1
|
Wei Z, Liu J, Liu H, Zhang X. Klotho alleviates sepsis-associated myocardial inflammation and apoptosis. Eur J Pharmacol 2025; 998:177653. [PMID: 40252895 DOI: 10.1016/j.ejphar.2025.177653] [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: 11/24/2024] [Revised: 04/02/2025] [Accepted: 04/17/2025] [Indexed: 04/21/2025]
Abstract
Klotho (KL) protects against various pathological phenotypes. The present study was designed to investigate the effect of KL on sepsis-associated cardiac dysfunction, and whether KL modulated sepsis-associated cardiac dysfunction via oxidative stress and apoptosis. Experiments were carried out in mice treated with lipopolysaccharide (LPS) to induce the septic model. The expression of KL was reduced in the heart of septic mice induced by LPS administration. The cardiac dysfunction of septic mice was deteriorated after KL KO, and was alleviated by KL upregulation. The inflammatory factors of tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β) and IL-6 were increased in the heart after LPS treatment, and these increases were further enhanced in KL deficiency mice, and were alleviated in KL overexpressed mice. The apoptosis biomarkers of B-cell lymphoma 2 (Bcl2) and Bcl2-associated X protein (Bax) levels were changed in the heart of septic mice, which were exacerbated by KL KO and were ameliorated by KL overexpression. These results indicated that KL involved in the regulation of sepsis-induced cardiac dysfunction via alleviating inflammation and apoptosis. Upregulation KL may be a therapeutic strategy to improve cardiac function in septic patients in the future.
Collapse
Affiliation(s)
- Zhongcheng Wei
- Department of Cardiology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China
| | - Juan Liu
- Department of Rheumatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China
| | - Hailang Liu
- Department of Cardiology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China
| | - Xiwen Zhang
- Department of Cardiology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China.
| |
Collapse
|
2
|
Cucinotta L, Palermo N, Ardizzone A, Capra AP, Campolo M, Esposito E, Casili G, Lanza M. The Inhibition of Prolyl Endopeptidase (PREP) by KYP-2047 Treatment to Reduce Myocardial Ischemia/Reperfusion Injury. Antioxidants (Basel) 2025; 14:442. [PMID: 40298805 PMCID: PMC12024445 DOI: 10.3390/antiox14040442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/02/2025] [Accepted: 04/05/2025] [Indexed: 04/30/2025] Open
Abstract
Myocardial ischemia-reperfusion injury (MI/R) is a negative and adverse cardiovascular outcome following myocardial ischemia, cardiac surgery, or circulatory arrest. Prolyl endopeptidase (PREP) appears to be involved in inflammatory responses, so it could be a possible therapeutic target for counteracting ischemia injury. This study aimed to investigate the role of PREP inhibitor, KYP-2047 (4-phenylbutanoyl-l-prolyl-2(S)-cyanopyrolidine), in the modulation of molecular and biochemical processes involved in MI/R. MI/R was induced through coronary artery occlusion (15 min), followed by reperfusion (2 h). KYP-2047 was intraperitoneally administrated at doses of 2.5 mg/kg and 5 mg/kg 24 h before the surgical procedures. The hearts were removed and processed for analysis. KYP-2047 treatment limited ischemic myocardial-induced histological damage and neutrophil accumulation, limiting inflammation, fibrosis, and apoptosis processes. Additionally, KYP-2047 was able to modulate p-38 and p-ERK expression, suggesting an improving role in recovering cardiac function. These findings highlighted the protective effects of KYP-2047 pretreatment in MI/R injury, suggesting PREP as a potential target therapy for the pathogenesis of MI/R. Although the molecular mechanisms underlying the action of KYP-2047 are still to be explored, these results suggested that the regulation of NF-κB, apoptosis, and MAPK pathways by KYP-2047 treatment could preventatively limit the damage caused by MI/R.
Collapse
Affiliation(s)
- Laura Cucinotta
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy
| | - Nicoletta Palermo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria, 1, 98125 Messina, Italy
| | - Alessio Ardizzone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy
| | - Anna Paola Capra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy
| | - Michela Campolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy
| | - Giovanna Casili
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy
| | - Marika Lanza
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy
| |
Collapse
|
3
|
Zi C, Ma X, Zheng M, Zhu Y. VDAC1-NF-κB/p65-mediated S100A16 contributes to myocardial ischemia/reperfusion injury by regulating oxidative stress and inflammatory response via calmodulin/CaMKK2/AMPK pathway. Eur J Pharmacol 2025; 987:177158. [PMID: 39613175 DOI: 10.1016/j.ejphar.2024.177158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/05/2024] [Accepted: 11/27/2024] [Indexed: 12/01/2024]
Abstract
Myocardial injury triggers intense inflammatory reactions and oxidative stress responses. S100 calcium-binding protein A16 (S100A16), a multi-functional calcium (Ca2+)-binding protein, participates in inflammatory responses and contributes to ischemia/reperfusion (I/R) injury. Nevertheless, the precise mechanism by which S100A16 operates in myocardial I/R injury remains uncertain. Cardiac I/R injury was produced by ligation/release of the left anterior descending artery, and mouse cardiac cells were subjected to hypoxia/reoxygenation (H/R) to determine the biological effects in vitro. We demonstrated that S100A16 was upregulated in the ischemic hearts and cardiac cells after I/R and H/R injury. Adenovirus-mediated S100A16 inhibition led to a considerable improvement in cardiac function with a reduced infarct size, accompanied by a reduction in cardiomyocyte apoptosis. Similar effects of S100A16 inhibition on inflammation and reactive oxygen species (ROS) production were observed in cultured cardiomyocytes. Importantly, we showed that I/R and H/R treatment upregulated the expression of voltage-dependent anion channel 1 (VDAC1), which subsequently activated NF-κB/p65 to facilitate the binding of NF-κB/p65 to the S100A16 promoter, thereby activating the transcription and expression of S100A16. Mechanically, S100A16 responded to increasing Ca2+ and interacted with calmodulin (CaM) to regulate the activation of calcium/calmodulin-dependent protein kinase 2 (CAMKK2)/AMPK pathway. In conclusion, VDAC1 sustained the NF-κB p65 pathway activation to elicit increased S100A16 expression, contributing to myocardial damage and heart failure post-I/R via the CaM/CaMKK2/AMPK pathway. This study revealed a crucial role of the VDAC1-S100A16 axis in the process of myocardial I/R injury, providing novel molecular targets for the treatment of cardiac conditions associated with I/R injury.
Collapse
Affiliation(s)
- Congna Zi
- Department of Anesthesiology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, China.
| | - Xian Ma
- Department of Blood Transfusion, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, China.
| | - Maodong Zheng
- Department of Pharmacy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, China
| | - Ying Zhu
- Department of Anesthesiology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, China
| |
Collapse
|
4
|
He S, Li C, Lu M, Lin F, Hu S, Zhang J, Peng L, Li L. Comprehensive analysis of scRNA-seq and bulk RNA-seq reveals the non-cardiomyocytes heterogeneity and novel cell populations in dilated cardiomyopathy. J Transl Med 2025; 23:17. [PMID: 39762897 PMCID: PMC11702085 DOI: 10.1186/s12967-024-05983-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is one of the most common causes of heart failure. Infiltration and alterations in non-cardiomyocytes of the human heart involve crucially in the occurrence of DCM and associated immunotherapeutic approaches. METHODS We constructed a single-cell transcriptional atlas of DCM and normal patients. Then, the xCell algorithm, EPIC algorithm, MCP counter algorithm, and CIBERSORT method were applied to identify DCM-related cell types with a high degree of precision and specificity using RNA-seq datasets. We further analyzed the heterogeneity among cell types, performed trajectory analysis, examined transcription factor regulatory networks, investigated metabolic heterogeneity, and conducted intercellular communication analysis. Finally, we used bulk RNA-seq data to confirm the roles of M2-like2 subpopulations and GAS6 in DCM. RESULTS We integrated and analyzed Single-cell sequencing (scRNA-seq) data from 7 DCM samples and 3 normal heart tissue samples, totaling 70,958 single-cell data points. Based on gene-specific expression and prior marker genes, we identified 9 distinct subtypes, including fibroblasts, endothelial cells, myeloid cells, pericytes, T/NK cells, smooth muscle cells, neuronal cells, B cells, and cardiomyocytes. Using machine learning methods to quantify bulk RNA-seq data, we found significant differences in fibroblasts, T cells, and macrophages between DCM and normal samples. Further analysis revealed high heterogeneity in tissue preference, gene expression, functional enrichment, immunodynamics, transcriptional regulatory factors, metabolic changes, and communication patterns in fibroblasts and myeloid cells. Among fibroblast subpopulations, proliferative F3 cells were implicated in the fibroblast transition process in DCM, while myofibroblast F6 cells promoted the fibroblast transition to a late cell state in DCM. Additionally, two subpopulations of M2 macrophages, M2-like1 and M2-like2, were identified with distinct features. The M2-like2 cell subpopulation, which was enriched in glycolysis and fatty acid metabolism, involved in inflammation inhibition and fibrosis promotion. Cell‒cell communication analysis indicated the GAS6-MERTK axis might exhibit interaction between M2 macrophage and M2-like1 macrophage. Furthermore, deconvolution analysis for bulk RNA-seq data revealed a significant increase in M2-like2 subpopulations in DCM, suggesting a more important role for this cell population in DCM. CONCLUSIONS We revealed the heterogeneity of non-cardiomyocytes in DCM and identified subpopulations of myofibroblast and macrophages engaged in DCM, which suggested a potential significance of non-cardiomyocytes in treatment of DCM.
Collapse
Affiliation(s)
- Siyu He
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, School of Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmias Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Stem Cell Research Center, Medical School, Tongji University, Shanghai, 200120, China
| | - Chunyu Li
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, School of Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmias Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Stem Cell Research Center, Medical School, Tongji University, Shanghai, 200120, China
| | - Mingxin Lu
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, School of Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmias Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Stem Cell Research Center, Medical School, Tongji University, Shanghai, 200120, China
| | - Fang Lin
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, School of Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmias Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Stem Cell Research Center, Medical School, Tongji University, Shanghai, 200120, China
| | - Sangyu Hu
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, School of Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmias Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Stem Cell Research Center, Medical School, Tongji University, Shanghai, 200120, China
| | - Junfang Zhang
- Stem Cell Research Center, Medical School, Tongji University, Shanghai, 200120, China
| | - Luying Peng
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, School of Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200120, China.
- Shanghai Arrhythmias Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
- Stem Cell Research Center, Medical School, Tongji University, Shanghai, 200120, China.
- Department of Cell and Genetics, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Li Li
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, School of Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200120, China.
- Shanghai Arrhythmias Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
- Stem Cell Research Center, Medical School, Tongji University, Shanghai, 200120, China.
