8677, Japan
| | - Yoichi Yoshida
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (B.-S.Z.)
- Comprehensive Stroke Center, Chiba University Hospital, Chiba 260-8677, Japan
| | - Tomoo Matsutani
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (B.-S.Z.)
| | - Seiichiro Mine
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (B.-S.Z.)
- Department of Neurological Surgery, Chiba Prefectural Sawara Hospital, Chiba 287-0003, Japan
- Department of Neurological Surgery, Chiba Cerebral and Cardiovascular Center, Chiba 290-0512, Japan
| | - Toshio Machida
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (B.-S.Z.)
- Department of Neurological Surgery, Chiba Cerebral and Cardiovascular Center, Chiba 290-0512, Japan
- Department of Neurosurgery, Eastern Chiba Medical Center, Chiba 283-8686, Japan
| | - Minoru Takemoto
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Department of Diabetes, Metabolism and Endocrinology, School of Medicine, International University of Health and Welfare, Chiba 286-8686, Japan
| | - Hiroki Yamagata
- Department of Diabetes, Metabolism and Endocrinology, School of Medicine, International University of Health and Welfare, Chiba 286-8686, Japan
| | - Aiko Hayashi
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Koutaro Yokote
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Yoshio Kobayashi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Hirotaka Takizawa
- Port Square Kashiwado Clinic, Kashiwado Memorial Foundation, Chiba 260-0025, Japan
| | - Hideyuki Kuroda
- Medical Project Division, Research Development Center, Fujikura Kasei Co., Saitama 340-0203, Japan
| | - Hideaki Shimada
- Department of Clinical Oncology, Graduate School of Medicine, Toho University, Tokyo 143-8541, Japan (H.S.)
- Department of Gastroenterological Surgery, Graduate School of Medicine, Toho University, Tokyo 143-8541, Japan
| | - Yasuo Iwadate
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (B.-S.Z.)
| | - Takaki Hiwasa
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (B.-S.Z.)
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Department of Clinical Oncology, Graduate School of Medicine, Toho University, Tokyo 143-8541, Japan (H.S.)
- Comprehensive Stroke Center, Chiba University Hospital, Chiba 260-8677, Japan
| |
Collapse
|
9
|
Neves LT, Paz LV, Wieck A, Mestriner RG, de Miranda Monteiro VAC, Xavier LL. Environmental Enrichment in Stroke Research: an Update. Transl Stroke Res 2024; 15:339-351. [PMID: 36717476 DOI: 10.1007/s12975-023-01132-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 01/25/2023] [Accepted: 01/25/2023] [Indexed: 02/01/2023]
Abstract
Environmental enrichment (EE) refers to different forms of stimulation, where the environment is designed to improve the levels of sensory, cognitive, and motor stimuli, inducing stroke recovery in animal models. Stroke is a leading cause of mortality and neurological disability among older adults, hence the importance of developing strategies to improve recovery for such patients. This review provides an update on recent findings, compiling information regarding the parameters affected by EE exposure in both preclinical and clinical studies. During stroke recovery, EE exposure has been shown to improve both the cognitive and locomotor aspects, inducing important neuroplastic alterations, increased angiogenesis and neurogenesis, and modified gene expression, among other effects. There is a need for further research in this field, particularly in those aspects where the evidence is inconclusive. Moreover, it is necessary refine and adapt the EE paradigms for application in human patients.
Collapse
Affiliation(s)
- Laura Tartari Neves
- Programa de Pós-Graduação Em Biologia Celular E Molecular, Laboratório deBiologiaCelular ETecidual, Pontifical Catholic University of Rio Grande Do Sul, PUCRS. Escola de Ciências da Saúde E da Vida, Av. Ipiranga 6681, Prédio 12C, Sala 104, Porto Alegre, Rio Grande Do Sul, CEP, 90619-900, Brazil
| | - Lisiê Valéria Paz
- Programa de Pós-Graduação Em Biologia Celular E Molecular, Laboratório deBiologiaCelular ETecidual, Pontifical Catholic University of Rio Grande Do Sul, PUCRS. Escola de Ciências da Saúde E da Vida, Av. Ipiranga 6681, Prédio 12C, Sala 104, Porto Alegre, Rio Grande Do Sul, CEP, 90619-900, Brazil
| | - Andréa Wieck
- Programa de Pós-Graduação Em Biologia Celular E Molecular, Laboratório deBiologiaCelular ETecidual, Pontifical Catholic University of Rio Grande Do Sul, PUCRS. Escola de Ciências da Saúde E da Vida, Av. Ipiranga 6681, Prédio 12C, Sala 104, Porto Alegre, Rio Grande Do Sul, CEP, 90619-900, Brazil
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690 - Jardim Botânico, Porto Alegre, RS, 90610-000, Brazil
| | - Régis Gemerasca Mestriner
- Programa de Pós-Graduação Em Biologia Celular E Molecular, Laboratório deBiologiaCelular ETecidual, Pontifical Catholic University of Rio Grande Do Sul, PUCRS. Escola de Ciências da Saúde E da Vida, Av. Ipiranga 6681, Prédio 12C, Sala 104, Porto Alegre, Rio Grande Do Sul, CEP, 90619-900, Brazil
| | - Valentina Aguiar Cardozo de Miranda Monteiro
- Programa de Pós-Graduação Em Biologia Celular E Molecular, Laboratório deBiologiaCelular ETecidual, Pontifical Catholic University of Rio Grande Do Sul, PUCRS. Escola de Ciências da Saúde E da Vida, Av. Ipiranga 6681, Prédio 12C, Sala 104, Porto Alegre, Rio Grande Do Sul, CEP, 90619-900, Brazil
| | - Léder Leal Xavier
- Programa de Pós-Graduação Em Biologia Celular E Molecular, Laboratório deBiologiaCelular ETecidual, Pontifical Catholic University of Rio Grande Do Sul, PUCRS. Escola de Ciências da Saúde E da Vida, Av. Ipiranga 6681, Prédio 12C, Sala 104, Porto Alegre, Rio Grande Do Sul, CEP, 90619-900, Brazil.
| |
Collapse
|
10
|
Du Y, Li L, Li X, Tan J, Qin Y, Lv Y, Zhai X. Synergistic effects and molecular mechanisms of DL-3-n-butylphthalide combined with dual antiplatelet therapy in acute ischemic stroke. Int Immunopharmacol 2024; 129:111592. [PMID: 38295546 DOI: 10.1016/j.intimp.2024.111592] [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: 11/09/2023] [Accepted: 01/23/2024] [Indexed: 02/02/2024]
Abstract
DL-3-n-butylphthalide (NBP) is isolated from the seeds of Apium graveolens L., and has been recently used as a neuroprotective agent for acute ischemic stroke. The present study aimed to determine the efficacy and safety of the combined use of dual antiplatelet therapy (DAPT) and NBP for treating of acute ischemic stroke in rats and to explore the synergistic mechanism of this treatment strategy in rat middle cerebral artery occlusion models. The efficacy of DAPT combined with NBP was evaluated by determining neurological deficits, infarction status, and histological changes. Changes in body weight, blood glucose level, blood count, and serum biochemical parameters were detected to evaluate the safety. To explore the synergistic pharmacological mechanism, the mRNA expression and protein levels of key proteins in the pyroptosis-inflammatory pathway, and the pyroptosis ratio of microglias were examined. Compared with the administration of NBP or DAPT alone, combination of them significantly improved neurological deficits, reduced infarct area, and repaired tissue injury and inflammation after cerebral ischemia. No hepatorenal toxicity was observed. The mRNA expression and protein levels of key proteins in the pyroptosis-inflammation pathway, and the pyroptosis ratio of microglias were significantly downregulated in the combined administration group than in the monotherapy group. We demonstrated that the combined use of NBP and DAPT exhibits better efficacy and high safety and plays a synergistic role by inhibiting the pyroptosis-inflammation pathway in the brain tissues, particularly in microglial cells.
Collapse
Affiliation(s)
- Yujing Du
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Linjie Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xixuan Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jingxuan Tan
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yanjie Qin
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yongning Lv
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Clinical Research Centre for Precision Medicine for Critical Illness, Wuhan, China.
| | - Xuejia Zhai
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Clinical Research Centre for Precision Medicine for Critical Illness, Wuhan, China.
| |
Collapse
|
11
|
Yang Y, Zhu Y, Liu C, Cheng J, He F. Taohong Siwu decoction reduces acute myocardial ischemia-reperfusion injury by promoting autophagy to inhibit pyroptosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117515. [PMID: 38042386 DOI: 10.1016/j.jep.2023.117515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 11/19/2023] [Accepted: 11/25/2023] [Indexed: 12/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Taohong Siwu decoction (TSD) is a classic traditional Chinese medicine (TCM) prescription used to promote the blood circulation and alleviate blood stasis. TSD consists of Paeonia lactiflora Pall., Conioselinum anthriscoides (H. Boissieu) Pimenov & Kljuykov, Rehmannia glutinosa (Gaertn.) DC., Prunus persica (L.) Batsch, Angelica sinensis (Oliv.) Diels, and Carthamus creticus L. in the ratio of 3:2:4:3:3:2. Studies on the effects of TSD on myocardial ischemia-reperfusion injury (MIRI) from the perspective of autophagy and pyroptosis have not been reported. AIM OF THE STUDY Investigate the effect of TSD on MIRI and explore the underlying mechanisms. MATERIALS AND METHODS We searched the main components and corresponding potential targets of TSD on The Pharmacology of Traditional Chinese Medicine Systems database for target prediction. We identified targets for MIRI on Online Mendelian Inheritance in Man and GeneCards databases. The intersection of the compound target and disease target was obtained and a protein-protein interaction network constructed. We undertook enrichment analyses using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes databases. The results of network pharmacology were verified by in vivo experiments in mice. RESULTS In mice, TSD significantly reduced the volume of the myocardial infarct, significantly reduced serum levels of cardiac troponin-nI (CTnI), creatine kinase-myocardial band (CK-MB), malonaldehyde (MDA), interleukin (IL)-6, increased the activity of superoxide dismutase (SOD) and IL-10 level, reduced the level of pyroptosis in myocardial tissue, increased the number of autophagosomes, and significantly reduced the fluorescence intensity of apoptosis-associated speck-like protein (ASC), Nod-like receptor protein 3 (NLRP3), and caspase-1. TSD administration increased the protein expression of microtubule-associated protein light chain 3 (LC3), but reduced the protein expression of p62, NLRP3, ASC, caspase-1, cleaved caspase-1, pro-caspase-1, gasdermin D (GSDMD), GSDMD-N-terminal, IL-18, and IL-1β. Administration of 3-Methyladenin could reverse the effect of TSD in inhibiting inflammation and the release of proinflammatory factors. CONCLUSION TSD treatment alleviated MIRI by promoting autophagy to suppress activation of the NLRP3 inflammasome and reducing the release of proinflammatory factors.