- Department of Cell and Genetics, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
5
|
Pan C, Shen R, Ding Y, Li Z, Dong C, Zhang J, Zhu R, Yu K, Zeng Q. Interleukin-38 ameliorates myocardial Ischemia-Reperfusion injury via inhibition of NLRP3 inflammasome activation in fibroblasts through the IL-1R8/SYK axis. Int Immunopharmacol 2024; 143:113428. [PMID: 39447412 DOI: 10.1016/j.intimp.2024.113428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024]
Abstract
OBJECTIVE Although IL-38 is recognized for its regulatory role in a spectrum of chronic inflammatory diseases, investigations into its cardiac physiological and pathophysiological functions are nascent. Our aim was to delineate the biological impact of IL-38 in the context of myocardial ischemia-reperfusion injury (MIRI) and to uncover the mechanisms through which it exerts its effects. METHODS AND RESULTS In this study, we used an MIRI mouse model, LPS/ATP stimulation, and a hypoxia/reoxygenation cell model to determine the regulatory influence of IL-38 on MIRI. We observed that the administration of recombinant IL-38 to mice led to a reduction in infarct size, an enhancement in cardiac function, and a suppression of NLRP3 inflammasome activation. In contrast, genetic deletion of IL-38 was associated with an increase in infarct size, worsening of cardiac function, and upregulation of NLRP3 inflammasome activity. The detrimental effects associated with the absence of IL-38 were mitigated by the administration of a specific NLRP3 inhibitor, suggesting that the inhibition of NLRP3 is a critical component of the protective effect mediated by IL-38 in MIRI. In vitro assays revealed that IL-38 inhibited NLRP3 inflammasome activation in cardiac fibroblasts through the engagement of IL-1R8 and the modulation of SYK phosphorylation. Silencing of IL-1R8 negated the suppressive effect of IL-38 on the NLRP3 inflammasome. CONCLUSION IL-38 acts as a potent negative regulator of inflammasome activation after MIRI. It achieves this regulatory effect within cardiac fibroblasts by inhibiting SYK phosphorylation, a process mediated by IL-1R8.
Collapse
Affiliation(s)
- Chengliang Pan
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Rui Shen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Yan Ding
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Zhiyang Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Chen Dong
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jiangmei Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Ruirui Zhu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Kunwu Yu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China.
| | - Qiutang Zeng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China.
| |
Collapse
|
6
|
Duan Z, Huang Z, Lei W, Zhang K, Xie W, Jin H, Wu M, Wang N, Li X, Xu A, Zhou H, Wu F, Li Y, Lin Z. Bone Morphogenetic Protein 9 Protects Against Myocardial Infarction by Improving Lymphatic Drainage Function and Triggering DECR1-Mediated Mitochondrial Bioenergetics. Circulation 2024; 150:1684-1701. [PMID: 39315433 DOI: 10.1161/circulationaha.123.065935] [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: 06/14/2023] [Accepted: 08/01/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND BMP9 (bone morphogenetic protein 9) is a member of the TGF-β (transforming growth factor β) family of cytokines with pleiotropic effects on glucose metabolism, fibrosis, and lymphatic development. However, the role of BMP9 in myocardial infarction (MI) remains elusive. METHODS The expressional profiles of BMP9 in cardiac tissues and plasma samples of subjects with MI were determined by immunoassay or immunoblot. The role of BMP9 in MI was determined by evaluating the impact of BMP9 deficiency and replenishment with adeno-associated virus-mediated BMP9 expression or recombinant human BMP9 protein in mice. RESULTS We show that circulating BMP9 and its cardiac levels are markedly increased in humans and mice with MI and are negatively associated with cardiac function. It is important to note that BMP9 deficiency exacerbates left ventricular dysfunction, increases infarct size, and augments cardiac fibrosis in mice with MI. In contrast, replenishment of BMP9 significantly attenuates these adverse effects. We further demonstrate that BMP9 improves lymphatic drainage function, thereby leading to a decrease of cardiac edema. In addition, BMP9 increases the expression of mitochondrial DECR1 (2,4-dienoyl-CoA [coenzyme A] reductase 1), a rate-limiting enzyme involved in β-oxidation, which, in turn, promotes cardiac mitochondrial bioenergetics and mitigates MI-induced cardiomyocyte injury. Moreover, DECR1 deficiency exacerbates MI-induced cardiac damage in mice, whereas this adverse effect is restored by the treatment of adeno-associated virus-mediated DECR1. Consistently, DECR1 deletion abrogates the beneficial effect of BMP9 against MI-induced cardiomyopathy and cardiac damage in mice. CONCLUSIONS These results suggest that BMP9 protects against MI by fine-tuning the multiorgan cross-talk among the liver, lymph, and the heart.
Collapse
Affiliation(s)
- Zikun Duan
- Affiliated Dongguan Songshan Lake Central Hospital (Z.D., F.W., Z.L.), Guangdong Medical University, Dongguan, China
| | - Zhouqing Huang
- Department of Cardiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (Z.H., W.X., H.Z., Z.L.)
| | - Wei Lei
- Precision Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China (W.L.)
| | - Ke Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, China (K.Z., H.J., M.W., N.W., X.L., Z.L.)
| | - Wei Xie
- Department of Cardiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (Z.H., W.X., H.Z., Z.L.)
| | - Hua Jin
- School of Pharmaceutical Sciences, Wenzhou Medical University, China (K.Z., H.J., M.W., N.W., X.L., Z.L.)
| | - Maolan Wu
- School of Pharmaceutical Sciences, Wenzhou Medical University, China (K.Z., H.J., M.W., N.W., X.L., Z.L.)
| | - Ningrui Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, China (K.Z., H.J., M.W., N.W., X.L., Z.L.)
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, China (K.Z., H.J., M.W., N.W., X.L., Z.L.)
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, China (A.X.)
| | - Hao Zhou
- Department of Cardiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (Z.H., W.X., H.Z., Z.L.)
| | - Fan Wu
- Affiliated Dongguan Songshan Lake Central Hospital (Z.D., F.W., Z.L.), Guangdong Medical University, Dongguan, China
- Innovation Center of Cardiometabolic Disease (F.W., Z.L.), Guangdong Medical University, Dongguan, China
| | - Yulin Li
- Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Anzhen Hospital of Capital Medical University, China (Y.L.)
| | - Zhuofeng Lin
- Affiliated Dongguan Songshan Lake Central Hospital (Z.D., F.W., Z.L.), Guangdong Medical University, Dongguan, China
- Innovation Center of Cardiometabolic Disease (F.W., Z.L.), Guangdong Medical University, Dongguan, China
- Department of Cardiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (Z.H., W.X., H.Z., Z.L.)
- School of Pharmaceutical Sciences, Wenzhou Medical University, China (K.Z., H.J., M.W., N.W., X.L., Z.L.)
| |
Collapse
|
7
|
Chen X, Yang Y, Zhou Z, Yu H, Zhang S, Huang S, Wei Z, Ren K, Jin Y. Unraveling the complex interplay between Mitochondria-Associated Membranes (MAMs) and cardiovascular Inflammation: Molecular mechanisms and therapeutic implications. Int Immunopharmacol 2024; 141:112930. [PMID: 39146786 DOI: 10.1016/j.intimp.2024.112930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/26/2024] [Accepted: 08/10/2024] [Indexed: 08/17/2024]
Abstract
Cardiovascular diseases (CVDs) represent a significant public health concern because of their associations with inflammation, oxidative stress, and abnormal remodeling of the heart and blood vessels. In this review, we discuss the intricate interplay between mitochondria-associated membranes (MAMs) and cardiovascular inflammation, highlighting their role in key cellular processes such as calcium homeostasis, lipid metabolism, oxidative stress management, and ERS. We explored how these functions impact the pathogenesis and progression of various CVDs, including myocardial ischemia-reperfusion injury, atherosclerosis, diabetic cardiomyopathy, cardiovascular aging, heart failure, and pulmonary hypertension. Additionally, we examined current therapeutic strategies targeting MAM-related pathways and proteins, emphasizing the potential of MAMs as therapeutic targets. Our review aims to provide new insights into the mechanisms of cardiovascular inflammation and propose novel therapeutic approaches to improve cardiovascular health outcomes.
Collapse
Affiliation(s)
- Xing Chen
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Yang Yang
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Zheng Zhou
- Department of Geriatric Endocrinology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Haihan Yu
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Shuwei Zhang
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Siyuan Huang
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Ziqing Wei
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| | - Yage Jin
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| |
Collapse
|
8
|
Pan L, Fu M, Tang XL, Ling Y, Su Y, Ge J. Kirenol Ameliorates Myocardial Ischemia-Reperfusion Injury by Promoting Mitochondrial Function and Inhibiting Inflammasome Activation. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07635-4. [PMID: 39531114 DOI: 10.1007/s10557-024-07635-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE Macrophage-mediated inflammation plays a crucial role in the pathophysiological process of myocardial ischemia/reperfusion (I/R) injury. Recent studies have highlighted the importance of mitochondrial function and inflammasome activation in the inflammatory process. Kirenol, a well-known natural compound, has been shown to regulate inflammation in various diseases. This study investigated whether Kirenol could exert anti-inflammatory effects on macrophages during myocardial I/R injury. METHODS Mouse myocardial I/R models were established by 45 min of ischemia followed by 24 h of reperfusion. Saline or Kirenol treatment was administered. In vivo assessments included the evaluation of cardiac function, infarcted area, and immune cell infiltration. Subsequently, bone marrow-derived macrophages (BMDMs) were isolated, and mitochondrial function and pyroptosis were assessed. Furthermore, the study compared the cardioprotective effects of Kirenol with a specific NOX1/NOX4 inhibitor, GKT137831. RESULTS Kirenol gavage improved cardiac function, decreased infarct area, and alleviated inflammatory infiltration in mice subjected to myocardial I/R injury. Mechanistically, Kirenol inhibited NOX1 and NOX4 and enhanced mitochondrial function, ultimately attenuating the pyroptosis of macrophages. The therapeutic effects of Kirenol and GKT137831 were not significantly different. CONCLUSION This study demonstrates that Kirenol mitigates myocardial I/R injury by inhibiting NOX1 and NOX4, restoring mitochondrial function, and ameliorating macrophage pyroptosis.
Collapse
Affiliation(s)
- Lei Pan
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Fenglin Road 180, Shanghai, 200032, Xuhui District, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Mingqiang Fu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Fenglin Road 180, Shanghai, 200032, Xuhui District, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiang-Lin Tang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Fenglin Road 180, Shanghai, 200032, Xuhui District, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Yunlong Ling
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Fenglin Road 180, Shanghai, 200032, Xuhui District, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Yangang Su
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Fenglin Road 180, Shanghai, 200032, Xuhui District, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, China.
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
- NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China.