Collapse
Affiliation(s)
- Yuming Yang
- College of the Nursing, Anhui University of Chinese Medicine, Hefei, China
| | - Ying Zhu
- College of the Nursing, Anhui University of Chinese Medicine, Hefei, China
| | - Changyi Liu
- College of the Nursing, Anhui University of Chinese Medicine, Hefei, China
| | - Jing Cheng
- College of the Nursing, Anhui University of Chinese Medicine, Hefei, China.
| | - Fei He
- Department of Cardiology, Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
12
|
Ni D, Lei C, Liu M, Peng J, Yi G, Mo Z. Cell death in atherosclerosis. Cell Cycle 2024; 23:495-518. [PMID: 38678316 PMCID: PMC11135874 DOI: 10.1080/15384101.2024.2344943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 04/14/2024] [Indexed: 04/29/2024] Open
Abstract
A complex and evolutionary process that involves the buildup of lipids in the arterial wall and the invasion of inflammatory cells results in atherosclerosis. Cell death is a fundamental biological process that is essential to the growth and dynamic equilibrium of all living things. Serious cell damage can cause a number of metabolic processes to stop, cell structure to be destroyed, or other irreversible changes that result in cell death. It is important to note that studies have shown that the two types of programmed cell death, apoptosis and autophagy, influence the onset and progression of atherosclerosis by controlling these cells. This could serve as a foundation for the creation of fresh atherosclerosis prevention and treatment strategies. Therefore, in this review, we summarized the molecular mechanisms of cell death, including apoptosis, pyroptosis, autophagy, necroptosis, ferroptosis and necrosis, and discussed their effects on endothelial cells, vascular smooth muscle cells and macrophages in the process of atherosclerosis, so as to provide reference for the next step to reveal the mechanism of atherosclerosis.
Collapse
Affiliation(s)
- Dan Ni
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, Guangxi, China
| | - Cai Lei
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Minqi Liu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, Guangxi, China
- Guangxi Province Postgraduate Co-training Base for Cooperative Innovation in Basic Medicine (Guilin Medical University and Yueyang Women & Children’s Medical Center), Yueyang, China
| | - Jinfu Peng
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Guanghui Yi
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Zhongcheng Mo
- Guangxi Key Laboratory of Diabetic Systems Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, Guangxi, China
- Guangxi Province Postgraduate Co-training Base for Cooperative Innovation in Basic Medicine (Guilin Medical University and Yueyang Women & Children’s Medical Center), Yueyang, China
| |
Collapse
|
13
|
Yang Y, Lin X. Potential relationship between autophagy and ferroptosis in myocardial ischemia/reperfusion injury. Genes Dis 2023; 10:2285-2295. [PMID: 37554184 PMCID: PMC10404879 DOI: 10.1016/j.gendis.2022.02.012] [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: 09/29/2021] [Revised: 01/19/2022] [Accepted: 02/08/2022] [Indexed: 11/19/2022] Open
Abstract
Autophagy is an evolutionarily conserved process involved in the degradation of long-lived proteins and excessive or dysfunctional organelles. As a pivotal cellular response, autophagy has been extensively studied and is known to be involved in various diseases. Ferroptosis is a recently discovered form of regulated cell death characterized by iron overload, leading to the accumulation of lethal levels of lipid hydroperoxides. Recently, an increasing number of studies have revealed a link between autophagy and ferroptosis. Myocardial ischemia/reperfusion injury (MIRI) is an urgent dilemma after myocardial infarction recanalization, which is regulated by several cell death pathways, including autophagy and ferroptosis. However, the potential relationship between autophagy and ferroptosis in MIRI remains unexplored. In this study, we briefly review the mechanisms of autophagy and ferroptosis, including their roles in MIRI. Moreover, we provide an overview of the potential crosstalk in MIRI. Clarifying the relationship between different cell death pathways may provide new ideas for the treatment of MIRI in the future.
Collapse
Affiliation(s)
- Yu Yang
- Cardiology Department, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Anhui Medical University, Hefei, Anhui 230032, China
| | - Xianhe Lin
- Cardiology Department, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| |
Collapse
|
14
|
Lu LQ, Li NS, Li MR, Peng JY, Tang LJ, Luo XJ, Peng J. DL-3-n-butylphthalide improves the endothelium-dependent vasodilation in high-fat diet-fed ApoE -/- mice via suppressing inflammation, endothelial necroptosis and apoptosis. Eur J Pharmacol 2023; 956:175938. [PMID: 37536623 DOI: 10.1016/j.ejphar.2023.175938] [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: 03/07/2023] [Revised: 07/03/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
Impaired endothelium-dependent vasodilation in atherosclerosis is a high-risk factor for myocardial infarction and ischemic stroke, and inflammation, necroptosis and apoptosis contribute to endothelial dysfunction in atherosclerosis. Although DL-3-n-butylphthalide (NBP) has been widely used in treating ischemic stroke, its effect on endothelium-dependent vasodilation remains unknown. This study aims to explore whether NBP is able to improve endothelium-dependent vasodilation in atherosclerosis and the underlying mechanisms. Male ApoE-/- mice were fed with a high-fat diet (HFD) for 9-16 weeks to establish a model of atherosclerosis. NBP were given to the mice after eating HFD for 6 weeks and atorvastatin served as a positive control. The endothelium-dependent vasodilation, the blood flow velocity, the atherosclerotic lesion area, the serum levels of lipids, inflammatory cytokines and necroptosis-relevant proteins (RIPK1, RIPK3 and MLKL), and the endothelial necroptosis and apoptosis within the aorta were measured. Human umbilical vein endothelial cells (HUVECs) were incubated with oxidized low-density lipoprotein (ox-LDL) for 48 h to mimic endothelial injury in atherosclerosis, lactate dehydrogenase release, the ratio of necroptosis and apoptosis and the expression of necroptosis-relevant proteins were examined. Similar to atorvastatin, NBP improves endothelium-dependent vasodilation, decreases aortic flow velocity and reduces atherosclerotic lesion area in HFD-fed ApoE-/- mice, concomitant with a reduction in serum lipids, inflammatory cytokines and necroptosis-relevant proteins, and endothelial necroptosis and apoptosis. Consistently, NBP inhibited necroptosis and apoptosis in ox-LDL-treated HUVECs. Based on these observations, we conclude that NBP exerts beneficial effects on improving the endothelium-dependent vasodilation in atherosclerosis via suppressing inflammation, endothelial necroptosis and apoptosis.
Collapse
Affiliation(s)
- Li-Qun Lu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Nian-Sheng Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Ming-Rui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Jiao-Yang Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Li-Jing Tang
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
| |
Collapse
|
15
|
Fang Z, Lee H, Liu J, Wong KA, Brown LM, Li X, Xiaoli AM, Yang F, Zhang M. Complement C3 Reduces Apoptosis via Interaction with the Intrinsic Apoptotic Pathway. Cells 2023; 12:2282. [PMID: 37759504 PMCID: PMC10528058 DOI: 10.3390/cells12182282] [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: 08/04/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Myocardial ischemia/reperfusion (I/R) elicits an acute inflammatory response involving complement factors. Recently, we reported that myocardial necrosis was decreased in complement C3-/- mice after heart I/R. The current study used the same heart model to test the effect of C3 on myocardial apoptosis and investigated if C3 regulation of apoptosis occurred in human cardiomyocytes. Comparative proteomics analyses found that cytochrome c was present in the myocardial C3 complex of WT mice following I/R. Incubation of exogenous human C3 reduced apoptosis in a cell culture system of human cardiomyocytes that did not inherently express C3. In addition, human C3 inhibited the intrinsic apoptosis pathway in a cell-free apoptosis system. Finally, human pro-C3 was found to bind with an apoptotic factor, pro-caspase 3, in a cell-free system. Thus, we present firsthand evidence showing that C3 readily reduces myocardial apoptosis via interaction with the intrinsic apoptotic pathway.
Collapse
Affiliation(s)
- Zhou Fang
- Departments of Anesthesiology, SUNY Downstate Health Science University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (Z.F.); (H.L.); (J.L.); (K.A.W.); (X.L.)
| | - Haekyung Lee
- Departments of Anesthesiology, SUNY Downstate Health Science University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (Z.F.); (H.L.); (J.L.); (K.A.W.); (X.L.)
| | - Junying Liu
- Departments of Anesthesiology, SUNY Downstate Health Science University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (Z.F.); (H.L.); (J.L.); (K.A.W.); (X.L.)
| | - Karen A. Wong
- Departments of Anesthesiology, SUNY Downstate Health Science University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (Z.F.); (H.L.); (J.L.); (K.A.W.); (X.L.)
| | - Lewis M. Brown
- Quantitative Proteomics and Metabolomics Center, Department of Biological Sciences, Columbia University, New York, NY 10027, USA;
| | - Xiang Li
- Departments of Anesthesiology, SUNY Downstate Health Science University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (Z.F.); (H.L.); (J.L.); (K.A.W.); (X.L.)
| | - Alus M. Xiaoli
- Department of Medicine/Endocrinology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (A.M.X.); (F.Y.)
| | - Fajun Yang
- Department of Medicine/Endocrinology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (A.M.X.); (F.Y.)
| | - Ming Zhang
- Departments of Anesthesiology, SUNY Downstate Health Science University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (Z.F.); (H.L.); (J.L.); (K.A.W.); (X.L.)
- Departments of Cell Biology, SUNY Downstate Health Science University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| |
Collapse
|
16
|
Wang R, Chen X, Li X, Wang K. Molecular therapy of cardiac ischemia-reperfusion injury based on mitochondria and ferroptosis. J Mol Med (Berl) 2023; 101:1059-1071. [PMID: 37505243 DOI: 10.1007/s00109-023-02346-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/05/2023] [Accepted: 06/27/2023] [Indexed: 07/29/2023]
Abstract
Excessive death of myocardial cells can lead to various cardiovascular diseases and even develop into heart failure, so developing ideal treatment plans based on pathogenesis is of great significance for cardiopathy. After the heart undergoes ischemia‒reperfusion (I/R), myocardial cells accumulate a large amount of peroxides, leading to mitochondrial dysfunction and inducing ferroptosis. Ferroptosis is a form of iron-dependent regulatory cell death (RCD) caused by imbalanced redox and iron metabolism that leads to severe cell damage through the accumulation of peroxides. The mechanism of ferroptosis is highly correlated with mitochondrial metabolism. Myocardial cells are rich in a large number of mitochondria, which serve as energy supply centers and are prone to producing reactive oxygen species (ROS), providing opportunities for oxidative stress caused by ferroptosis. Ferroptosis is related to various cardiovascular diseases, and potential treatment methods designed around ferroptosis may alter the pathological progression of cardiovascular diseases. Therefore, this review investigates the regulatory mechanisms of ferroptosis, exploring the close pathological and physiological connections between ferroptosis and mitochondrial and cardiac I/R injury. Targeting ferroptosis and mitochondria for intervention may be an effective plan for preventing and treating cardiac I/R injury.
Collapse
Affiliation(s)
- Ruiquan Wang
- Key Laboratory of Birth Regulation and Control Technologyof , National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
- Institute for Translational Medicine, College of Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Xinzhe Chen
- Institute for Translational Medicine, College of Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Xinmin Li
- Institute for Translational Medicine, College of Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China.
| | - Kun Wang
- Key Laboratory of Birth Regulation and Control Technologyof , National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China.
- Institute for Translational Medicine, College of Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China.
| |
Collapse
|
17
|
Zhang J, Qian J, Zhang W, Chen X. The pathophysiological role of receptor-interacting protein kinase 3 in cardiovascular disease. Biomed Pharmacother 2023; 165:114696. [PMID: 37329707 DOI: 10.1016/j.biopha.2023.114696] [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: 02/17/2023] [Revised: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 06/19/2023] Open
Abstract
Recent studies have found that receptor interacting protein kinase 3 (RIPK3) can mediate CaMK Ⅱ phosphorylation and oxidation, open mitochondrial permeability transition pore (mPTP), and induce myocardial necroptosis. The increased expression or phosphorylation of RIPK3 is one of the important markers of necroptosis; Inhibition of CaMK Ⅱ phosphorylation or oxidation significantly reduces RIPK3 mediated myocardial necroptosis; Studies have shown that necroptosis plays an important role in the occurrence and development of cardiovascular diseases; Using the selective inhibitor GSK '872 of RIPK3 can effectively inhibit the occurrence and development of cardiovascular diseases, and can reverse cardiovascular and cardiac dysfunction caused by overexpression of RIPK3. In this review, we provide a brief overview of the current knowledge on RIPK3 in mediating necroptosis, inflammatory response, and oxidative stress, and discussed the role of RIPK3 in cardiovascular diseases such as atherosclerosis, myocardial ischaemia, myocardial infarction, and heart failure.
Collapse
Affiliation(s)
- Jingjing Zhang
- School of Medicine, Nantong University, Nantong, Jiangsu 226001, China
| | - Jianan Qian
- School of Pharmacy, Nantong University, Nantong, Jiangsu 226001, China
| | - Wei Zhang
- School of Medicine, Nantong University, Nantong, Jiangsu 226001, China; School of Pharmacy, Nantong University, Nantong, Jiangsu 226001, China.
| | - Xianfen Chen
- Department of Pharmacy, Nantong First People's Hospital, the Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China.
| |
Collapse
|
18
|
Wang Y, Chen H, Fan X, Xu C, Li M, Sun H, Song J, Jia F, Wei W, Jiang F, Li G, Zhong D. Bone marrow mesenchymal stem cell-derived exosomal miR-193b-5p reduces pyroptosis after ischemic stroke by targeting AIM2. J Stroke Cerebrovasc Dis 2023; 32:107235. [PMID: 37393689 DOI: 10.1016/j.jstrokecerebrovasdis.2023.107235] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 06/17/2023] [Accepted: 06/26/2023] [Indexed: 07/04/2023] Open
Abstract
BACKGROUND Ischemic stroke represents a major factor causing global morbidity and death. Bone marrow mesenchymal stem cell (BMSC)-derived exosomes (Exos) have important effects on treating ischemic stroke. Here, we investigated the therapeutic mechanism by which BMSC-derived exosomal miR-193b-5p affects ischemic stroke. METHODS luciferase assay was performed to evaluate the regulatory relationship of miR-193b-5p with absent in melanoma 2 (AIM2). Additionally, an oxygen-glucose deprivation/reperfusion (OGD/R) model was constructed for the in vitro assay, while a middle cerebral artery occlusion (MCAO) model was developed for the in vivo assay. After exosome therapy, lactate dehydrogenase and MTT assays were conducted to detect cytotoxicity and cell viability, while PCR, ELISA, western blotting assay, and immunofluorescence staining were performed to detect changes in the levels of pyroptosis-related molecules. TTC staining and TUNEL assays were performed to assess cerebral ischemia/reperfusion (I/R) injury. RESULTS In the luciferase assay, miR-193b-5p showed direct binding to the 3'-untranslated region of AIM2. In both in vivo and in vitro assays, the injected exosomes could access the sites of ischemic injury and could be internalized. In the in vitro assay, compared to normal BMSC-Exos, miR-193b-5p-overexpressing BMSC-Exos showed greater effects on increasing cell viability and attenuating cytotoxicity; AIM2, GSDMD-N, and cleaved caspase-1 levels; and IL-1β/IL-18 generation. In the in vivo assay, compared to normal BMSC-Exos, miR-193b-5p-overexpressing BMSC-Exos showed greater effects on decreasing the levels of these pyroptosis-related molecules and infarct volume. CONCLUSION BMSC-Exos attenuate the cerebral I/R injury in vivo and in vitro by inhibiting AIM2 pathway-mediated pyroptosis through miR-193b-5p delivery.
Collapse
Affiliation(s)
- Yingju Wang
- Department of Neurology, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin 150001, Heilongjiang Province, PR China
| | - Hongping Chen
- Department of Neurology, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin 150001, Heilongjiang Province, PR China
| | - Xuehui Fan
- Department of Neurology, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin 150001, Heilongjiang Province, PR China
| | - Chen Xu
- Department of Neurology, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin 150001, Heilongjiang Province, PR China
| | - Meng Li
- Department of Neurology, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin 150001, Heilongjiang Province, PR China
| | - Hongxue Sun
- Department of Neurology, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin 150001, Heilongjiang Province, PR China
| | - Jihe Song
- Department of Neurology, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin 150001, Heilongjiang Province, PR China
| | - Feihong Jia
- Department of Neurology, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin 150001, Heilongjiang Province, PR China
| | - Wan Wei
- Department of Neurology, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin 150001, Heilongjiang Province, PR China
| | - Fangchao Jiang
- Department of Neurology, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin 150001, Heilongjiang Province, PR China
| | - Guozhong Li
- Department of Neurology, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin 150001, Heilongjiang Province, PR China; Department of Neurology, Heilongjiang Provincial Hospital, 405 Guogeli Street, Harbin 150036, Heilongjiang Province, PR China.
| | - Di Zhong
- Department of Neurology, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin 150001, Heilongjiang Province, PR China.
| |
Collapse
|
19
|
Li F, Hu Z, Huang Y, Zhan H. Dexmedetomidine ameliorates diabetic cardiomyopathy by inhibiting ferroptosis through the Nrf2/GPX4 pathway. J Cardiothorac Surg 2023; 18:223. [PMID: 37430319 DOI: 10.1186/s13019-023-02300-7] [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: 03/30/2023] [Accepted: 06/05/2023] [Indexed: 07/12/2023] Open
Abstract
OBJECTIVE Dexmedetomidine (DEX) has been shown to have anti-apoptotic effects in diabetes mellitus, but its role in mitigating diabetic cardiomyopathy (DCM) through ferroptosis regulation is unclear. METHODS An in vitro DCM model was established using H9C2 cells induced with high glucose (HG) and treated with DEX at varying doses and a nuclear factor erythroid 2-realated factor 2 (Nrf2) specific inhibitor ML385. Cell viability was evaluated using the MTT method after treatment with DEX or mannitol (MAN), and the dosage of DEX used in subsequent experimentation was determined. The effects of HG-induced high osmotic pressure were assessed using MAN as a control. Cell apoptosis was evaluated using flow cytometry. Protein levels of Bcl2, Bax, nuclear Nrf2, and glutathione peroxidase 4 (GPX4) were measured using Western blot. Superoxide dismutase (SOD) activity, malondialdehyde (MDA) levels, Fe2+ concentration and reactive oxygen species (ROS) levels were measured using corresponding kits and dichlorodihydrofluorescein diacetate, respectively. RESULTS Treatment with DEX or MAN had no effect on H9C2 cell viability. HG induction reduced H9C2 cell viability, increased cell apoptosis, upregulated levels of Bax, Fe2+, MDA, and ROS, and downregulated Bcl2 protein levels, SOD activity, and protein levels of nuclear Nrf2 and GPX4. DEX inhibited HG-induced H9C2 cell apoptosis, promoted Nrf2 nuclear translocation, and activated the Nrf2/GPX4 pathway. Inhibition of Nrf2 partially reversed the protective effects of DEX against HG-evoked H9C2 cell injury. CONCLUSION Our findings demonstrate that DEX attenuates HG-induced cardiomyocyte injury by inhibiting ferroptosis through the Nrf2/GPX4 pathway, providing potential therapeutic targets for DCM treatment.
Collapse
Affiliation(s)
- Fan Li
- Department of Anesthesiology, First Afiliated Hospital of Xinjiang Medical University, Xinjiang Perioperative Organ Protection Laboratory (XJDX1411), No.1 Liyushan Road, Urumqi, Xinjiang, 830054, China
| | - Zhenfei Hu
- Department of Anesthesiology, First Afiliated Hospital of Xinjiang Medical University, Xinjiang Perioperative Organ Protection Laboratory (XJDX1411), No.1 Liyushan Road, Urumqi, Xinjiang, 830054, China
| | - Yidan Huang
- Department of Anesthesiology, First Afiliated Hospital of Xinjiang Medical University, Xinjiang Perioperative Organ Protection Laboratory (XJDX1411), No.1 Liyushan Road, Urumqi, Xinjiang, 830054, China
| | - Haiting Zhan
- Department of Anesthesiology, First Afiliated Hospital of Xinjiang Medical University, Xinjiang Perioperative Organ Protection Laboratory (XJDX1411), No.1 Liyushan Road, Urumqi, Xinjiang, 830054, China.
| |
Collapse
|
20
|
Wang Y, Guo L, Zhang Z, Fu S, Huang P, Wang A, Liu M, Ma X. A bibliometric analysis of myocardial ischemia/reperfusion injury from 2000 to 2023. Front Cardiovasc Med 2023; 10:1180792. [PMID: 37383699 PMCID: PMC10293770 DOI: 10.3389/fcvm.2023.1180792] [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: 03/06/2023] [Accepted: 05/22/2023] [Indexed: 06/30/2023] Open
Abstract
Background Myocardial ischemia/reperfusion injury (MIRI) refers to the more severe damage that occurs in the previously ischemic myocardium after a short-term interruption of myocardial blood supply followed by restoration of blood flow within a certain period of time. MIRI has become a major challenge affecting the therapeutic efficacy of cardiovascular surgery. Methods A scientific literature search on MIRI-related papers published from 2000 to 2023 in the Web of Science Core Collection database was conducted. VOSviewer was used for bibliometric analysis to understand the scientific development and research hotspots in this field. Results A total of 5,595 papers from 81 countries/regions, 3,840 research institutions, and 26,202 authors were included. China published the most papers, but the United States had the most significant influence. Harvard University was the leading research institution, and influential authors included Lefer David J., Hausenloy Derek J., Yellon Derek M., and others. All keywords can be divided into four different directions: risk factors, poor prognosis, mechanisms and cardioprotection. Conclusion Research on MIRI is flourishing. It is necessary to conduct an in-depth investigation of the interaction between different mechanisms and multi-target therapy will be the focus and hotspot of MIRI research in the future.
Collapse
Affiliation(s)
- Yifei Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Lijun Guo
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Zhibo Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Shuangqing Fu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Pingping Huang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Anzhu Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mi Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Xiaochang Ma
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| |
Collapse
|
21
|
Tian K, Yang Y, Zhou K, Deng N, Tian Z, Wu Z, Liu X, Zhang F, Jiang Z. The role of ROS-induced pyroptosis in CVD. Front Cardiovasc Med 2023; 10:1116509. [PMID: 36873396 PMCID: PMC9978107 DOI: 10.3389/fcvm.2023.1116509] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Cardiovascular disease (CVD) is the number one cause of death in the world and seriously threatens human health. Pyroptosis is a new type of cell death discovered in recent years. Several studies have revealed that ROS-induced pyroptosis plays a key role in CVD. However, the signaling pathway ROS-induced pyroptosis has yet to be fully understood. This article reviews the specific mechanism of ROS-mediated pyroptosis in vascular endothelial cells, macrophages, and cardiomyocytes. Current evidence shows that ROS-mediated pyroptosis is a new target for the prevention and treatment of cardiovascular diseases such as atherosclerosis (AS), myocardial ischemia-reperfusion injury (MIRI), and heart failure (HF).