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Beijing, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Fenglin Road 180, Shanghai, 200032, Xuhui District, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
9
|
Todorova VK, Azhar G, Stone A, Malapati SJ, Che Y, Zhang W, Makhoul I, Wei JY. Neutrophil Biomarkers Can Predict Cardiotoxicity of Anthracyclines in Breast Cancer. Int J Mol Sci 2024; 25:9735. [PMID: 39273682 PMCID: PMC11395913 DOI: 10.3390/ijms25179735] [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: 08/11/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
Doxorubicin (DOX), a commonly used anticancer agent, causes cardiotoxicity that begins with the first dose and may progress to heart failure years after treatment. An inflammatory response associated with neutrophil recruitment has been recognized as a mechanism of DOX-induced cardiotoxicity. This study aimed to validate mRNA expression of the previously identified biomarkers of DOX-induced cardiotoxicity, PGLYRP1, CAMP, MMP9, and CEACAM8, and to assay their protein expression in the peripheral blood of breast cancer patients. Blood samples from 40 breast cancer patients treated with DOX-based chemotherapy were collected before and after the first chemotherapy cycle and > 2 years after treatment. The protein and gene expression of PGLYRP1/Tag7, CAMP/LL37, MMP9/gelatinase B, and CEACAM8/CD66b were determined using ELISA and reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Receiver operating characteristic (ROC) curve analysis was used to determine the diagnostic value of each candidate biomarker. Patients with cardiotoxicity (n = 20) had significantly elevated levels of PGLYRP1, CAMP, MMP9, and CEACAM8 at baseline, after the first dose of DOX-based chemotherapy, and at > 2 years after treatment relative to patients without cardiotoxicity (n = 20). The first dose of DOX induced significantly higher levels of all examined biomarkers in both groups of patients. At > 2 years post treatment, the levels of all but MMP9 dropped below the baseline. There was a good correlation between the expression of mRNA and the target proteins. We demonstrate that circulating levels of PGLYRP1, CAMP, MMP9, and CEACAM8 can predict the cardiotoxicity of DOX. This novel finding may be of value in the early identification of patients at risk for cardiotoxicity.
Collapse
Affiliation(s)
- Valentina K Todorova
- Division of Hematology/Oncology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | - Gohar Azhar
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Annjanette Stone
- Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | - Sindhu J Malapati
- Division of Hematology/Oncology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Yingni Che
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Wei Zhang
- Department of Mathematics and Statistics, University of Arkansas at Little Rock, Little Rock, AR 72205, USA
| | - Issam Makhoul
- Division of Hematology/Oncology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jeanne Y Wei
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
10
|
Zhang Y, Cao Y, Xin Y, Liu Y. Significance of detecting cardiac troponin I and creatine kinase MB in critically Ill children without primary cardiac illness. Front Pediatr 2024; 12:1445651. [PMID: 39286452 PMCID: PMC11402721 DOI: 10.3389/fped.2024.1445651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024] Open
Abstract
Objective To investigate the incidence of myocardial injury in children with critically ill children without primary cardiac disease and the association between elevated cardiac troponin I (cTnl) and creatine kinase MB (CK-MB) concentrations and disease progression and prognosis to guide early treatment. Methods The serum cTnI and CK-MB concentrations of 292 children with critically ill children without primary cardiac disease in Yantai Yuhuangding Hospital between January 2021 and January 2024 were retrospectively analyzed within 24 h after entering the Pediatric Intensive Care Unit (PICU). The children were divided into normal and abnormal groups according to the myocardial marker results. The abnormal group was further divided into the cTnI-elevated, CK-MB-elevated, single-elevated (cTnI- or CK-MB-elevated) and double-elevated (cTnI- and CK-MB-elevated) groups. The differences in the clinical indicators and their relationships with prognosis for the groups were compared. Results The incidence of myocardial injury among the critically ill children without primary cardiac disease was 55.1%. The incidence of myocardial injury in children with infectious diarrhea combined with moderate and severe dehydration reached 85.19%. The pediatric critical illness score; frequency of use of vasoactive drugs; hypotension, shock, heart failure, respiratory failure, and multiple organ dysfunction syndrome; and mortality indexes differed significantly for the normal and abnormal myocardial marker groups (P < 0.05). The single-elevated and normal groups only showed a difference in mortality (P < 0.017). The cTnI and CK-MB concentrations were negatively correlated with prognosis (P < 0.01). Conclusion Myocardial injury, as evidenced by elevated cardiac biomarkers, is common in critically ill children without primary cardiac illness. cTnI and CK-MB are associated with outcomes. Shock, heart failure, and multiple organ dysfunction syndromes are independently associated with simultaneous elevations of CK-MB and cTnI concentrations. Further prospective studies are needed to elucidate the clinical utility of these biomarkers.
Collapse
Affiliation(s)
- Yangyang Zhang
- Department of Pediatrics, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Yinyin Cao
- Department of Pediatrics, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Yi Xin
- Department of Pediatrics, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Yongming Liu
- Clinical Laboratory, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, Shandong, China
| |
Collapse
|
11
|
Wan H, Yang X, Zhang Y, Liu X, Li Y, Qin Y, Yan H, Gui L, Li K, Zhang L, Yang L, Zhang B, Wang Y. Polyphenol-Reinforced Glycocalyx-Like Hydrogel Coating Induced Myocardial Regeneration and Immunomodulation. ACS NANO 2024; 18:21512-21522. [PMID: 39096486 DOI: 10.1021/acsnano.4c06332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/05/2024]
Abstract
Although minimally invasive interventional occluders can effectively seal heart defect tissue, they still have some limitations, including poor endothelial healing, intense inflammatory response, and thrombosis formation. Herein, a polyphenol-reinforced medicine/peptide glycocalyx-like coating was prepared on cardiac occluders. A coating consisting of carboxylated chitosan, epigallocatechin-3-gallate (EGCG), tanshinone IIA sulfonic sodium (TSS), and hyaluronic acid grafted with 3-aminophenylboronic acid was prepared. Subsequently, the mercaptopropionic acid-GGGGG-Arg-Glu-Asp-Val peptide was grafted by the thiol-ene "click" reaction. The coating showed good hydrophilicity and free radical-scavenging ability and could release EGCG-TSS. The results of biological experiments suggested that the coating could reduce thrombosis by promoting endothelialization, and promote myocardial repair by regulating the inflammatory response. The functions of regulating cardiomyocyte apoptosis and metabolism were confirmed, and the inflammatory regulatory functions of the coating were mainly dependent on the NF-kappa B and TNF signaling pathway.
Collapse
Affiliation(s)
- Huining Wan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Xiaohui Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yutong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Xiyu Liu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yanyan Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yumei Qin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Hui Yan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Lan Gui
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Ke Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Longjian Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Li Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Bo Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
- Bioengineering Department, University of California, Los Angeles, California 90095, United States
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| |
Collapse
|
12
|
Nakamori S, Amyar A, Fahmy AS, Ngo LH, Ishida M, Nakamura S, Omori T, Moriwaki K, Fujimoto N, Imanaka-Yoshida K, Sakuma H, Dohi K, Manning WJ, Nezafat R. Cardiovascular Magnetic Resonance Radiomics to Identify Components of the Extracellular Matrix in Dilated Cardiomyopathy. Circulation 2024; 150:7-18. [PMID: 38808522 PMCID: PMC11216881 DOI: 10.1161/circulationaha.123.067107] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 04/30/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Current cardiovascular magnetic resonance sequences cannot discriminate between different myocardial extracellular space (ECSs), including collagen, noncollagen, and inflammation. We sought to investigate whether cardiovascular magnetic resonance radiomics analysis can distinguish between noncollagen and inflammation from collagen in dilated cardiomyopathy. METHODS We identified data from 132 patients with dilated cardiomyopathy scheduled for an invasive septal biopsy who underwent cardiovascular magnetic resonance at 3 T. Cardiovascular magnetic resonance imaging protocol included native and postcontrast T1 mapping and late gadolinium enhancement (LGE). Radiomic features were computed from the midseptal myocardium, near the biopsy region, on native T1, extracellular volume (ECV) map, and LGE images. Principal component analysis was used to reduce the number of radiomic features to 5 principal radiomics. Moreover, a correlation analysis was conducted to identify radiomic features exhibiting a strong correlation (r>0.9) with the 5 principal radiomics. Biopsy samples were used to quantify ECS, myocardial fibrosis, and inflammation. RESULTS Four histopathological phenotypes were identified: low collagen (n=20), noncollagenous ECS expansion (n=49), mild to moderate collagenous ECS expansion (n=42), and severe collagenous ECS expansion (n=21). Noncollagenous expansion was associated with the highest risk of myocardial inflammation (65%). Although native T1 and ECV provided high diagnostic performance in differentiating severe fibrosis (C statistic, 0.90 and 0.90, respectively), their performance in differentiating between noncollagen and mild to moderate collagenous expansion decreased (C statistic: 0.59 and 0.55, respectively). Integration of ECV principal radiomics provided better discrimination and reclassification between noncollagen and mild to moderate collagen (C statistic, 0.79; net reclassification index, 0.83 [95% CI, 0.45-1.22]; P<0.001). There was a similar trend in the addition of native T1 principal radiomics (C statistic, 0.75; net reclassification index, 0.93 [95% CI, 0.56-1.29]; P<0.001) and LGE principal radiomics (C statistic, 0.74; net reclassification index, 0.59 [95% CI, 0.19-0.98]; P=0.004). Five radiomic features per sequence were identified with correlation analysis. They showed a similar improvement in performance for differentiating between noncollagen and mild to moderate collagen (native T1, ECV, LGE C statistic, 0.75, 0.77, and 0.71, respectively). These improvements remained significant when confined to a single radiomic feature (native T1, ECV, LGE C statistic, 0.71, 0.70, and 0.64, respectively). CONCLUSIONS Radiomic features extracted from native T1, ECV, and LGE provide incremental information that improves our capability to discriminate noncollagenous expansion from mild to moderate collagen and could be useful for detecting subtle chronic inflammation in patients with dilated cardiomyopathy.
Collapse
Affiliation(s)
- Shiro Nakamori
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
- Department of Cardiology and Nephrology, University Graduate School of Medicine, Tsu, Mie, Japan
| | - Amine Amyar
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Ahmed S Fahmy
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Long H. Ngo
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Masaki Ishida
- Department of Radiology, and University Graduate School of Medicine, Tsu, Mie, Japan
| | - Satoshi Nakamura
- Department of Radiology, and University Graduate School of Medicine, Tsu, Mie, Japan
| | - Taku Omori
- Department of Cardiology and Nephrology, University Graduate School of Medicine, Tsu, Mie, Japan
| | - Keishi Moriwaki
- Department of Cardiology and Nephrology, University Graduate School of Medicine, Tsu, Mie, Japan
| | - Naoki Fujimoto
- Department of Cardiology and Nephrology, University Graduate School of Medicine, Tsu, Mie, Japan
| | - Kyoko Imanaka-Yoshida
- Department of Pathology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Hajime Sakuma
- Department of Radiology, and University Graduate School of Medicine, Tsu, Mie, Japan
| | - Kaoru Dohi
- Department of Cardiology and Nephrology, University Graduate School of Medicine, Tsu, Mie, Japan
| | - Warren J Manning
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
- Department of Radiology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Reza Nezafat
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
13
|
Ma J, Wang X, Jia Y, Tan F, Yuan X, Du J. The roles of B cells in cardiovascular diseases. Mol Immunol 2024; 171:36-46. [PMID: 38763105 DOI: 10.1016/j.molimm.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/30/2024] [Accepted: 05/04/2024] [Indexed: 05/21/2024]
Abstract
Damage to the heart can start the repair process and cause cardiac remodeling. B cells play an important role in this process. B cells are recruited to the injured place and activate cardiac remodeling through secreting antibodies and cytokines. Different types of B cells showed specific functions in the heart. Among all types of B cells, heart-associated B cells play a vital role in the heart by secreting TGFβ1. B cells participate in the activation of fibroblasts and promote cardiac fibrosis. Four subtypes of B cells in the heart revealed the relationship between the B cells' heterogeneity and cardiac remodeling. Many cardiovascular diseases like atherosclerosis, heart failure (HF), hypertension, myocardial infarction (MI), and dilated cardiomyopathy (DCM) are related to B cells. The primary mechanisms of these B cell-related activities will be discussed in this review, which may also suggest potential novel therapeutic targets.