Collapse
Affiliation(s)
- Kaijiang Tian
- The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China
| | - Yu Yang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China
| | - Kun Zhou
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China
| | - Nianhua Deng
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China
| | - Zhen Tian
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China
| | - Zefan Wu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China
| | - Xiyan Liu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China
| | - Fan Zhang
- The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Zhisheng Jiang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China
| |
Collapse
|
22
|
Zhang YY, Tian J, Peng ZM, Liu B, Peng YW, Zhang XJ, Hu ZY, Luo XJ, Peng J. Caspofungin Suppresses Brain Cell Necroptosis in Ischemic Stroke Rats via Up-Regulation of Pellino3. Cardiovasc Drugs Ther 2023; 37:9-23. [PMID: 34495409 DOI: 10.1007/s10557-021-07231-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/14/2021] [Indexed: 01/14/2023]
Abstract
PURPOSE Pellino3, an ubiquitin E3 ligase, prevents the formation of the death-induced signaling complex in response to TNF-α by targeting receptor-interacting protein kinase 1 (RIPK1), and bioinformatics analysis predicted an interaction between Pellino3 and caspofungin, a common antifungal drug used in clinics. This study aimed to explore the effect of caspofungin on brain injury in ischemic stroke and the underlying mechanisms. METHODS Ischemic stroke injury was induced in Sprague Dawley rats by occlusion of the middle cerebral artery (MCA) for 2 h, followed by 24 h reperfusion. PC12 cells were deprived of both oxygen and glucose for 8 h and then were cultured for 24 h with oxygen and glucose to mimic an ischemic stroke in vitro. RESULTS Animal experiments showed brain injury (increase in neurological deficit score and infarct volume) concomitant with a downregulation of Pellino3, a decreased ubiquitination of RIPK1, and an up-regulation of necroptosis-associated proteins [RIPK1, RIPK3, mixed lineage kinase domain-like protein (MLKL), p-RIPK1, p-RIPK3, and p-MLKL]. Administration of caspofungin (6 mg/kg, i.m.) at 1 h and 6 h after ischemia significantly improved neurological function, reduced infarct volume, up-regulated Pellino3 levels, increased RIPK1 ubiquitination, and down-regulated protein levels of RIPK1, p-RIPK1, p-RIPK3, and p-MLKL. PC12 cells deprived of oxygen/glucose developed signs of cellular injury (LDH release and necroptosis) concomitant with downregulation of Pellino3, decreased ubiquitination of RIPK1, and elevated necroptosis-associated proteins. These changes were reversed by overexpression of Pellino3. CONCLUSION We conclude that Pellino3 has an important role in counteracting necroptosis via ubiquitination of RIPK1 and caspofungin can suppress the brain cell necroptosis in ischemic stroke through upregulation of Pellino3.
Collapse
Affiliation(s)
- Yi-Yue Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Jing Tian
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Zi-Mei Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Bin Liu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Ya-Wei Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Xiao-Jie Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Zhong-Yang Hu
- Department of Neurology, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
| |
Collapse
|
23
|
Metformin confers longitudinal cardiac protection by preserving mitochondrial homeostasis following myocardial ischemia/reperfusion injury. Eur J Nucl Med Mol Imaging 2023; 50:825-838. [PMID: 36322187 DOI: 10.1007/s00259-022-06008-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022]
Abstract
PURPOSE Myocardial ischemia-reperfusion (I/R) injury is associated with systemic oxidative stress, cardiac mitochondrial homeostasis, and cardiomyocyte apoptosis. Metformin has been recognized to attenuate cardiomyocyte apoptosis. However, the longitudinal effects and pathomechanism of metformin on the regulation of myocardial mitohormesis following I/R treatment remain unclear. This study aimed to investigate the longitudinal effects and mechanism of metformin in regulating cardiac mitochondrial homeostasis by serial imaging with the 18-kDa translocator protein (TSPO)-targeted positron emission tomography (PET) tracer 18F-FDPA. METHODS Myocardial I/R injury was established in Sprague-Dawley rats, which were treated with or without metformin (150 mg/kg per day). Serial gated 18F-FDG and 18F-FDPA PET imaging were performed at 1, 4, and 8 weeks after surgery, followed by analysis of ventricular remodelling and cardiac mitochondrial homeostasis. The correlation between Hsp60 and 18F-FDPA uptake was analyzed. After PET imaging, the activity of antioxidant enzymes, immunostaining, and western blot analysis were performed to analyze the spatio-temporal effects and pathomechanism of metformin for cardiac protection after myocardial I/R injury. RESULTS Oxidative stress and apoptosis increased 1 week after myocardial I/R injury (before significant progression of ventricular remodelling). TSPO expression was correlated with Hsp60 expression and was co-localized with inflammatory CD68+ macrophages in the infarct area, and TSPO uptake was associated with an upregulation of AMPK-p/AMPK and a downregulation of Bcl-2/Bax. However, these effects were reversed with metformin treatment. Eight weeks after myocardial I/R injury (representing the advanced stage of heart failure), 18F-FDPA uptake in myocardial cells in the distal non-infarct area increased without CD68+ expression, whereas the activity decreased with metformin treatment. CONCLUSION Taken together, these results show that a prolonged metformin treatment has pleiotropic protective effects against myocardial I/R injury associated with a regional and temporal dynamic balance between mitochondrial homeostasis and cardiac outcome, which were assessed by TSPO-targeted imaging during cardiac remodelling.
Collapse
|
24
|
Peng ZM, Zhang YY, Wei D, Zhang XJ, Liu B, Peng J, Luo XJ. MALT1 promotes necroptosis in stroke rat brain via targeting the A20/RIPK3 pathway. Arch Biochem Biophys 2023; 735:109502. [PMID: 36603698 DOI: 10.1016/j.abb.2023.109502] [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: 09/20/2022] [Revised: 12/30/2022] [Accepted: 01/01/2023] [Indexed: 01/04/2023]
Abstract
Necroptosis has been demonstrated to contribute to brain injury in ischemic stroke, whereas A20 can exert anti-necroptosis effect via deubiquitinating receptor-interacting protein kinase (RIPK3) at k63 and it can be cleaved by MALT1. This study aims to explore whether MALT1 is upregulated in the brain during ischemic stroke and promotes brain cell necroptosis through enhancing the degradation of A20. Ischemic stroke model was established in Sprague Dawley rats by occlusion of the middle cerebral artery (MCA) for 2 h, followed by 24 h reperfusion, which showed brain injury (increase in neurological deficit score and infarct volume) concomitant with an upregulation of MALT1, a decrease in A20 level, and increases in necroptosis-associated protein levels [RIPK3, mixed lineage kinase domain-like protein (MLKL) and p-MLKL] and k63-ubiquitination of RIPK3 in brain tissues. Administration of MALT1 inhibitor (Ml-2) at 8 or 15 mg/kg (i.p.) at 1 h after ischemia significantly improved neurological function and reduced infarct volume together with a downregulation of MALT1, an increase in A20 level and decreases in necroptosis-associated protein levels and k63-ubiquitination of RIPK3. Similarly, knockdown of MALT1 could also reduce oxygen-glucose deprivation/reoxygenation (OGD/R)-induced injury in the cultured HT22 cells coincident with an increase in A20 level and decreases in necroptosis-associated protein levels and k63-ubiquitination of RIPK3. Based on these observations, we conclude that MALT1 promotes necroptosis in stroke rat brain via enhancing the degradation of A20, which leads to a decrease in the capability of A20 to deubiquitinate RIPK3 at k63 and a subsequent compromise in counteraction against the brain cell necroptosis.
Collapse
Affiliation(s)
- Zi-Mei Peng
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, China; Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Yi-Yue Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Dan Wei
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Xiao-Jie Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China; Department of Pharmacy, Xiangya Hospital, Central South University, Hunan, China
| | - Bin Liu
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China; Department of Pharmacy, Xiangya Hospital, Central South University, Hunan, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
| |
Collapse
|
25
|
Ma S, He L, Zuo Q, Zhang G, Guo Y. Canagliflozin Regulates Ferroptosis, Potentially via Activating AMPK/PGC-1α/Nrf2 Signaling in HFpEF Rats. CARDIOVASCULAR INNOVATIONS AND APPLICATIONS 2023. [DOI: 10.15212/cvia.2022.0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Aims: Sodium-glucose cotransporter-2 (SGLT2) inhibitors have been found to ameliorate major adverse cardiovascular events in patients with heart failure with preserved ejection fraction (HFpEF), but the exact mechanism is unknown. Ferroptosis is a form of programmed necrosis. Herein, we verified that canagliflozin (CANA) ameliorates heart function in HFpEF rats, partly by regulating ferroptosis, which may be activated by AMPK/PGC-1α/Nrf2 signaling.
Methods: An HFpEF model was established and subjected to CANA treatment. Blood pressure was monitored, and echocardiography was performed at the 12th week. Pathological examination was performed, and expression of ferroptosis-associated proteins and AMPK/PGC-1α/Nrf2 signaling related proteins was detected.
Results: CANA had an antihypertensive effect and increased E/A ratios in HFpEF rats. Myocardial pathology was ameliorated, on the basis of decreased cross-sectional area and intercellular fibrosis. Acyl-CoA synthetase long-chain family member 4 (ACSL4) expression increased, whereas ferritin heavy chain 1 (FTH1) expression decreased in HFpEF rats, which showed iron overload. CANA reversed changes in ACSL4 and FTH1, and decreased iron accumulation, but did not alter glutathione peroxidase 4 (GPX4) expression. The expression of AMPK/PGC-1α/Nrf2 signaling related proteins and heme oxygenase 1 (HO-1) in the HFpEF group decreased but was reverted after CANA treatment.
Conclusions: CANA regulates ferroptosis, potentially via activating AMPK/PGC-1α/Nrf2 signaling in HFpEF rats.
Collapse
Affiliation(s)
- Sai Ma
- Department of Pain, Hebei General Hospital, Shijiazhuang 050051, Hebei, China
| | - Lili He
- Department of Geriatric Cardiology, Hebei General Hospital, Shijiazhuang 050051, Hebei, China
| | - Qingjuan Zuo
- Department of Geriatric Cardiology, Hebei General Hospital, Shijiazhuang 050051, Hebei, China
| | - Guorui Zhang
- Department of Cardiology, The Third Hospital of Shijiazhuang City affiliated with Hebei Medical University, Shijiazhuang 050011, Hebei, China
| | - Yifang Guo
- Department of Geriatric Cardiology, Hebei General Hospital, Shijiazhuang 050051, Hebei, China
| |
Collapse
|
26
|
Du H, He Y, Zhu J, Zhou H, Shao C, Yang J, Wan H. Danhong injection alleviates cerebral ischemia-reperfusion injury by inhibiting mitochondria-dependent apoptosis pathway and improving mitochondrial function in hyperlipidemia rats. Biomed Pharmacother 2023; 157:114075. [PMID: 36481401 DOI: 10.1016/j.biopha.2022.114075] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/25/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Cerebral ischemia threatens human health and life. Hyperlipidemia is a risk of cerebral ischemia. Danhong injection (DHI) is a traditional Chinese medical preparation for the treatment of cerebrovascular diseases. However, the effects of DHI on mitochondria-dependent apoptosis and mitochondrial function following cerebral ischemia in hyperlipidemia rats are not clear. In this study, SD rats were fed by high-fat diet for six weeks to establish the hyperlipidemia model, except for the sham and ischemia-reperfusion (I/R) groups. Hyperlipidemia rats were assigned into I/R + high-fat diet (HFD) group, DHI 1 mL/kg group, and DHI 2 mL/kg group. DHI was administrated to the drug group via caudal vein for seven consecutive days (once per day). Subsequently, rats underwent middle cerebral artery occlusion (MCAO) for 1 h and reperfusion for 24 h. The results showed that DHI significantly reduced cerebral infarction volume, ameliorated neurological function, improved pathological changes, and inhibited apoptosis. DHI could significantly restore the levels of mitochondrial respiratory chain complexes I-IV, increase the ATP content and COX activity, and decrease the level of OFR in the ischemic brain mitochondria of hyperlipidemia rats after I/R. DHI significantly regulated the levels of cytochrome c (Cyt c), Apaf1, Bax, Bcl-2, Caspase-3, and Caspase-9 in brain tissue, and improved mitochondrial dynamics (Mfn1, Mfn2, OPA1, Drp1, and Fis1). The results indicate that DHI could alleviate ischemic brain injury in hyperlipidemia rats, and the mechanism may be to improve mitochondrial function by restoring the mitochondrial respiratory chain and changing the protein balance of mitochondrial fusion and fission, and inhibiting mitochondria-dependent apoptosis.