Collapse
Affiliation(s)
- Jian Ma
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaotong Wang
- Department of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yuewang Jia
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fangyan Tan
- Department of Nephrology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Yuan
- Department of Nephrology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Jianlin Du
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
14
|
Tang J, Tam E, Song E, Xu A, Sweeney G. Crosstalk between myocardial autophagy and sterile inflammation in the development of heart failure. AUTOPHAGY REPORTS 2024; 3:2320605. [PMID: 40395524 PMCID: PMC11864620 DOI: 10.1080/27694127.2024.2320605] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/02/2024] [Accepted: 02/13/2024] [Indexed: 05/22/2025]
Abstract
Heart failure, a leading driver of global mortality, remains a topic of intense contemporary research interest due to the prevailing unmet need in cardiometabolic therapeutics. Numerous mechanisms with the potential to influence the onset and development of heart failure remain incompletely understood. Firstly, myocardial autophagy, which involves lysosomal degradation of damaged cellular components, confers context-dependent beneficial and detrimental effects. Secondly, sterile inflammation may arise following cardiac stress and exacerbate the progression of heart failure. Inflammation changes in a temporal manner and its onset must be adequately resolved to limit progression of heart failure. Mitochondria are an important factor in contributing to sterile inflammation by releasing damage associated molecular patterns (DAMPs) including mitochondrial DNA (mtDNA). Accordingly, this is one reason why the selective autophagy of mitochondria to maintain optimal function is important in determining cardiac function. In this review, we examine the increasing evidence suggesting crosstalk between autophagy and sterile inflammation together with their role in the development of heart failure. In particular, this is exemplified in the preclinical models of ischaemia/reperfusion injury and pressure overload induced heart failure. We also highlight potential therapeutic approaches focusing on autophagy and addressing sterile inflammation, aiming to enhance outcomes in heart failure.
Collapse
Affiliation(s)
- Jialing Tang
- Department of Biology, York University, Toronto, ON, Canada
| | - Eddie Tam
- Department of Biology, York University, Toronto, ON, Canada
| | - Erfei Song
- Department of Medicine, School of Clinical Medicine, State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Metabolic and Bariatric Surgery, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Aimin Xu
- Department of Medicine, School of Clinical Medicine, State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON, Canada
| |
Collapse
|
15
|
Samani SL, Barlow SC, Freeburg LA, Jones TL, Poole M, Sarzynski MA, Zile MR, Shazly T, Spinale FG. Left ventricle function and post-transcriptional events with exercise training in pigs. PLoS One 2024; 19:e0292243. [PMID: 38306359 PMCID: PMC10836705 DOI: 10.1371/journal.pone.0292243] [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] [Received: 10/13/2022] [Accepted: 09/14/2023] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Standardized exercise protocols have been shown to improve overall cardiovascular fitness, but direct effects on left ventricular (LV) function, particularly diastolic function and relation to post-transcriptional molecular pathways (microRNAs (miRs)) are poorly understood. This project tested the central hypothesis that adaptive LV remodeling resulting from a large animal exercise training protocol, would be directly associated with specific miRs responsible for regulating pathways relevant to LV myocardial stiffness and geometry. METHODS AND RESULTS Pigs (n = 9; 25 Kg) underwent a 4 week exercise training protocol (10 degrees elevation, 2.5 mph, 10 min, 5 days/week) whereby LV chamber stiffness (KC) and regional myocardial stiffness (rKm) were measured by Doppler/speckle tracking echocardiography. Age and weight matched non-exercise pigs (n = 6) served as controls. LV KC fell by approximately 50% and rKm by 30% following exercise (both p < 0.05). Using an 84 miR array, 34 (40%) miRs changed with exercise, whereby 8 of the changed miRs (miR-19a, miR-22, miR-30e, miR-99a, miR-142, miR-144, miR-199a, and miR-497) were correlated to the change in KC (r ≥ 0.5 p < 0.05) and mapped to matrix and calcium handling processes. Additionally, miR-22 and miR-30e decreased with exercise and mapped to a localized inflammatory process, the inflammasome (NLRP-3, whereby a 2-fold decrease in NLRP-3 mRNA occurred with exercise (p < 0.05). CONCLUSION Chronic exercise reduced LV chamber and myocardial stiffness and was correlated to miRs that map to myocardial relaxation processes as well as local inflammatory pathways. These unique findings set the stage for utilization of myocardial miR profiling to identify underlying mechanisms by which exercise causes changes in LV myocardial structure and function.
Collapse
Affiliation(s)
- Stephanie L. Samani
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, United States of America
- Columbia VA Health Care System, Columbia, SC, United States of America
| | - Shayne C. Barlow
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, United States of America
| | - Lisa A. Freeburg
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, United States of America
- Columbia VA Health Care System, Columbia, SC, United States of America
| | - Traci L. Jones
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, United States of America
| | - Marlee Poole
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, United States of America
| | - Mark A. Sarzynski
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC, United States of America
| | - Michael R. Zile
- Division of Cardiology, RHJ Department of Veterans Affairs Medical Center, Medical University of South Carolina, Charleston, SC, United States of America
| | - Tarek Shazly
- College of Engineering and Computing, University of South Carolina, Columbia, SC, United States of America
| | - Francis G. Spinale
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, United States of America
- Columbia VA Health Care System, Columbia, SC, United States of America
- College of Engineering and Computing, University of South Carolina, Columbia, SC, United States of America
- Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC, United States of America
| |
Collapse
|
16
|
Liu N, Su H, Lou Y, Kong J. The improvement of homocysteine-induced myocardial inflammation by vitamin D depends on activation of NFE2L2 mediated MTHFR. Int Immunopharmacol 2024; 127:111437. [PMID: 38150882 DOI: 10.1016/j.intimp.2023.111437] [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: 10/15/2023] [Revised: 12/14/2023] [Accepted: 12/20/2023] [Indexed: 12/29/2023]
Abstract
OBJECTIVES Myocardial inflammation underlies a broad spectrum of conditions that cause damage to the myocardium and lead to structural and functional defects. Homocysteine (Hcy) is closely related to the occurrence and development of cardiovascular diseases. We investigated the mechanism underlying the effects of vitamin D as a prophylactic treatment for Hcy-induced cardiac inflammation. METHODS The levels of 25(OH)D3 and Hcy were assessed using ELISA kits. Expression levels of the vitamin D receptor (VDR), NFE2 like bZIP transcription factor 2 (NFE2L2), methylenetetrahydrofolate reductase (MTHFR) and inflammatory factors were examined by Western blotting, immunohistochemistry and real time polymerase chain reaction. NFE2L2/MTHFR-knockdown HL-1 cells and NFE2L2+/- mouse were used to test the effects of vitamin D. RESULTS We found the levels of Hcy in the serum and myocardial tissue of mice in the Hcy + CCE group were lower than in the Hcy groups, which was opposed to the trend exhibited by the serum 25(OH)D3 level of mice. The mRNA and protein expression levels of the inflammatory factors in cardiac tissues and cardiomyocytes were strongly decreased by the Hcy treatment, compared to the Hcy + CCE/Hcy + 1,25(OH)2D3 groups. Moreover, the results revealed that the level of nuclear NFE2L2 in Hcy + CCE/Hcy + 1,25(OH)2D3 group was increased compared to Hcy group with a reciprocal decrease in the level of cytosolic NFE2L2 in vivo and in vitro. Similarly, the MTHFR mRNA and protein expression in the Hcy + CCE group was higher than the Hcy group. We determined that NFE2L2 promoted the expression of MTHFR. However, based on Hcy treatment, the combination of 1,25(OH)2D3 and MTHFR-/- reversed the decline in IL-6 and TNFα expression caused by 1,25(OH)2D3 alone. Chromatin immunoprecipitation and luciferase reporter assays showed the up-regulation effect of VDR on NFE2L2 and NFE2L2 on MTHFR. CONCLUSIONS Our findings indicate that vitamin D/VDR could improve Hcy-induced myocardial inflammation through activation of NFE2L2 mediated MTHFR.
Collapse
Affiliation(s)
- Ning Liu
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Han Su
- Department of Health Management, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yan Lou
- School of Fundamental Sciences, China Medical University, Shenyang 110122, China.
| | - Juan Kong
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
17
|
El-Awaisi J, Mitchell JL, Ranasinghe A, Kalia N. Interleukin-36 is vasculoprotective in both sexes despite sex-specific changes in the coronary microcirculation response to IR injury. Front Cardiovasc Med 2023; 10:1227499. [PMID: 37753164 PMCID: PMC10518412 DOI: 10.3389/fcvm.2023.1227499] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/25/2023] [Indexed: 09/28/2023] Open
Abstract
Aims Risks and outcomes of myocardial infarction (MI) are different between men and women and some studies have demonstrated that the latter have a higher risk of mortality. Whilst there are many reasons for this, it may also partially be linked to stronger innate and adaptive immune responses mounted by females compared to males. However, little is known about how sex impacts the coronary microvessels, the site where inflammatory processes take place, after an MI. Intravital and laser speckle microscopy was used to image coronary microvessels and ventricular perfusion in vivo in response to myocardial ischaemia-reperfusion (IR) injury in male and female mice. Interleukin-36 (IL-36) is the latest addition to the IL-1 superfamily of pro-inflammatory cytokines and has recently been shown to mediate inflammation in a number of non-cardiovascular diseases. Its role in mediating potential sex-related microcirculatiory pertubations in the heart are unknown. Therefore, the vasculoprotective efficacy of an IL-36 receptor antagonist (IL-36Ra) was also investigated. Methods and results Immunostaining and flow cytometry demonstrated higher expression of IL-36 and its receptor in female hearts, an observation confirmed in human samples. Intravital imaging of the anaesthetised mouse beating heart identified significantly greater neutrophil recruitment in female hearts, but a greater burden of thrombotic disease in male hearts. Male mice had reduced functional capillary density and were unable to restore perfusion to baseline values as effectively as females. However, female mice had significantly larger infarcts. Interestingly, IL-36Ra decreased inflammation, improved perfusion, and reduced infarct size in both sexes despite increasing platelet presence in male hearts. Mechanistically, this was explained by IL-36Ra attenuating endothelial oxidative damage and VCAM-1 expression. Importantly, IL-36Ra administration during ischaemia was critical for vasculoprotection to be realised. Conclusion This novel study identified notable sex-related differences in the coronary microcirculatory response to myocardial IR injury which may explain why some studies have noted poorer outcomes in women after MI. Whilst contemporary MI treatment focuses on anti-platelet strategies, the heightened presence of neutrophils in female IR injured coronary microvessels necessitates the development of an effective anti-inflammatory approach for treating female patients. We also emphasise the importance of early intervention during the ischaemic period in order to maximise therapeutic effectiveness.