Collapse
Affiliation(s)
- Haixia Du
- Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Yu He
- Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Jiaqi Zhu
- Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Huifen Zhou
- Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Chongyu Shao
- Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Jiehong Yang
- Zhejiang Chinese Medical University, Hangzhou, PR China.
| | - Haitong Wan
- Zhejiang Chinese Medical University, Hangzhou, PR China.
| |
Collapse
|
27
|
Chen C, Chen W, Zhou X, Li Y, Pan X, Chen X. Hyperbaric oxygen protects HT22 cells and PC12 cells from damage caused by oxygen-glucose deprivation/reperfusion via the inhibition of Nrf2/System Xc-/GPX4 axis-mediated ferroptosis. PLoS One 2022; 17:e0276083. [PMID: 36355759 PMCID: PMC9648730 DOI: 10.1371/journal.pone.0276083] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 09/28/2022] [Indexed: 11/12/2022] Open
Abstract
This study was to investigate the protective effect of hyperbaric oxygen (HBO) on HT22 and PC12 cell damage caused by oxygen-glucose deprivation/reperfusion-induced ferroptosis. A 2-h oxygen-glucose deprivation and 24-h reperfusion model on HT22 and PC12 cells was used to simulate cerebral ischemia-reperfusion injury. Cell viabilities were detected by Cell Counting Kit-8 (CCK-8) method. The levels of reactive oxygen species (ROS) and lipid reactive oxygen species (Lipid ROS) were detected by fluorescent probes Dihydroethidium (DHE) and C11 BODIPY 581/591. Iron Colorimetric Assay Kit, malondialdehyde (MDA) and glutathione (GSH) activity assay kits were used to detect intracellular iron ion, MDA and GSHcontent. Cell ferroptosis-related ultrastructures were visualized using transmission electron microscopy (TEM). Furthermore, PCR and Western blot analyses were used to detect the expressions of ferroptosis-related genes and proteins. After receiving oxygen-glucose deprivation/reperfusion, the viabilities of HT22 and PC12 cells were significantly decreased; ROS, Lipid ROS, iron ions and MDA accumulation occurred in the cells; GSH contents decreased; TEM showed that cells were ruptured and blebbed, mitochondria atrophied and became smaller, mitochondrial ridges were reduced or even disappeared, and apoptotic bodies appeared. And the expressions of Nrf2, SLC7A11 and GPX4 genes were reduced; the expressions of p-Nrf2/Nrf2, xCT and GPX4 proteins were reduced. Notably, these parameters were significantly reversed by HBO, indicating that HBO can protect HT22 cells and PC12 cells from damage caused by oxygen-glucosedeprivation/reperfusion via the inhibition of Nrf2/System Xc-/GPX4 axis-mediated ferroptosis.
Collapse
Affiliation(s)
- Chunxia Chen
- Department of Pharmacy, The People’s Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, Guangxi, P. R. China
| | - Wan Chen
- Department of Emergency, The People’s Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, Guangxi, P. R. China
| | - Xing Zhou
- Department of Pharmacy, The People’s Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, Guangxi, P. R. China
| | - Yaoxuan Li
- Department of Neurology, The People’s Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, China
| | - Xiaorong Pan
- Department of Hyperbaric Oxygen, The People’s Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, Guangxi, P. R. China
| | - Xiaoyu Chen
- Department of Pharmacy, The People’s Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, Guangxi, P. R. China
| |
Collapse
|
28
|
Polymyxin B Reduces Brain Injury in Ischemic Stroke Rat Through a Mechanism Involving Targeting ESCRT-III Machinery and RIPK1/RIPK3/MLKL Pathway. J Cardiovasc Transl Res 2022; 15:1129-1142. [PMID: 35239171 DOI: 10.1007/s12265-022-10224-1] [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: 09/13/2021] [Accepted: 02/21/2022] [Indexed: 02/05/2023]
Abstract
Endosomal sorting complex required for transport III (ESCRT-III) machinery is a key component to counteract the mixed lineage kinase domain-like pseudokinase (MLKL)-induced plasma membrane broken in cells undergoing necroptosis. Based on the bioinformatics analysis, polymyxin B, a polypeptide antibiotic, is predicted to simultaneously interact with ESCRT-III subunits and necroptosis-relevant proteins. This study aims to explore whether polymyxin B could reduce necroptosis in the stroke rat brain via enhancing the ESCRT-III machinery and/or suppressing the RIPK1/RIPK3/MLKL pathway. The stroke rats showed evident brain injury, concomitant with the downregulation of ESCRT-III subunits and the upregulation of necroptosis-relevant proteins. Post-ischemic administration of polymyxin B could alleviate the brain injury, accompanied by restoration of the levels of ESCRT-III subunits and suppression of necroptosis-relevant proteins. And, polymyxin B exerted similar effects in hypoxia-treated HT22 cells. We conclude that polymyxin B can reduce necroptosis in the stroke rat brain via enhancing the ESCRT-III machinery and suppressing the RIPK1/RIPK3/MLKL pathway simultaneously.
Collapse
|
29
|
Emerging roles of ferroptosis in cardiovascular diseases. Cell Death Dis 2022; 8:394. [PMID: 36127318 PMCID: PMC9488879 DOI: 10.1038/s41420-022-01183-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/29/2022] [Accepted: 09/06/2022] [Indexed: 11/26/2022]
Abstract
The mechanism of cardiovascular diseases (CVDs) is complex and threatens human health. Cardiomyocyte death is an important participant in the pathophysiological basis of CVDs. Ferroptosis is a new type of iron-dependent programmed cell death caused by excessive accumulation of iron-dependent lipid peroxides and reactive oxygen species (ROS) and abnormal iron metabolism. Ferroptosis differs from other known cell death pathways, such as apoptosis, necrosis, necroptosis, autophagy and pyroptosis. Several compounds have been shown to induce or inhibit ferroptosis by regulating related key factors or signalling pathways. Recent studies have confirmed that ferroptosis is associated with the development of diverse CVDs and may be a potential therapeutic drug target for CVDs. In this review, we summarize the characteristics and related mechanisms of ferroptosis and focus on its role in CVDs, with the goal of inspiring novel treatment strategies.
Collapse
|
30
|
Li Y, Gao Y, Li G. Preclinical multi-target strategies for myocardial ischemia-reperfusion injury. Front Cardiovasc Med 2022; 9:967115. [PMID: 36072870 PMCID: PMC9444048 DOI: 10.3389/fcvm.2022.967115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
Despite promising breakthroughs in diagnosing and treating acute coronary syndromes, cardiovascular disease’s high global mortality rate remains indisputable. Nearly half of these patients died of ischemic heart disease. Primary percutaneous coronary intervention (PCI) and coronary artery bypass grafting can rapidly restore interrupted blood flow and become the most effective method for salvaging viable myocardium. However, restoring blood flow could increase the risk of other complications and myocardial cell death attributed to myocardial ischemia-reperfusion injury (IRI). How to reduce the damage of blood reperfusion to ischemic myocardium has become an urgent problem to be solved. In preclinical experiments, many treatments have substantial cardioprotective effects against myocardial IRI. However, the transition from these cardioprotective therapies to clinically beneficial therapies for patients with acute myocardial infarction remains elusive. The reasons for the failure of the clinical translation may be multi-faceted, and three points are summarized here: (1) Our understanding of the complex pathophysiological mechanisms of myocardial IRI is far from enough, and the classification of specific therapeutic targets is not rigorous, and not clear enough; (2) Most of the clinical patients have comorbidities, and single cardioprotective strategies including ischemia regulation strategies cannot exert their due cardioprotective effects under conditions of hyperglycemia, hypertension, hyperlipidemia, and aging; (3) Most preclinical experimental results are based on adult, healthy animal models. However, most clinical patients had comorbidities and received multiple drug treatments before reperfusion therapy. In 2019, COST Action proposed a multi-target drug combination initiative for prospective myocardial IRI; the optimal cardioprotective strategy may be a combination of additive or synergistic multi-target therapy, which we support. By establishing more reasonable preclinical models, screening multi-target drug combinations more in line with clinical practice will benefit the translation of clinical treatment strategies.
Collapse
|
31
|
Cell Death Mechanisms in Cerebral Ischemia-Reperfusion Injury. Neurochem Res 2022; 47:3525-3542. [PMID: 35976487 DOI: 10.1007/s11064-022-03697-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/11/2022] [Accepted: 07/14/2022] [Indexed: 10/15/2022]
Abstract
Ischemic stroke is one of the major causes of morbidity and mortality, affecting millions of people worldwide. Inevitably, the interruption of cerebral blood supply after ischemia may promote a cascade of pathophysiological processes. Moreover, the subsequent restoration of blood flow and reoxygenation may further aggravate brain tissue injury. Although recombinant tissue plasminogen activator (rt-PA) is the only approved therapy for restoring blood perfusion, the reperfusion injury and the narrow therapeutic time window restrict its application for most stroke patients. Increasing evidence indicates that multiple cell death mechanisms are relevant to cerebral ischemia-reperfusion injury, including apoptosis, necrosis, necroptosis, autophagy, pyroptosis, ferroptosis, and so on. Therefore, it is crucial to comprehend various cell death mechanisms and their interactions. In this review, we summarize the various signaling pathways underlying cerebral ischemia-reperfusion injury and elaborate on the crosstalk between the different mechanisms.
Collapse
|
32
|
Han R, Huang H, Xia W, Liu J, Luo H, Tang J, Xia Z. Perspectives for Forkhead box transcription factors in diabetic cardiomyopathy: Their therapeutic potential and possible effects of salvianolic acids. Front Cardiovasc Med 2022; 9:951597. [PMID: 36035917 PMCID: PMC9403618 DOI: 10.3389/fcvm.2022.951597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/27/2022] [Indexed: 11/15/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is the primary cause of morbidity and mortality in diabetic cardiovascular complications, which initially manifests as cardiac hypertrophy, myocardial fibrosis, dysfunctional remodeling, and diastolic dysfunction, followed by systolic dysfunction, and eventually end with acute heart failure. Molecular mechanisms underlying these pathological changes in diabetic hearts are complicated and multifactorial, including but not limited to insulin resistance, oxidative stress, lipotoxicity, cardiomyocytes apoptosis or autophagy, inflammatory response, and myocardial metabolic dysfunction. With the development of molecular biology technology, accumulating evidence illustrates that members of the class O of Forkhead box (FoxO) transcription factors are vital for maintaining cardiomyocyte metabolism and cell survival, and the functions of the FoxO family proteins can be modulated by a wide variety of post-translational modifications including phosphorylation, acetylation, ubiquitination, arginine methylation, and O-glycosylation. In this review, we highlight and summarize the most recent advances in two members of the FoxO family (predominately FoxO1 and FoxO3a) that are abundantly expressed in cardiac tissue and whose levels of gene and protein expressions change as DCM progresses, with the goal of providing valuable insights into the pathogenesis of diabetic cardiovascular complications and discussing their therapeutic potential and possible effects of salvianolic acids, a natural product.