Collapse
Affiliation(s)
- Juma El-Awaisi
- Microcirculation Research Group, Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Joanne L Mitchell
- Microcirculation Research Group, Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Aaron Ranasinghe
- Consultant Cardiac and Heart/Lung Transplant Consultant, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Trust, Birmingham, United Kingdom
| | - Neena Kalia
- Microcirculation Research Group, Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
18
|
Distefano R, Ilieva M, Madsen JH, Rennie S, Uchida S. DoxoDB: A Database for the Expression Analysis of Doxorubicin-Induced lncRNA Genes. Noncoding RNA 2023; 9:39. [PMID: 37489459 PMCID: PMC10366827 DOI: 10.3390/ncrna9040039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 07/26/2023] Open
Abstract
Cancer and cardiovascular disease are the leading causes of death worldwide. Recent evidence suggests that these two life-threatening diseases share several features in disease progression, such as angiogenesis, fibrosis, and immune responses. This has led to the emergence of a new field called cardio-oncology. Doxorubicin is a chemotherapy drug widely used to treat cancer, such as bladder and breast cancer. However, this drug causes serious side effects, including acute ventricular dysfunction, cardiomyopathy, and heart failure. Based on this evidence, we hypothesize that comparing the expression profiles of cells and tissues treated with doxorubicin may yield new insights into the adverse effects of the drug on cellular activities. To test this hypothesis, we analyzed published RNA sequencing (RNA-seq) data from doxorubicin-treated cells to identify commonly differentially expressed genes, including long non-coding RNAs (lncRNAs) as they are known to be dysregulated in diseased tissues and cells. From our systematic analysis, we identified several doxorubicin-induced genes. To confirm these findings, we treated human cardiac fibroblasts with doxorubicin to record expression changes in the selected doxorubicin-induced genes and performed a loss-of-function experiment of the lncRNA MAP3K4-AS1. To further disseminate the analyzed data, we built the web database DoxoDB.
Collapse
Affiliation(s)
- Rebecca Distefano
- Department of Biology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Mirolyuba Ilieva
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, DK-2450 Copenhagen SV, Denmark
| | - Jens Hedelund Madsen
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, DK-2450 Copenhagen SV, Denmark
| | - Sarah Rennie
- Department of Biology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Shizuka Uchida
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, DK-2450 Copenhagen SV, Denmark
| |
Collapse
|
19
|
Sidheeque Hassan V, Hanifa M, Navik U, Bali A. Exogenous fetuin-A protects against sepsis-induced myocardial injury by inhibiting oxidative stress and inflammation in mice. Fundam Clin Pharmacol 2023; 37:607-617. [PMID: 36647295 DOI: 10.1111/fcp.12870] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/23/2022] [Accepted: 01/09/2023] [Indexed: 01/18/2023]
Abstract
Sepsis-induced myocardial injury is a consequence of septicemia and is one of the major causes of death in intensive care units. A serum glycoprotein called fetuin-A is secreted largely by the liver, tongue, placenta, and adipose tissue. Fetuin-A has a variety of biological and pharmacological properties. The anti-inflammatory and antioxidant glycoprotein fetuin-A has shown its efficacy in a number of inflammatory disorders including sepsis. However, its protective role against sepsis-induced myocardial injury remains elusive. The purpose of this work is to explore the role of fetuin-A in mouse models of myocardial injury brought on by cecal ligation and puncture (CLP). CLP significantly induced the myocardial injury assessed in terms of elevated myocardial markers (serum CK-MB, cTnI levels), inflammatory markers (IL-6, TNF-α) in the serum, and oxidative stress markers (increased MDA levels and decreased reduced glutathione) in heart tissue homogenate following 24 h of ligation and puncture. Further, hematoxylin and eosin (H&E) staining showed considerable histological alterations in the myocardial tissue of sepsis-developed mice. Interestingly, fetuin-A pretreatment (50 and 100 mg/kg) for 4 days before the CLP procedure significantly improved the myocardial injury and was evaluated in perspective of a reduction in the CK-MB, cTnI levels, IL-6, and TNF-α in sepsis-developed animals. Fetuin-A pretreatment significantly attenuated the oxidative stress and improved the myocardial morphology in a dose-dependent manner. The present study provides preliminary evidence that fetuin-A exerts protection against sepsis-induced cardiac dysfunction in vivo via suppression of inflammation and oxidative damage.
Collapse
Affiliation(s)
- V Sidheeque Hassan
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Mohd Hanifa
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Anjana Bali
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| |
Collapse
|
20
|
Wang M, Mei K, Chao C, Di D, Qian Y, Wang B, Zhang X. Rheumatoid arthritis increases the risk of heart failure-current evidence from genome-wide association studies. Front Endocrinol (Lausanne) 2023; 14:1154271. [PMID: 37288294 PMCID: PMC10242133 DOI: 10.3389/fendo.2023.1154271] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 05/04/2023] [Indexed: 06/09/2023] Open
Abstract
Background Numerous studies have demonstrated that rheumatoid arthritis (RA) is related to increased incidence of heart failure (HF), but the underlying association remains unclear. In this study, the potential association of RA and HF was clarified using Mendelian randomization analysis. Methods Genetic tools for RA, HF, autoimmune disease (AD), and NT-proBNP were acquired from genome-wide studies without population overlap. The inverse variance weighting method was employed for MR analysis. Meanwhile, the results were verified in terms of reliability by using a series of analyses and assessments. Results According to MR analysis, its genetic susceptibility to RA may lead to increased risk of heart failure (OR=1.02226, 95%CI [1.005495-1.039304], P=0.009067), but RA was not associated with NT-proBNP. In addition, RA was a type of AD, and the genetic susceptibility of AD had a close relation to increased risk of heart failure (OR=1.045157, 95%CI [1.010249-1.081272], P=0.010825), while AD was not associated with NT-proBNP. In addition, the MR Steiger test revealed that RA was causal for HF and not the opposite (P = 0.000). Conclusion The causal role of RA in HF was explored to recognize the underlying mechanisms of RA and facilitate comprehensive HF evaluation and treatment of RA.
Collapse
Affiliation(s)
| | | | | | | | | | - Bin Wang
- *Correspondence: Bin Wang, ; Xiaoying Zhang,
| | | |
Collapse
|
21
|
Wang J, Song Y, Xie W, Zhao J, Wang Y, Yu W. Therapeutic angiogenesis based on injectable hydrogel for protein delivery in ischemic heart disease. iScience 2023; 26:106577. [PMID: 37192972 PMCID: PMC10182303 DOI: 10.1016/j.isci.2023.106577] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023] Open
Abstract
Ischemic heart disease (IHD) remains the leading cause of death and disability worldwide and leads to myocardial necrosis and negative myocardial remodeling, ultimately leading to heart failure. Current treatments include drug therapy, interventional therapy, and surgery. However, some patients with severe diffuse coronary artery disease, complex coronary artery anatomy, and other reasons are unsuitable for these treatments. Therapeutic angiogenesis stimulates the growth of the original blood vessels by using exogenous growth factors to generate more new blood vessels, which provides a new treatment for IHD. However, direct injection of these growth factors can cause a short half-life and serious side effects owing to systemic spread. Therefore, to overcome this problem, hydrogels have been developed for temporally and spatially controlled delivery of single or multiple growth factors to mimic the process of angiogenesis in vivo. This paper reviews the mechanism of angiogenesis, some important bioactive molecules, and natural and synthetic hydrogels currently being applied for bioactive molecule delivery to treat IHD. Furthermore, the current challenges of therapeutic angiogenesis in IHD and its potential solutions are discussed to facilitate real translation into clinical applications in the future.
Collapse
Affiliation(s)
- Junke Wang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 26000, China
- Qingdao Medical College, Qingdao University, Qingdao, Shandong 266071, China
| | - Yancheng Song
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 26000, China
| | - Wenjie Xie
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Shandong, Qingdao, Shandong 26000, China
| | - Jiang Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Ying Wang
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong 26000, China
- Corresponding author
| | - Wenzhou Yu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 26003, China
- Corresponding author
| |
Collapse
|
22
|
Zuo B, Zhu S, Wang G, Li Z. Transcriptome analysis reveals ADAMTS15 is a potential inflammation-related gene in remote ischemic postconditioning. Front Cardiovasc Med 2023; 10:1089151. [PMID: 37234367 PMCID: PMC10206167 DOI: 10.3389/fcvm.2023.1089151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 04/17/2023] [Indexed: 05/27/2023] Open
Abstract
Background Remote ischemic postconditioning (RIPostC) induced by brief episodes of the limb ischemia is a potential therapeutic strategy for myocardial ischemia/reperfusion injury, achieved by reducing cardiomyocyte death, inflammation and so on. The actual mechanisms underlying cardioprotection conferred by RIPostC remain unclear. Exploring gene expression profiles in myocardium at transcriptional level is helpful to deepen the understanding on the cardioprotective mechanisms of RIPostC. This study aims to investigate the effect of RIPostC on gene expressions in rat myocardium using transcriptome sequencing. Methods Rat myocardium samples from the RIPostC group, the control group (myocardial ischemia/reperfusion group) and the sham group were performed transcriptome analysis using RNA sequencing. The levels of cardiac IL-1β, IL-6, IL-10 and TNFα were analyzed by Elisa. The expression levels of candidate genes were verified by qRT-PCR technique. Infarct size was measured by Evans blue and TTC staining. Apoptosis was assessed by TUNEL assays and caspase-3 levels were detected using western blotting. Results RIPostC can markedly decrease infarct size and reduce the levels of cardiac IL-1β, IL-6 and increase the level of cardiac IL-10. This transcriptome analysis showed that 2 genes were up-regulated (Prodh1 and ADAMTS15) and 5 genes (Caspase-6, Claudin-5, Sccpdh, Robo4 and AABR07011951.1) were down-regulated in the RIPostC group. Go annotation analysis showed that Go terms mainly included cellular process, metabolic process, cell part, organelle, catalytic activity and binding. The KEGG annotation analysis of DEGs found only one pathway, amino acid metabolism, was up-regulated. The relative mRNA expression levels of ADAMTS15, Caspase-6, Claudin-5 and Prodh1 were verified by qRT-PCR, which were consistent with the RNA-seq results. In addition, the relative expression of ADAMTS15 was negatively correlated with the level of cardiac IL-1β (r = -0.748, P = 0.005) and positively correlated with the level of cardiac IL-10 (r = 0.698, P = 0.012). A negative correlation statistical trend was found between the relative expression of ADAMTS15 and the level of cardiac IL-6 (r = -0.545, P = 0.067). Conclusions ADAMTS15 may be a potential inflammation-related gene in regulation of cardioprotection conferred by remote ischemic postconditioning and a possible therapeutic target for myocardial ischemia reperfusion injury in the future.