Collapse
Affiliation(s)
- Ronghui Han
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hemeng Huang
- Department of Emergency, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Weiyi Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Orthopaedics and Traumatology, The Univerisity of Hong Kong, Hong Kong, China
- *Correspondence: Weiyi Xia,
| | - Jingjin Liu
- Department of Cardiology, Shenzhen People’s Hospital and The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Hui Luo
- Marine Biomedical Research Institution, Guangdong Medical University, Zhanjiang, China
| | - Jing Tang
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Hong Kong, China
- Zhengyuan Xia,
| |
Collapse
|
33
|
Zhang X, Ren Z, Xu W, Jiang Z. Necroptosis in atherosclerosis. Clin Chim Acta 2022; 534:22-28. [PMID: 35809652 DOI: 10.1016/j.cca.2022.07.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 11/26/2022]
Abstract
Atherosclerosis, a chronic inflammatory disease, is a leading cause of death worldwide. Vascular endothelial cells (VECs), vascular smooth muscle cells (VSMCs) and macrophages play extremely vital roles in the formation of atherosclerotic plaques and subsequent atherosclerosis. Necroptosis, a caspase-independent programmed cell necrosis, occurs in advanced atherosclerotic plaques and has been implicated in VEC, VSMC and macrophage function. Although necroptosis may have considered as a defensive line against intracellular infection, it can induce a pro-inflammatory state, which will accelerate the disease process. Accordingly, necroptosis plays an important pathophysiologic role. In this review, we explore the role of necroptosis in VECs, VSMCs and macrophages in atherosclerotic plaques and their connection to atherosclerosis.
Collapse
Affiliation(s)
- Xiaofan Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Hengyang Medical School, University of South China, Hengyang 421001, China.
| | - Zhong Ren
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Hengyang Medical School, University of South China, Hengyang 421001, China.
| | - Wenxin Xu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Hengyang Medical School, University of South China, Hengyang 421001, China.
| | - Zhisheng Jiang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Hengyang Medical School, University of South China, Hengyang 421001, China.
| |
Collapse
|
34
|
Molecular Mechanisms of Parthanatos and Its Role in Diverse Diseases. Int J Mol Sci 2022; 23:ijms23137292. [PMID: 35806303 PMCID: PMC9266317 DOI: 10.3390/ijms23137292] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 12/12/2022] Open
Abstract
Differential evolution of apoptosis, programmed necrosis, and autophagy, parthanatos is a form of cell death mediated by poly(ADP-ribose) polymerase 1 (PARP1), which is caused by DNA damage. PARP1 hyper-activation stimulates apoptosis-inducing factor (AIF) nucleus translocation, and accelerates nicotinamide adenine dinucleotide (NAD+) and adenosine triphosphate (ATP) depletion, leading to DNA fragmentation. The mechanisms of parthanatos mainly include DNA damage, PARP1 hyper-activation, PAR accumulation, NAD+ and ATP depletion, and AIF nucleus translocation. Now, it is reported that parthanatos widely exists in different diseases (tumors, retinal diseases, neurological diseases, diabetes, renal diseases, cardiovascular diseases, ischemia-reperfusion injury...). Excessive or defective parthanatos contributes to pathological cell damage; therefore, parthanatos is critical in the therapy and prevention of many diseases. In this work, the hallmarks and molecular mechanisms of parthanatos and its related disorders are summarized. The questions raised by the recent findings are also presented. Further understanding of parthanatos will provide a new treatment option for associated conditions.
Collapse
|
35
|
Roles and Mechanisms of Regulated Necrosis in Corneal Diseases: Progress and Perspectives. J Ophthalmol 2022; 2022:2695212. [PMID: 35655803 PMCID: PMC9152437 DOI: 10.1155/2022/2695212] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/24/2022] [Accepted: 05/09/2022] [Indexed: 11/21/2022] Open
Abstract
Regulated necrosis is defined as cell death characterized by loss of the cell membrane integrity and release of the cytoplasmic content. It contributes to the development and progression of some diseases, including ischemic stroke injury, liver diseases, hypertension, and cancer. Various forms of regulated necrosis, particularly pyroptosis, necroptosis, and ferroptosis, have been implicated in the pathogenesis of corneal disease. Regulated necrosis of corneal cells enhances inflammatory reactions in the adjacent corneal tissues, leading to recurrence and aggravation of corneal disease. In this review, we summarize the molecular mechanisms of pyroptosis, necroptosis, and ferroptosis in corneal diseases and discuss the roles of regulated necrosis in inflammation regulation, tissue repair, and corneal disease outcomes.
Collapse
|
36
|
Xu Y, Chen R, Yan J, Zang G, Shao C, Wang Z. CD137 Signal Mediates Cardiac Ischemia-Reperfusion Injury by Regulating the Necrosis of Cardiomyocytes. J Cardiovasc Transl Res 2022; 15:1163-1175. [PMID: 35419772 DOI: 10.1007/s12265-022-10240-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/18/2022] [Indexed: 11/28/2022]
Abstract
The injury of cardiomyocytes after ischemia-reperfusion is the main reason of cardiac dysfunction. Necrosis is one of the methods of programmed cell death and cardiomyocyte necrosis occurs in the process of reperfusion. The activation of CD137 signal is involved in various diseases. In vivo experiments proved that CD137-/- mice have less heart damage than wild-type mice after ischemia-reperfusion. In vitro experiments, we found that after inhibiting the CD137 signal, the degree of necrosis of HL-1 cells was reduced and it was caused by reducing the Ca2 + overload in the mitochondria, which caused the reduction of mPTP opening. Ca2 + overload in mitochondria induced by activation of CD137 signal was caused by increased Ca2 + released into mitochondria by activation of IP3R and increased MCU level. These results indicate that CD137 signaling aggravates cardiac ischemia-reperfusion injury by inducing myocardial cell necrosis.
Collapse
Affiliation(s)
- Yao Xu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Rui Chen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Jinchuan Yan
- Health Science Center, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Guangyao Zang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Chen Shao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, China.
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, China.
| |
Collapse
|
37
|
Shi M, Chen J, Liu T, Dai W, Zhou Z, Chen L, Xie Y. Protective Effects of Remimazolam on Cerebral Ischemia/Reperfusion Injury in Rats by Inhibiting of NLRP3 Inflammasome-Dependent Pyroptosis. Drug Des Devel Ther 2022; 16:413-423. [PMID: 35210755 PMCID: PMC8863189 DOI: 10.2147/dddt.s344240] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/26/2022] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Remimazolam is a novel benzodiazepine γ-aminobutyric acid A (GABAa) receptor agonist used for sedation and the induction as well as maintenance of general anesthesia. Previous research proved that anesthetic agents acting on GABAa receptor, such as thiopentone, propofol and midazolam, have protective actions for cerebral ischemia/reperfusion (I/R) injury. We here probed into remimazolam for its protective effect and potential mechanism of action against cerebral I/R injury. MATERIAL AND METHODS A rat model of middle cerebral artery occlusion (MCAO) with focal transient cerebral I/R injury was established and was given tail vein injection of gradient remimazolam (5, 10, 20 mg/kg) after 2 h of ischemia. Following 24 h of reperfusion, neurological function, brain infarct volume, morphology of cerebral cortical neurons, and expressions of corticocerebral NLRP3, ASC, caspase-1, GSDMD, IL-1β and IL-18 were evaluated. RESULTS The results showed that remimazolam could effectively improve the neurological dysfunction, reduce the infarct volume and alleviate the damage of cortical neurons after I/R injury. Notably, the expression of NLRP3 inflammasome pathway was down-regulated, suggesting that remimazolam exerted protective actions on I/R injury by suppressing pyroptosis with decreased expression and release of inflammatory factors, and the involvement of the NLRP3 inflammasome pathway might be the core during that process. Overall, our results indicate that NLRP3 inflammation is a promising target. CONCLUSION Based on this mechanism, remimazolam may be one of the ideal anesthetic drugs for patients with ischemic stroke.
Collapse
Affiliation(s)
- Min Shi
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Jing Chen
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Tianxiao Liu
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Weixin Dai
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Zhan Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Lifei Chen
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Yubo Xie
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| |
Collapse
|
38
|
Lin L, Zhang MX, Zhang L, Zhang D, Li C, Li YL. Autophagy, Pyroptosis, and Ferroptosis: New Regulatory Mechanisms for Atherosclerosis. Front Cell Dev Biol 2022; 9:809955. [PMID: 35096837 PMCID: PMC8793783 DOI: 10.3389/fcell.2021.809955] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/27/2021] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disorder characterized by the gradual buildup of plaques within the vessel wall of middle-sized and large arteries. The occurrence and development of atherosclerosis and the rupture of plaques are related to the injury of vascular cells, including endothelial cells, smooth muscle cells, and macrophages. Autophagy is a subcellular process that plays an important role in the degradation of proteins and damaged organelles, and the autophagy disorder of vascular cells is closely related to atherosclerosis. Pyroptosis is a proinflammatory form of regulated cell death, while ferroptosis is a form of regulated nonapoptotic cell death involving overwhelming iron-dependent lipid peroxidation. Both of them exhibit distinct features from apoptosis, necrosis, and autophagy in morphology, biochemistry, and genetics. However, a growing body of evidence suggests that pyroptosis and ferroptosis interact with autophagy and participate in the development of cancers, degenerative brain diseases and cardiovascular diseases. This review updated the current understanding of autophagy, pyroptosis, and ferroptosis, finding potential links and their effects on atherogenesis and plaque stability, thus providing ways to develop new pharmacological strategies to address atherosclerosis and stabilize vulnerable, ruptured plaques.
Collapse
Affiliation(s)
- Lin Lin
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mu-Xin Zhang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lei Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dan Zhang
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chao Li
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yun-Lun Li
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, China.,Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
39
|
Huang L, Zhou Y, Gou ZX, Zhang F, Lu LQ. Docosahexaenoic acid reduces hypoglycemia-induced neuronal necroptosis via the peroxisome proliferator-activated receptor γ/nuclear factor-κB pathway. Brain Res 2022; 1774:147708. [PMID: 34785255 DOI: 10.1016/j.brainres.2021.147708] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 10/31/2021] [Accepted: 11/01/2021] [Indexed: 11/02/2022]
Abstract
DHA has been shown to be neuroprotective and important to neurogenesis, but its role in HG-induced brain injury and the underlying mechanisms remain unknown. To elucidate the therapeutic effect of DHA, we established a mouse model with insulin-induced hypoglycemic brain injury and an in vitro model of HT-22 cells using a sugar-free medium. DHA treatment significantly reduced neuronal death and improved HG-induced learning and memory deficits. Moreover, DHA inhibited neuronal necroptosis and decreased the concentrations of TNF-α, IL-1β and TNFR1. DHA also activated PPAR-γ and suppressed the NF-κB pathway in mouse brain tissues. In vitro, DHA treatment restored the viability and decreased necroptosis of HT-22 cells treated with glucose deprivation. However, the inhibition of PPAR-γ with T0070907 reversed neuroprotective and anti-necroptosis effects of DHA in HG-induced brain injury, which is associated with the activation of the downstream NF-κB pathway. We conclude that DHA displays a protective effect against HG-induced brain injury through the PPAR-γ/NF-κB pathway and represents a promising method to prevent HG-induced brain injury.
Collapse
Affiliation(s)
- Lin Huang
- Department of Neonatology, Sichuan Provincial Maternity and Child Health Care Hospital, No. 290 West Second Street, Shayan Road, 610031, Sichuan, China; Clinical Medical College and The First Affiliated Hospital of ChengDu Medical College Chengdu 610000, Sichuan, China
| | - Yue Zhou
- Clinical Medical College and The First Affiliated Hospital of ChengDu Medical College Chengdu 610000, Sichuan, China
| | - Zhi-Xian Gou
- Clinical Medical College and The First Affiliated Hospital of ChengDu Medical College Chengdu 610000, Sichuan, China
| | - Feng Zhang
- Clinical Medical College and The First Affiliated Hospital of ChengDu Medical College Chengdu 610000, Sichuan, China
| | - Li-Qun Lu
- Clinical Medical College and The First Affiliated Hospital of ChengDu Medical College Chengdu 610000, Sichuan, China.
| |
Collapse
|
40
|
Xu J, Zhang M, Liu F, Shi L, Jiang X, Chen C, Wang J, Diao M, Khan ZU, Zhang M. Mesenchymal Stem Cells Alleviate Post-resuscitation Cardiac and Cerebral Injuries by Inhibiting Cell Pyroptosis and Ferroptosis in a Swine Model of Cardiac Arrest. Front Pharmacol 2021; 12:793829. [PMID: 34955860 PMCID: PMC8696260 DOI: 10.3389/fphar.2021.793829] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022] Open
Abstract
Following cardiopulmonary resuscitation (CPR), the ensuing cardiac and cerebral injuries contribute to the poor outcome of cardiac arrest (CA) victims, in which the pathogenetic process is possibly driven by cell pyroptosis and ferroptosis. Mesenchymal stem cells (MSCs) have been shown to be a promising strategy for post-resuscitation cardiac and cerebral protection in rat, but its effectiveness in the clinically relevant swine model and the potential protective mechanism remain unknown. The present study was designed to investigate whether MSCs administration could alleviate post-resuscitation cardiac and cerebral injuries through the inhibition of cell pyroptosis and ferroptosis in swine. Twenty-four male domestic swine were randomly divided into three groups: sham, CPR, and MSC. A dose of 2.5×106/kg of MSCs derived from human embryonic stem cells was intravenously infused at 1.5, and 3 days prior to CA. The animal model was established by 8 min of CA and then 8 min of CPR. After resuscitation, cardiac, cerebral function and injury biomarkers were regularly evaluated for a total of 24 h. At 24 h post-resuscitation, pyroptosis-related proteins (NLRP3, ASC, cleaved caspase-1, GSDMD), proinflammatory cytokines (IL-1β, IL-18), ferroptosis-related proteins (ACSL4, GPX4) and iron deposition in the heart, cortex and hippocampus were measured. Consequently, significantly greater cardiac, cerebral dysfunction and injuries after resuscitation were observed in the CPR and MSC groups compared with the sham group. However, the severity of cardiac and cerebral damage were significantly milder in the MSC group than in the CPR group. In addition, the expression levels of NLRP3, ASC, cleaved caspase-1, GSDMD and ACSL4, the contents of IL-1β and IL-18, and the level of iron deposition were significantly higher while the expression level of GPX4 was significantly lower in the heart, cortex and hippocampus in all resuscitated animals compared with the sham group. Nevertheless, MSCs administration significantly decreased post-resuscitation cardiac, cerebral pyroptosis and ferroptosis compared to the CPR group. Our results showed that the administration of MSCs significantly alleviated post-resuscitation cardiac and cerebral injuries in swine, in which the protective effects were related to the inhibition of cell pyroptosis and ferroptosis.
Collapse
Affiliation(s)
- Jiefeng Xu
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou, China
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| | - Minhai Zhang
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou, China
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| | - Fei Liu
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou, China
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| | - Lin Shi
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou, China
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| | - Xiangkang Jiang
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou, China
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| | - Chuang Chen
- Department of Emergency Medicine, Zhejiang Hospital, Hangzhou, China
| | | | - Mengyuan Diao
- Department of Intensive Care Medicine, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zafar Ullah Khan
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou, China
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| | - Mao Zhang
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou, China
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| |
Collapse
|
41
|
Luo MY, Su JH, Gong SX, Liang N, Huang WQ, Chen W, Wang AP, Tian Y. Ferroptosis: New Dawn for Overcoming the Cardio-Cerebrovascular Diseases. Front Cell Dev Biol 2021; 9:733908. [PMID: 34858973 PMCID: PMC8632439 DOI: 10.3389/fcell.2021.733908] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/25/2021] [Indexed: 12/21/2022] Open
Abstract
The dynamic balance of cardiomyocytes and neurons is essential to maintain the normal physiological functions of heart and brain. If excessive cells die in tissues, serious Cardio-Cerebrovascular Diseases would occur, namely, hypertension, myocardial infarction, and ischemic stroke. The regulation of cell death plays a role in promoting or alleviating Cardio-Cerebrovascular Diseases. Ferroptosis is an iron-dependent new type of cell death that has been proved to occur in a variety of diseases. In our review, we focus on the critical role of ferroptosis and its regulatory mechanisms involved in Cardio-Cerebrovascular Diseases, and discuss the important function of ferroptosis-related inhibitors in order to propose potential implications for the prevention and treatment of Cardio-Cerebrovascular Diseases.
Collapse
Affiliation(s)
- Meng-Yi Luo
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| | - Jian-Hui Su
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| | - Shao-Xin Gong
- Department of Pathology, First Affiliated Hospital, University of South China, Hengyang, China
| | - Na Liang
- Department of Anesthesiology, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Wen-Qian Huang
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| | - Wei Chen
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| | - Ai-Ping Wang
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| | - Ying Tian
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| |
Collapse
|
42
|
Vasconcelos AG, Barros ALAN, Cabral WF, Moreira DC, da Silva IGM, Silva-Carvalho AÉ, de Almeida MP, Albuquerque LFF, dos Santos RC, S. Brito AK, Saldanha-Araújo F, Arcanjo DDR, C. Martins MDC, dos S. Borges TK, Báo SN, Plácido A, Eaton P, Kuckelhaus SAS, Leite JRSA. Promising self-emulsifying drug delivery system loaded with lycopene from red guava (Psidium guajava L.): in vivo toxicity, biodistribution and cytotoxicity on DU-145 prostate cancer cells. Cancer Nanotechnol 2021. [DOI: 10.1186/s12645-021-00103-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Self-emulsifying drug delivery systems (SEDDSs) have attracted attention because of their effects on solubility and bioavailability of lipophilic compounds. Herein, a SEDDS loaded with lycopene purified from red guava (nanoLPG) was produced. The nanoemulsion was characterized using dynamic light scattering (DLS), zeta potential measurement, nanoparticle tracking analysis (NTA), transmission electron microscopy (TEM), Fourier-transform infrared spectroscopy (FTIR), lycopene content quantification, radical scavenging activity and colloidal stability in cell culture medium. Then, in vivo toxicity and tissue distribution in orally treated mice and cytotoxicity on human prostate carcinoma cells (DU-145) and human peripheral blood mononuclear cells (PBMC) were evaluated.
Results
NanoLPG exhibited physicochemical properties with a size around 200 nm, negative zeta-potential, and spherical morphology. The size, polydispersity index, and zeta potential parameters suffered insignificant alterations during the 12 month storage at 5 °C, which were associated with lycopene stability at 5 °C for 10 months. The nanoemulsion showed partial aggregation in cell culture medium at 37 °C after 24 h. NanoLPG at 0.10 mg/mL exhibited radical scavenging activity equivalent to 0.043 ± 0.002 mg Trolox/mL. The in vivo studies did not reveal any significant changes in clinical, behavioral, hematological, biochemical, and histopathological parameters in mice orally treated with nanoLPG at 10 mg/kg for 28 days. In addition, nanoLPG successfully delivered lycopene to the liver, kidney and prostate in mice, improved its cytotoxicity against DU-145 prostate cancer cells—probably by pathway independent on classical necrosis and apoptosis—and did not affect PBMC viability.
Conclusions
Thus, nanoLPG stands as a promising and biosafe lycopene delivery system for further development of nanotechnology-based health products.
Graphical Abstract
Collapse
|
43
|
Liu J, Zheng J, Xu Y, Cao W, Wang J, Wang B, Zhao L, Zhang X, Liao W. Enriched Environment Attenuates Pyroptosis to Improve Functional Recovery After Cerebral Ischemia/Reperfusion Injury. Front Aging Neurosci 2021; 13:717644. [PMID: 34646128 PMCID: PMC8504677 DOI: 10.3389/fnagi.2021.717644] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/01/2021] [Indexed: 12/16/2022] Open
Abstract
Enriched environment (EE) is a complex containing social, cognitive, and motor stimuli. Exposure to EE can promote functional recovery after ischemia/reperfusion (I/R) injury. However, the underlying mechanisms remained unclear. Pyroptosis has recently been identified and demonstrated a significant role in ischemic stroke. The purpose of this study was to explore the effect of EE on neuronal pyroptosis after cerebral I/R injury. In the current study, middle cerebral artery occlusion/reperfusion (MCAO/R) was applied to establish the cerebral I/R injury model. Behavior tests including the modified Neurological Severity Scores (mNSS) and the Morris Water Maze (MWM) were performed. The infarct volume was evaluated by Nissl staining. To evaluate the levels of pyroptosis-related proteins, the levels of GSDMD-N and nod-like receptor protein 1/3 (NLRP1/3) inflammasome-related proteins were examined. The mRNA levels of IL-1β and IL-18 were detected by Quantitative Real-Time PCR (qPCR). The secretion levels of IL-1β and IL-18 were analyzed by ELISA. Also, the expression of p65 and p-p65 were detected. The results showed that EE treatment improved functional recovery, reduced infarct volume, attenuated neuronal pyroptosis after cerebral I/R injury. EE treatment also suppressed the activities of NLRP1/NLRP3 inflammasomes. These may be affected by inhabiting the NF-κB p65 signaling pathway. Our findings suggested that neuronal pyroptosis was probably the neuroprotective mechanism that EE treatment rescued neurological deficits after I/R injury.
Collapse
Affiliation(s)
- Jingying Liu
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jun Zheng
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yang Xu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenyue Cao
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jinchen Wang
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Biru Wang
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Linyao Zhao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin Zhang
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Weijing Liao
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
44
|
Zhang B, Chen X, Ru F, Gan Y, Li B, Xia W, Dai G, He Y, Chen Z. Liproxstatin-1 attenuates unilateral ureteral obstruction-induced renal fibrosis by inhibiting renal tubular epithelial cells ferroptosis. Cell Death Dis 2021; 12:843. [PMID: 34511597 PMCID: PMC8435531 DOI: 10.1038/s41419-021-04137-1] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 08/16/2021] [Accepted: 09/02/2021] [Indexed: 12/23/2022]
Abstract
Renal fibrosis is a common pathological process that occurs with diverse etiologies in chronic kidney disease. However, its regulatory mechanisms have not yet been fully elucidated. Ferroptosis is a form of non-apoptotic regulated cell death driven by iron-dependent lipid peroxidation. It is currently unknown whether ferroptosis is initiated during unilateral ureteral obstruction (UUO)-induced renal fibrosis and its role has not been determined. In this study, we demonstrated that ureteral obstruction induced ferroptosis in renal tubular epithelial cells (TECs) in vivo. The ferroptosis inhibitor liproxstatin-1 (Lip-1) reduced iron deposition, cell death, lipid peroxidation, and inhibited the downregulation of GPX4 expression induced by UUO, ultimately inhibiting ferroptosis in TECs. We found that Lip-1 significantly attenuated UUO-induced morphological and pathological changes and collagen deposition of renal fibrosis in mice. In addition, Lip-1 attenuated the expression of profibrotic factors in the UUO model. In vitro, we used RSL3 treatment and knocked down of GPX4 level by RNAi in HK2 cells to induce ferroptosis. Our results indicated HK2 cells secreted various profibrotic factors during ferroptosis. Lip-1 was able to inhibit ferroptosis and thereby inhibit the secretion of the profibrotic factors during the process. Incubation of kidney fibroblasts with culture medium from RSL3-induced HK2 cells promoted fibroblast proliferation and activation, whereas Lip-1 impeded the profibrotic effects. Our study found that Lip-1 may relieve renal fibrosis by inhibiting ferroptosis in TECs. Mechanistically, Lip-1 could reduce the activation of surrounding fibroblasts by inhibiting the paracrine of profibrotic factors in HK2 cells. Lip-1 may potentially be used as a therapeutic approach for the treatment of UUO-induced renal fibrosis.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Xiang Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Feng Ru
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Yu Gan
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Bingsheng Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Weiping Xia
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Guoyu Dai
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Yao He
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.
| | - Zhi Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.
| |
Collapse
|
45
|
Communications between Mitochondria and Endoplasmic Reticulum in the Regulation of Metabolic Homeostasis. Cells 2021; 10:cells10092195. [PMID: 34571844 PMCID: PMC8468463 DOI: 10.3390/cells10092195] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 12/18/2022] Open
Abstract
Mitochondria associated membranes (MAM), which are the contact sites between endoplasmic reticulum (ER) and mitochondria, have emerged as an important hub for signaling molecules to integrate the cellular and organelle homeostasis, thus facilitating the adaptation of energy metabolism to nutrient status. This review explores the dynamic structural and functional features of the MAM and summarizes the various abnormalities leading to the impaired insulin sensitivity and metabolic diseases.