Collapse
Affiliation(s)
- Bo Zuo
- Department of Cardiology, Cardiovascular Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Department of Cardiology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Sha Zhu
- Department of Neurology, Peking University International Hospital, Beijing, China
| | - Guisong Wang
- Department of Cardiology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Zhengpeng Li
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| |
Collapse
|
23
|
Li J, Liu W, Peng F, Cao X, Xie X, Peng C. The multifaceted biology of lncR-Meg3 in cardio-cerebrovascular diseases. Front Genet 2023; 14:1132884. [PMID: 36968595 PMCID: PMC10036404 DOI: 10.3389/fgene.2023.1132884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/28/2023] [Indexed: 03/12/2023] Open
Abstract
Cardio-cerebrovascular disease, related to high mortality and morbidity worldwide, is a type of cardiovascular or cerebrovascular dysfunction involved in various processes. Therefore, it is imperative to conduct additional research into the pathogenesis and new therapeutic targets of cardiovascular and cerebrovascular disorders. Long non-coding RNAs (lncRNAs) have multiple functions and are involved in nearly all cellular biological processes, including translation, transcription, signal transduction, and cell cycle control. LncR-Meg3 is one of them and is becoming increasingly popular. By binding proteins or directly or competitively binding miRNAs, LncR-Meg3 is involved in apoptosis, inflammation, oxidative stress, endoplasmic reticulum stress, epithelial-mesenchymal transition, and other processes. Recent research has shown that LncR-Meg3 is associated with acute myocardial infarction and can be used to diagnose this condition. This article examines the current state of knowledge regarding the expression and regulatory function of LncR-Meg3 in relation to cardiovascular and cerebrovascular diseases. The abnormal expression of LncR-Meg3 can influence neuronal cell death, inflammation, apoptosis, smooth muscle cell proliferation, etc., thereby aggravating or promoting the disease. In addition, we review the bioactive components that target lncR-Meg3 and propose some potential delivery vectors. A comprehensive and in-depth analysis of LncR-Meg3’s role in cardiovascular disease suggests that targeting LncR-Meg3 may be an alternative therapy in the near future, providing new options for slowing the progression of cardiovascular disease.
Collapse
Affiliation(s)
- Jing Li
- Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of standardization of Chinese herbal medicine of MOE, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenxiu Liu
- Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of standardization of Chinese herbal medicine of MOE, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fu Peng
- Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of standardization of Chinese herbal medicine of MOE, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
- *Correspondence: Fu Peng, ; Xiaofang Xie, ; Cheng Peng,
| | - Xiaoyu Cao
- Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of standardization of Chinese herbal medicine of MOE, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofang Xie
- Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of standardization of Chinese herbal medicine of MOE, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Fu Peng, ; Xiaofang Xie, ; Cheng Peng,
| | - Cheng Peng
- Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of standardization of Chinese herbal medicine of MOE, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Fu Peng, ; Xiaofang Xie, ; Cheng Peng,
| |
Collapse
|
24
|
Detection of myocardial dysfunction using global longitudinal strain with speckle-tracking echocardiography in patients with vs without rheumatoid arthritis: a systematic review and meta-analysis. J Echocardiogr 2023; 21:23-32. [PMID: 35987937 DOI: 10.1007/s12574-022-00583-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 07/04/2022] [Accepted: 07/19/2022] [Indexed: 10/15/2022]
Abstract
OBJECTIVES Rheumatoid arthritis (RA) is a systemic autoimmune disorder primarily involving the peripheral joints. Systemic involvement can occur, including myocardial dysfunction. Speckle tracking echocardiography (STE) is a novel diagnostic study which is recently being used to detect subclinical cardiac dysfunction. Global longitudinal strain (GLS) by STE is more sensitive than standard echocardiographic parameters to detect occult cardiac dysfunction. METHODS A systematic search of PUBMED, EMBASE, Cochrane, and Google Scholar databases was performed to identify studies comparing the STE parameters between RA and non-RA patients. RESULTS Left ventricular (LV) GLS was significantly lower in patients with RA compared to non-RA patients with a standard mean difference (SMD) of -1.09 (-1.48--0.70, P < 0.001). LV Global Circumferential Strain (GCS) was reported in five studies, and it was found to be lower in RA patients with an SMD of -1.25 (-2.59--0.10; P < 0.0010). Meta regression analysis studies failed to show any significant impact of disease duration, activity, age, sex and BMI on LV GLS and RV GLS. CONCLUSIONS RA patients have lower LV GLS and LV GCS compared to controls suggesting impaired myocardial dysfunction. Further studies need to be done to delineate the importance of lower GLS in asymptomatic rheumatoid patients to guide disease management and risk factor modification in this selected population.
Collapse
|
25
|
De Alba-Alvarado MC, Torres-Gutiérrez E, Reynoso-Ducoing OA, Zenteno-Galindo E, Cabrera-Bravo M, Guevara-Gómez Y, Salazar-Schettino PM, Rivera-Fernández N, Bucio-Torres MI. Immunopathological Mechanisms Underlying Cardiac Damage in Chagas Disease. Pathogens 2023; 12:pathogens12020335. [PMID: 36839607 PMCID: PMC9959418 DOI: 10.3390/pathogens12020335] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
In Chagas disease, the mechanisms involved in cardiac damage are an active field of study. The factors underlying the evolution of lesions following infection by Trypanosoma cruzi and, in some cases, the persistence of its antigens and the host response, with the ensuing development of clinically observable cardiac damage, are analyzed in this review.
Collapse
Affiliation(s)
- Mariana Citlalli De Alba-Alvarado
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán, México City 04510, Mexico
| | - Elia Torres-Gutiérrez
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán, México City 04510, Mexico
| | - Olivia Alicia Reynoso-Ducoing
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán, México City 04510, Mexico
| | - Edgar Zenteno-Galindo
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán, México City 04510, Mexico
| | - Margarita Cabrera-Bravo
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán, México City 04510, Mexico
| | - Yolanda Guevara-Gómez
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán, México City 04510, Mexico
| | - Paz María Salazar-Schettino
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán, México City 04510, Mexico
| | - Norma Rivera-Fernández
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán, México City 04510, Mexico
- Correspondence: (N.R.-F.); (M.I.B-T.)
| | - Martha Irene Bucio-Torres
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán, México City 04510, Mexico
- Correspondence: (N.R.-F.); (M.I.B-T.)
| |
Collapse
|
26
|
Patil P, Doshi G. Deciphering the Role of Pyroptosis Impact on Cardiovascular Diseases. Curr Drug Targets 2023; 24:1166-1183. [PMID: 38164730 DOI: 10.2174/0113894501267496231102114410] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 01/03/2024]
Abstract
Pyroptosis has become a noteworthy area of focus in recent years due to its association with inflammatory diseases. Pyroptosis is a type of programmed cell death accompanied by an inflammatory response, and the discovery of the gasdermin family has expanded the study of pyroptosis. The primary characteristics of pyroptosis include cell expansion, membrane penetration, and the ejection of cell contents. In healthy physiology, pyroptosis is an essential part of the host's defence against pathogen infection. Excessive Pyroptosis, however, can lead to unchecked and persistent inflammatory responses, including the emergence of inflammatory diseases. More precisely, gasdermin family members have a role in the creation of membrane holes during pyroptosis, which leads to cell lysis. It is also related to how pro-inflammatory intracellular substances, including IL-1, IL-18, and High mobility group box 1 (HMGB1), are used. Two different signalling pathways, one of which is regulated by caspase-1 and the other by caspase-4/5/11, are the primary causes of pyroptosis. Cardiovascular diseases are often associated with cell death and acute or chronic inflammation, making this area of research particularly relevant. In this review, we first systematically summarize recent findings related to Pyroptosis, exploring its characteristics and the signalling pathway mechanisms, as well as various treatment strategies based on its modulation that has emerged from the studies. Some of these strategies are currently undergoing clinical trials. Additionally, the article elaborates on the scientific evidence indicating the role of Pyroptosis in various cardiovascular diseases. As a whole, this should shed insight into future paths and present innovative ideas for employing Pyroptosis as a strong disease-fighting weapon.
Collapse
Affiliation(s)
- Poonam Patil
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, VLM Road, Vile Parle (w), Mumbai, 400056, India
| | - Gaurav Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, VLM Road, Vile Parle (w), Mumbai, 400056, India
| |
Collapse
|
27
|
Wang X, Zhou H, Liu Q, Cheng P, Zhao T, Yang T, Zhao Y, Sha W, Zhao Y, Qu H. Targeting regulatory T cells for cardiovascular diseases. Front Immunol 2023; 14:1126761. [PMID: 36911741 PMCID: PMC9995594 DOI: 10.3389/fimmu.2023.1126761] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death and disability worldwide. The CVDs are accompanied by inflammatory progression, resulting in innate and adaptive immune responses. Regulatory T cells (Tregs) have an immunosuppressive function and are one of the subsets of CD4+T cells that play a crucial role in inflammatory diseases. Whether using Tregs as a biomarker for CVDs or targeting Tregs to exert cardioprotective functions by regulating immune balance, suppressing inflammation, suppressing cardiac and vascular remodeling, mediating immune tolerance, and promoting cardiac regeneration in the treatment of CVDs has become an emerging research focus. However, Tregs have plasticity, and this plastic Tregs lose immunosuppressive function and produce toxic effects on target organs in some diseases. This review aims to provide an overview of Tregs' role and related mechanisms in CVDs, and reports on the research of plasticity Tregs in CVDs, to lay a foundation for further studies targeting Tregs in the prevention and treatment of CVDs.
Collapse
Affiliation(s)
- Xinting Wang
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Zhou
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Cardiovascular Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qian Liu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Peipei Cheng
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tingyao Zhao
- Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tianshu Yang
- Department of Cardiovascular Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yue Zhao
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wanjing Sha
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanyan Zhao
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huiyan Qu
- Department of Cardiovascular Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
28
|
ElKhatib MAW, Isse FA, El-Kadi AOS. Effect of inflammation on cytochrome P450-mediated arachidonic acid metabolism and the consequences on cardiac hypertrophy. Drug Metab Rev 2022; 55:50-74. [PMID: 36573379 DOI: 10.1080/03602532.2022.2162075] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The incidence of heart failure (HF) is generally preceded by cardiac hypertrophy (CH), which is the enlargement of cardiac myocytes in response to stress. During CH, the metabolism of arachidonic acid (AA), which is present in the cell membrane phospholipids, is modulated. Metabolism of AA gives rise to hydroxyeicosatetraenoic acids (HETEs) and epoxyeicosatrienoic acids (EETs) via cytochrome P450 (CYP) ω-hydroxylases and CYP epoxygenases, respectively. A plethora of studies demonstrated the involvement of CYP-mediated AA metabolites in the pathogenesis of CH. Also, inflammation is known to be a characteristic hallmark of CH. In this review, our aim is to highlight the impact of inflammation on CYP-derived AA metabolites and CH. Inflammation is shown to modulate the expression of various CYP ω-hydroxylases and CYP epoxygenases and their respective metabolites in the heart. In general, HETEs such as 20-HETE and mid-chain HETEs are pro-inflammatory, while EETs are characterized by their anti-inflammatory and cardioprotective properties. Several mechanisms are implicated in inflammation-induced CH, including the modulation of NF-κB and MAPK. This review demonstrated the inflammatory modulation of cardiac CYPs and their metabolites in the context of CH and the anti-inflammatory strategies that can be employed in the treatment of CH and HF.