Collapse
|
46
|
Su JH, Luo MY, Liang N, Gong SX, Chen W, Huang WQ, Tian Y, Wang AP. Interleukin-6: A Novel Target for Cardio-Cerebrovascular Diseases. Front Pharmacol 2021; 12:745061. [PMID: 34504432 PMCID: PMC8421530 DOI: 10.3389/fphar.2021.745061] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/09/2021] [Indexed: 12/18/2022] Open
Abstract
Cardio-Cerebrovascular Disease is a collective term for cardiovascular disease and cerebrovascular disease, being a serious threat to human health. A growing number of studies have proved that the content of inflammatory factors or mediators determines the stability of vascular plaque and the incidence of cardio-cerebrovascular event, and involves in the process of Cardio-Cerebrovascular Diseases. Interleukin-6 is a widely used cytokine that causes inflammation and oxidative stress, which would further result in cardiac and cerebral injury. The increased expression of interleukin-6 is closely related to atherosclerosis, myocardial infarction, heart failure and ischemic stroke. It is a key risk factor for these diseases by triggering inflammatory reaction and inducing other molecules release. Therefore, interleukin-6 may become a potential target for Cardio-Cerebrovascular Diseases in the future. This paper is aimed to discuss the expression changes and pathological mechanisms of interleukin-6 in Cardio-Cerebrovascular Diseases, and to provide a novel strategy for the prevention and treatment of Cardio-Cerebrovascular Diseases.
Collapse
Affiliation(s)
- Jian-Hui Su
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| | - Meng-Yi Luo
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| | - Na- Liang
- Department of Anesthesiology, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Shao-Xin Gong
- Department of Pathology, First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Wei Chen
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| | - Wen-Qian Huang
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| | - Ying Tian
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Ai-Ping Wang
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
47
|
Wei Y, Zhu M, Li S, Hong T, Guo X, Li Y, Liu Y, Hou X, He B. Engineered Biomimetic Nanoplatform Protects the Myocardium Against Ischemia/Reperfusion Injury by Inhibiting Pyroptosis. ACS APPLIED MATERIALS & INTERFACES 2021; 13:33756-33766. [PMID: 34258997 DOI: 10.1021/acsami.1c03421] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Protection of cardiomyocytes against oxidative stress is vital to alleviate myocardial ischemia/reperfusion injury (MI/RI). However, antioxidative treatment is hampered by the lack of safe and effective therapeutics. Polydopamine (PDA), as a biodegradable class of nanomaterial with excellent antioxidant properties, has shown great potential in treating MI/RI. To achieve site-specific antioxidative efficacy, we established a PDA-based biomimetic nanoplatform (PDA@M), which consisted of a polydopamine core and a macrophage membrane shell to form a shell-core structure. By inheriting the inherent migration capability of macrophages, PDA@M was able to target the infarcted myocardium and exert an antioxidative effect to protect the myocardium. The results demonstrated that the accumulation of the membrane-wrapped nanoparticles (NPs) in the infarcted myocardium was greatly increased as compared with PDA alone, which effectively relieved the MI/RI-induced oxidative stress. PDA@M largely decreased the infarct size and improved the cardiac function post-MI/RI. Our study revealed that PDA@M could inhibit cell pyroptosis by suppressing the NLRP3/caspase-1 pathway, which is known to play a significant role in the antioxidant signaling pathway. In summary, PDA@M can target the infarcted myocardium and exert antioxidative and antipyroptosis functions to protect the myocardium against MI/RI-induced oxidative stress, suggesting that it may prove to be a potential therapeutic agent for MI/RI.
Collapse
Affiliation(s)
- Yazhong Wei
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Minfang Zhu
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Saiqi Li
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Ting Hong
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Xiaoyu Guo
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Yongyong Li
- Institute for Biomedical Engineering & Nano Science, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Yiqiong Liu
- Institute for Biomedical Engineering & Nano Science, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Xumin Hou
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Bin He
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| |
Collapse
|
48
|
Zhuo Y, Yuan R, Chen X, He J, Chen Y, Zhang C, Sun K, Yang S, Liu Z, Gao H. Tanshinone I exerts cardiovascular protective effects in vivo and in vitro through inhibiting necroptosis via Akt/Nrf2 signaling pathway. Chin Med 2021; 16:48. [PMID: 34183021 PMCID: PMC8240219 DOI: 10.1186/s13020-021-00458-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/16/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Tanshinone I (TI) is a primary component of Salvia miltiorrhiza Bunge (Danshen), which confers a favorable role in a variety of pharmacological activities including cardiovascular protection. However, the exact mechanism of the cardiovascular protection activity of TI remains to be illustrated. In this study, the cardiovascular protective effect and its mechanism of TI were investigated. METHODS In this study, tert-butyl hydroperoxide (t-BHP)-stimulated H9c2 cells model was employed to investigate the protective effect in vitro. The cell viability was determined by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide (MTT) assay and lactate dehydrogenase (LDH) kit. The reactive-oxygen-species (ROS) level and mitochondrial membrane potential (MMP) were investigated by the flow cytometry and JC-1 assay, respectively. While in vivo experiment, the cardiovascular protective effect of TI was determined by using myocardial ischemia-reperfusion (MI/R) model including hematoxylin-eosin (H&E) staining assay and determination of superoxide dismutase (SOD) and malondialdehyde (MDA). Tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) release were detected by Enzyme-linked immunosorbent assay (ELISA). Receptor interacting protein kinase 1 (RIP1), receptor interacting protein kinase 3 (RIP3), receptor interacting protein kinase 3 (MLKL), protein kinase B (Akt), Nuclear factor erythroid 2 related factor 2 (Nrf2), Heme oxygenase-1 (HO-1) and NAD(P)H: quinone oxidoreductase-1 (NQO-1) were determined by western blotting. RESULTS Our data demonstrated that TI pretreatment attenuated t-BHP and MI/R injury-induced necroptosis by inhibiting the expression of p-RIP1, p-RIP3, and p-MLKL. TI activated the Akt/Nrf2 pathway to promote the expression of antioxidant-related proteins such as phosphorylation of Akt, nuclear factor erythroid 2 related factor 2 (Nrf2), quinone oxidoreductase-1 (NQO-1) and heme oxygenase-1 (HO-1) expression in t-BHP-stimulated H9c2 cells. TI relieved oxidative stress by mitigating ROS generation and reversing MMP loss. In vivo experiment, TI made electrocardiograph (ECG) recovery better and lessened the degree of myocardial tissue damage. The counts of white blood cell (WBC), neutrophil (Neu), lymphocyte (Lym), and the release of TNF-α and IL-6 were reversed by TI treatment. SOD level was increased, while MDA level was decreased by TI treatment. CONCLUSION Collectively, our findings indicated that TI exerted cardiovascular protective activities in vitro and in vivo through suppressing RIP1/RIP3/MLKL and activating Akt/Nrf2 signaling pathways, which could be developed into a cardiovascular protective agent.
Collapse
Affiliation(s)
- Youqiong Zhuo
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Renyikun Yuan
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, China
| | - Xinxin Chen
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Jia He
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Yangling Chen
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Chenwei Zhang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Kaili Sun
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Shilin Yang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Zhenjie Liu
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Hongwei Gao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China.
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China.
| |
Collapse
|
49
|
Zhou Y, Liao J, Mei Z, Liu X, Ge J. Insight into Crosstalk between Ferroptosis and Necroptosis: Novel Therapeutics in Ischemic Stroke. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9991001. [PMID: 34257829 PMCID: PMC8257382 DOI: 10.1155/2021/9991001] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/30/2021] [Accepted: 06/14/2021] [Indexed: 12/16/2022]
Abstract
Ferroptosis is a nonapoptotic form of cell death characterized by iron-dependent accumulation of lipid hydroperoxides to lethal levels. Necroptosis, an alternative form of programmed necrosis, is regulated by receptor-interacting protein (RIP) 1 activation and by RIP3 and mixed-lineage kinase domain-like (MLKL) phosphorylation. Ferroptosis and necroptosis both play important roles in the pathological progress in ischemic stroke, which is a complex brain disease regulated by several cell death pathways. In the past few years, increasing evidence has suggested that the crosstalk occurs between necroptosis and ferroptosis in ischemic stroke. However, the potential links between ferroptosis and necroptosis in ischemic stroke have not been elucidated yet. Hence, in this review, we overview and analyze the mechanism underlying the crosstalk between necroptosis and ferroptosis in ischemic stroke. And we find that iron overload, one mechanism of ferroptosis, leads to mitochondrial permeability transition pore (MPTP) opening, which aggravates RIP1 phosphorylation and contributes to necroptosis. In addition, heat shock protein 90 (HSP90) induces necroptosis and ferroptosis by promoting RIP1 phosphorylation and suppressing glutathione peroxidase 4 (GPX4) activation. In this work, we try to deliver a new perspective in the exploration of novel therapeutic targets for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Yue Zhou
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Jun Liao
- Medical School, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Zhigang Mei
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei 443002, China
| | - Xun Liu
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Jinwen Ge
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- School of Medicine, Shaoyang University, Shaoyang, Hunan 422000, China
| |
Collapse
|
50
|
Karwi QG, Ho KL, Pherwani S, Ketema EB, Sun QY, Lopaschuk GD. Concurrent diabetes and heart failure: interplay and novel therapeutic approaches. Cardiovasc Res 2021; 118:686-715. [PMID: 33783483 DOI: 10.1093/cvr/cvab120] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Diabetes mellitus increases the risk of developing heart failure, and the co-existence of both diseases worsens cardiovascular outcomes, hospitalization and the progression of heart failure. Despite current advancements on therapeutic strategies to manage hyperglycemia, the likelihood of developing diabetes-induced heart failure is still significant, especially with the accelerating global prevalence of diabetes and an ageing population. This raises the likelihood of other contributing mechanisms beyond hyperglycemia in predisposing diabetic patients to cardiovascular disease risk. There has been considerable interest in understanding the alterations in cardiac structure and function in the diabetic patients, collectively termed as "diabetic cardiomyopathy". However, the factors that contribute to the development of diabetic cardiomyopathies is not fully understood. This review summarizes the main characteristics of diabetic cardiomyopathies, and the basic mechanisms that contribute to its occurrence. This includes perturbations in insulin resistance, fuel preference, reactive oxygen species generation, inflammation, cell death pathways, neurohormonal mechanisms, advanced glycated end-products accumulation, lipotoxicity, glucotoxicity, and posttranslational modifications in the heart of the diabetic. This review also discusses the impact of antihyperglycemic therapies on the development of heart failure, as well as how current heart failure therapies influence glycemic control in diabetic patients. We also highlight the current knowledge gaps in understanding how diabetes induces heart failure.
Collapse
Affiliation(s)
- Qutuba G Karwi
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Kim L Ho
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Simran Pherwani
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Ezra B Ketema
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Qiu Yu Sun
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Gary D Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|