Collapse
Affiliation(s)
| | - Fadumo Ahmed Isse
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | | |
Collapse
|
29
|
Zhang J, Hu Y, Wang H, Hou J, Xiao W, Wen X, Wang T, Long P, Jiang H, Wang Z, Liu H, Chen X. Advances in research on the protective mechanisms of traditional Chinese medicine (TCM) in myocardial ischaemia-reperfusion injury. PHARMACEUTICAL BIOLOGY 2022; 60:931-948. [PMID: 35587352 PMCID: PMC9132412 DOI: 10.1080/13880209.2022.2063342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/31/2022] [Accepted: 03/31/2022] [Indexed: 06/15/2023]
Abstract
CONTEXT Developing effective drugs to treat myocardial ischaemia-reperfusion (MI/R) injury is imperative. Traditional Chinese medicines (TCMs) have had considerable success in the treatment of cardiovascular diseases. Elucidating the mechanisms by which TCMs improve MI/R injury can supplement the literature on MI/R prevention and treatment. OBJECTIVE To summarise TCMs and their main protective mechanisms against MI/R injury reported over the past 40 years. METHODS Relevant literature published between 1980 and 2020 in Chinese and English was retrieved from the Web of Science, PubMed, SpringerLink, PubMed Central, Scopus, and Chinese National Knowledge Infrastructure (CNKI) databases. Search terms included 'medicinal plants', 'myocardial ischaemia reperfusion injury', 'Chinese medicine prescriptions', 'mechanisms', 'prevention', 'treatment' and 'protection'. For inclusion in the analysis, medicinal plants had to be searchable in the China Medical Information Platform and Plant Database. RESULTS We found 71 medicinal species (from 40 families) that have been used to prevent MI/R injury, of which Compositae species (8 species) and Leguminosae species (7 species) made up the majority. Most of the effects associated with these plants are described as antioxidant and anti-inflammatory. Furthermore, we summarised 18 kinds of Chinese compound prescriptions, including the compound Danshen tablet and Baoxin pill, which mainly reduce oxidative stress and regulate mitochondrial energy metabolism. DISCUSSION AND CONCLUSIONS We summarised TCMs that protect against MI/R injury and their pharmacological mechanisms. This in-depth explanation of the roles of TCMs in MI/R injury protection provides a theoretical basis for the research and development of TCM-based treatment drugs.
Collapse
Affiliation(s)
- Jiexin Zhang
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu/Affiliated Hospital of Southwest, Jiaotong University, Chengdu, Sichuan, China
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Yonghe Hu
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Han Wang
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu/Affiliated Hospital of Southwest, Jiaotong University, Chengdu, Sichuan, China
| | - Jun Hou
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Wenjing Xiao
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Xudong Wen
- Department of Gastroenterology, The First People’s Hospital of Chengdu, Chengdu, Sichuan, China
| | - Tingting Wang
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Pan Long
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Hezhong Jiang
- Faculty of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Zhanhao Wang
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu/Affiliated Hospital of Southwest, Jiaotong University, Chengdu, Sichuan, China
| | - Huawei Liu
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu/Affiliated Hospital of Southwest, Jiaotong University, Chengdu, Sichuan, China
| | - Xin Chen
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu/Affiliated Hospital of Southwest, Jiaotong University, Chengdu, Sichuan, China
| |
Collapse
|
30
|
Luo Y, Zhang H, Yu J, Wei L, Li M, Xu W. Stem cell factor/mast cell/CCL2/monocyte/macrophage axis promotes Coxsackievirus B3 myocarditis and cardiac fibrosis by increasing Ly6C high monocyte influx and fibrogenic mediators production. Immunology 2022; 167:590-605. [PMID: 36054617 DOI: 10.1111/imm.13556] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 02/10/2022] [Indexed: 06/15/2023] Open
Abstract
Mast cells (MCs), central players in allergy and parasitic infections, play key roles in inflammation and fibrosis. Here, the impact of MCs on the progression of Coxsackievirus B3 (CVB3)-induced viral myocarditis (VMC) and fibrosis was investigated using MC-deficient KitW-sh mice. Viral titres, cellular infiltrates and heart pathologies were evaluated and compared with wild-type (WT) mice during acute CVB3 infection of C57BL/6 mice. CVB3 infection induced an increased accumulation and degranulation of MCs in the hearts of mice during acute infection. MC-deficient KitW-sh mice had slightly higher viral titres, decreased VMC and cardiac fibrosis and improved cardiac dysfunction compared to WT mice via decreasing cardiac influx of Ly6Chigh monocytes/macrophages (Mo/Mφ). While bone marrow-derived MC reconstitution decreased viral titre and worsened improved survival and VMC severity in Wsh mice. MC-fibroblasts co-culture revealed a cardiac MC-fibroblasts crosstalk during early infection: fibroblasts trigger MC degranulation and secretion of CCL2 and tumour necrosis factor alpha (TNF-α) via producing early stem cell factor (SCF); while MCs-fibrogenic mediators (TNF-α) stimulate fibroblasts to increase CCL2, α-smooth muscle actin (SMA), collagen and transforming growth factor beta(TGFβ) expression, thus aggravating cardiac fibrosis. MCs and fibroblast-derived CCL2s are both essential for cardiac Ly6Chigh Mo/Mφ influx. Administration of recombinant mouse SCF to CVB3-infected mice aggravates VMC via accelerating MCs accumulation and cardiac influx of Ly6Chi Mo/Mφ. Collectively, our data highlight an early MC-fibroblast crosstalk and SCF/MC/CCL2/Mo/Mφ axis as important mechanisms required for triggering VMC and myocardial fibrosis. This finding indicates critical roles of MCs in initiating and modulating cardiac innate response to CVB3 and has an implication in developing new and more effective treatments for VMC.
Collapse
Affiliation(s)
- Yuan Luo
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Hongkai Zhang
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Jie Yu
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Lin Wei
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Min Li
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Wei Xu
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
31
|
Ling S, You Z, Li Y, Zhang J, Zhao S, He Y, Chen X. The role of γδ T17 cells in cardiovascular disease. J Leukoc Biol 2022; 112:1649-1661. [PMID: 36073777 DOI: 10.1002/jlb.3mr0822-761rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 06/23/2022] [Accepted: 08/03/2022] [Indexed: 01/04/2023] Open
Abstract
Due to the ability of γδ T cells to bridge adaptive and innate immunity, γδ T cells can respond to a variety of molecular cues and acquire the ability to induce a variety of cytokines such as IL-17 family, IFN-γ, IL-4, and IL-10. IL-17+ γδ T cells (γδ T17 cells) populations have recently received considerable interest as they are the major early source of IL-17A in many immune response models. However, the exact mechanism of γδ T17 cells is still poorly understood, especially in the context of cardiovascular disease (CVD). CVD is the leading cause of death in the world, and it tends to be younger. Here, we offer a review of the cardiovascular inflammatory and immune functions of γδ T17 cells in order to understand their role in CVD, which may be the key to developing new clinical applications.
Collapse
Affiliation(s)
- Shaoxue Ling
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| | - Zonghao You
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| | - Yang Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| | - Jian Zhang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| | - Shuwu Zhao
- School of Intergrative Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| | - Yongzhi He
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| | - Xi Chen
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| |
Collapse
|
32
|
Kell DB, Pretorius E. The potential role of ischaemia-reperfusion injury in chronic, relapsing diseases such as rheumatoid arthritis, Long COVID, and ME/CFS: evidence, mechanisms, and therapeutic implications. Biochem J 2022; 479:1653-1708. [PMID: 36043493 PMCID: PMC9484810 DOI: 10.1042/bcj20220154] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 02/07/2023]
Abstract
Ischaemia-reperfusion (I-R) injury, initiated via bursts of reactive oxygen species produced during the reoxygenation phase following hypoxia, is well known in a variety of acute circumstances. We argue here that I-R injury also underpins elements of the pathology of a variety of chronic, inflammatory diseases, including rheumatoid arthritis, ME/CFS and, our chief focus and most proximally, Long COVID. Ischaemia may be initiated via fibrin amyloid microclot blockage of capillaries, for instance as exercise is started; reperfusion is a necessary corollary when it finishes. We rehearse the mechanistic evidence for these occurrences here, in terms of their manifestation as oxidative stress, hyperinflammation, mast cell activation, the production of marker metabolites and related activities. Such microclot-based phenomena can explain both the breathlessness/fatigue and the post-exertional malaise that may be observed in these conditions, as well as many other observables. The recognition of these processes implies, mechanistically, that therapeutic benefit is potentially to be had from antioxidants, from anti-inflammatories, from iron chelators, and via suitable, safe fibrinolytics, and/or anti-clotting agents. We review the considerable existing evidence that is consistent with this, and with the biochemical mechanisms involved.
Collapse
Affiliation(s)
- Douglas B. Kell
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Kemitorvet 200, 2800 Kgs Lyngby, Denmark
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland 7602, South Africa
| | - Etheresia Pretorius
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland 7602, South Africa
| |
Collapse
|
33
|
Long K, Zhao Z, Chen J, Zhi L, Wang C, Liao D, Wang M, Gao P. Yang-xin-xue keli exerts therapeutic effects via regulating mitochondrial homeostasis and function in doxorubicin-induced rat heart failure. Front Pharmacol 2022; 13:931453. [PMID: 36110548 PMCID: PMC9468485 DOI: 10.3389/fphar.2022.931453] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Heart failure, especially chronic heart failure, is generally induced by the accumulation of reactive oxygen species (ROS), as well as the subsequent loss of mitochondrial permeability transition pore (mPTP) openings and pathological mitochondrial dysfunction. Herein, we explored the therapeutic effects of the Chinese medicine Yangxin Keli (YXXKL) on chronic heart failure and its underlying working mechanism. Methods: To mimic oxidative stress-induced chronic heart failure, a rat heart failure model was induced by the administration of DOX. Transthoracic echocardiography was performed to confirm the successful establishment of the heart failure model by observing significantly decreased cardiac function in the rats. Mitochondrial membrane potential, function, and ATP synthesis activity were measured after YXXKL was employed. Results The administration of YXXKL not only significantly improved cardiac function but also reversed the myocardium loss and fibrosis induced via DOX. Moreover, the administration of YXXKL also increased ATP synthesis and mitochondrial DNA mass in left ventricular tissues, which indicated that mitochondria may be a key target of YXXKL. Thus, we employed rat cardiomyocyte H9c2 and primary rat cardiac myocytes (RCMs) to induce oxidative stress-induced myocardial injury via DOX treatment. YXXKL-medicated serum promoted cell proliferation, which was inhibited by the addition of IC30 DOX, and the serum also inhibited cell apoptosis, which was promoted by the addition of IC50 DOX. YXKL-medicated serum was able to scavenge ROS and maintain the mitochondrial membrane potential as well as promote mitochondrial function, including the promotion of ATP synthesis, mitochondrial DNA mass, and transcriptional activity. Furthermore, we also observed that YXXKL-medicated serum inhibited DOX-induced autophagy/mitophagy by scavenging ROS. Conclusion: Taken together, we conclude that YXXKLI may exert therapeutic effects on oxidative stress-related heart failure via the regulation of mitochondria.
Collapse
Affiliation(s)
- Kunlan Long
- Intensive Care Unit, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ziyi Zhao
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Chen
- Intensive Care Unit, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lijia Zhi
- Intensive Care Unit, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chunxia Wang
- Intensive Care Unit, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dan Liao
- Intensive Care Unit, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Meng Wang
- Intensive Care Unit, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peiyang Gao
- Intensive Care Unit, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Peiyang Gao,
| |
Collapse
|
34
|
Wan Q, Xu C, Zhu L, Zhang Y, Peng Z, Chen H, Rao H, Zhang E, Wang H, Chu F, Ning X, Yang X, Yuan J, Wu Y, Huang Y, Hu S, Liu DP, Wang M. Targeting PDE4B (Phosphodiesterase-4 Subtype B) for Cardioprotection in Acute Myocardial Infarction via Neutrophils and Microcirculation. Circ Res 2022; 131:442-455. [PMID: 35899614 DOI: 10.1161/circresaha.122.321365] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Timely and complete restoration of blood flow is the most effective intervention for patients with acute myocardial infarction. However, the efficacy is limited by myocardial ischemia-reperfusion (MI/R) injury. PDE4 (phosphodiesterase-4) hydrolyzes intracellular cAMP and it has 4 subtypes A-D. This study aimed to delineate the role of PDE4B (phosphodiesterase-4 subtype B) in MI/R injury. METHODS Mice were subjected to 30-minute coronary artery ligation, followed by 24-hour reperfusion. Cardiac perfusion was assessed by laser Doppler flow. Vasomotor reactivities were determined in mouse and human coronary (micro-)arteries. RESULTS Cardiac expression of PDE4B, but not other PDE4 subtypes, was increased in mice following reperfusion. PDE4B was detected primarily in endothelial and myeloid cells of mouse and human hearts. PDE4B deletion strikingly reduced infarct size and improved cardiac function 24-hour or 28-day after MI/R. PDE4B in bone marrow-derived cells promoted MI/R injury and vascular PDE4B further exaggerated this injury. Mechanistically, PDE4B-mediated neutrophil-endothelial cell interaction and PKA (protein kinase A)-dependent expression of cell adhesion molecules, neutrophil cardiac infiltration, and release of proinflammatory cytokines. Meanwhile, PDE4B promoted coronary microcirculatory obstruction and vascular permeability in MI/R, without affecting flow restriction-induced thrombosis. PDE4B blockade increased flow-mediated vasodilatation and promoted endothelium-dependent dilatation of coronary arteries in a PKA- and nitric oxide-dependent manner. Furthermore, postischemia administration with piclamilast, a PDE4 pan-inhibitor, improved cardiac microcirculation, suppressed inflammation, and attenuated MI/R injury in mice. Incubation with sera from patients with acute myocardial infarction impaired acetylcholine-induced relaxations in human coronary microarteries, which was abolished by PDE4 inhibition. Similar protection against MI/R-related coronary injury was recapitulated in mice with PDE4B deletion or inhibition, but not with the pure vasodilator, sodium nitroprusside. CONCLUSIONS PDE4B is critically involved in neutrophil inflammation and microvascular obstruction, leading to MI/R injury. Selective inhibition of PDE4B might protect cardiac function in patients with acute myocardial infarction designated for reperfusion therapy.
Collapse
Affiliation(s)
- Qing Wan
- State Key Laboratory of Cardiovascular Disease (Q.W., C.X., L.Z., Y.Z., Z.P., H.C., H.R., F.C., X.N., X.Y., S.H., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chuansheng Xu
- State Key Laboratory of Cardiovascular Disease (Q.W., C.X., L.Z., Y.Z., Z.P., H.C., H.R., F.C., X.N., X.Y., S.H., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liyuan Zhu
- State Key Laboratory of Cardiovascular Disease (Q.W., C.X., L.Z., Y.Z., Z.P., H.C., H.R., F.C., X.N., X.Y., S.H., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuze Zhang
- State Key Laboratory of Cardiovascular Disease (Q.W., C.X., L.Z., Y.Z., Z.P., H.C., H.R., F.C., X.N., X.Y., S.H., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zekun Peng
- State Key Laboratory of Cardiovascular Disease (Q.W., C.X., L.Z., Y.Z., Z.P., H.C., H.R., F.C., X.N., X.Y., S.H., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hong Chen
- State Key Laboratory of Cardiovascular Disease (Q.W., C.X., L.Z., Y.Z., Z.P., H.C., H.R., F.C., X.N., X.Y., S.H., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haojie Rao
- State Key Laboratory of Cardiovascular Disease (Q.W., C.X., L.Z., Y.Z., Z.P., H.C., H.R., F.C., X.N., X.Y., S.H., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Erli Zhang
- Department of Cardiology (E.Z., J.Y., Y.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongyue Wang
- Department of Pathology (H.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Chu
- State Key Laboratory of Cardiovascular Disease (Q.W., C.X., L.Z., Y.Z., Z.P., H.C., H.R., F.C., X.N., X.Y., S.H., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Pharmacy, First Affiliated Hospital, Bengbu Medical College, Anhui, China (F.C.)
| | - Xuan Ning
- State Key Laboratory of Cardiovascular Disease (Q.W., C.X., L.Z., Y.Z., Z.P., H.C., H.R., F.C., X.N., X.Y., S.H., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuejian Yang
- State Key Laboratory of Cardiovascular Disease (Q.W., C.X., L.Z., Y.Z., Z.P., H.C., H.R., F.C., X.N., X.Y., S.H., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinqing Yuan
- Department of Cardiology (E.Z., J.Y., Y.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongjian Wu
- Department of Cardiology (E.Z., J.Y., Y.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Huang
- Department of Biomedical Sciences, The City University of Hong Kong, Hong Kong SAR, China (Y.H.)
| | - Shengshou Hu
- State Key Laboratory of Cardiovascular Disease (Q.W., C.X., L.Z., Y.Z., Z.P., H.C., H.R., F.C., X.N., X.Y., S.H., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Cardiovascular Surgery (S.H.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - De-Pei Liu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (D.-P.L.)
| | - Miao Wang
- State Key Laboratory of Cardiovascular Disease (Q.W., C.X., L.Z., Y.Z., Z.P., H.C., H.R., F.C., X.N., X.Y., S.H., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Clinical Pharmacology Center (M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
35
|
Liang R, Chen S, Jin Y, Tao L, Ji W, Zhu P, Li D, Zhang Y, Zhang W, Duan G. The CXCL10/CXCR3 Axis Promotes Disease Pathogenesis in Mice upon CVA2 Infection. Microbiol Spectr 2022; 10:e0230721. [PMID: 35604176 PMCID: PMC9241849 DOI: 10.1128/spectrum.02307-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 04/16/2022] [Indexed: 11/20/2022] Open
Abstract
Coxsackievirus A2 (CVA2) is an emerging pathogen that results in hand-foot-and-mouth disease (HFMD) outbreaks. Systemic inflammatory response and central nervous system inflammation are the main pathological features of fatal HFMD. However, the immunopathogenesis of CVA2 infection is poorly understood. We first detected the transcriptional levels of 81 inflammation-related genes in neonatal mice with CVA2 infection. Remarkably, CVA2 induced higher expression of chemokine (C-X-C motif) ligand 10 (CXCL10) in multiple organs and tissues. CXCL10 acts through its cognate receptor chemokine (C-X-C motif) receptor 3 (CXCR3) and regulates immune responses. CXCL10/CXCR3 activation contributes to the pathogenesis of many inflammatory diseases. Next, we found CXCL10 and CXCR3 expression to be significantly elevated in the organs and tissues from CVA2-infected mice at 5 days postinfection (dpi) using immunohistochemistry (IHC). To further explore the role of CXCL10/CXCR3 in CVA2 pathogenesis, an anti-CXCR3 neutralizing antibody (αCXCR3) or IgG isotype control antibody was used to treat CVA2-infected mice on the same day as infection and every 24 h until 5 dpi. Our results showed that αCXCR3 therapy relieved the clinical manifestations and pathological damage and improved the survival rate of CVA2-infected mice. Additionally, αCXCR3 treatment reduced viral loads and reversed the proinflammatory cytokine (interleukin 6 [IL-6], tumor necrosis factor alpha [TNF-α], and IL-1β) expression, apoptosis, and inflammatory cell infiltration induced by CVA2. Collectively, our study presents evidence for the involvement of the CXCL10/CXCR3 axis in CVA2 pathogenesis. The activation of CXCL10/CXCR3 contributes to CVA2 pathogenesis by inducing apoptosis, proinflammatory cytokine expression, and inflammatory cell infiltration, which can be reversed by αCXCR3 therapy. This study provides new insight into the pathogenesis of HFMD, which has an important guiding significance for the treatment of HFMD. IMPORTANCE Systemic inflammatory response and central nervous system inflammation are the main pathological features of fatal HFMD cases. We detected the expression of 81 inflammation-related genes and found higher expression of CXCL10 in CVA2-infected mice. Next, we confirmed CXCL10/CXCR3 activation using immunohistochemistry and found that anti-CXCR3 neutralizing antibody (αCXCR3) therapy could relieve the clinical manifestations and pathological damage and improve the survival rate of CVA2-infected mice. Additionally, αCXCR3 treatment reduced viral loads and reversed the proinflammatory cytokine (IL-6, TNF-α, and IL-1β) expression, apoptosis, and inflammatory cell infiltration induced by CVA2. Collectively, our study presents the first evidence for the involvement of the CXCL10/CXCR3 axis in CVA2 pathogenesis. The activation of CXCL10/CXCR3 contributes to CVA2 pathogenesis via inducing apoptosis, proinflammatory cytokine expression, and inflammatory cell infiltration, which can be reversed by αCXCR3 therapy. This study provides new insight into the pathogenesis of HFMD, which has an important guiding significance for the treatment of HFMD.
Collapse
Affiliation(s)
- Ruonan Liang
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Shuaiyin Chen
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yuefei Jin
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Ling Tao
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Wangquan Ji
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Peiyu Zhu
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Dong Li
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yu Zhang
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Weiguo Zhang
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
- Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
| | - Guangcai Duan
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Molecular Medicine, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
36
|
Wyatt L, Kamat G, Moyer J, Weaver AM, Diaz-Sanchez D, Devlin RB, Di Q, Schwartz JD, Cascio WE, Ward-Caviness CK. Associations between short-term exposure to PM 2.5 and cardiomyocyte injury in myocardial infarction survivors in North Carolina. Open Heart 2022; 9:openhrt-2021-001891. [PMID: 35750420 PMCID: PMC9234784 DOI: 10.1136/openhrt-2021-001891] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 05/23/2022] [Indexed: 12/20/2022] Open
Abstract
Objective Short-term ambient fine particulate matter (PM2.5) is associated with adverse cardiovascular events including myocardial infarction (MI). However, few studies have examined associations between PM2.5 and subclinical cardiomyocyte damage outside of overt cardiovascular events. Here we evaluate the impact of daily PM2.5 on cardiac troponin I, a cardiomyocyte specific biomarker of cellular damage. Methods We conducted a retrospective cohort study of 2924 patients identified using electronic health records from the University of North Carolina Healthcare System who had a recorded MI between 2004 and 2016. Troponin I measurements were available from 2014 to 2016, and were required to be at least 1 week away from a clinically diagnosed MI. Daily ambient PM2.5 concentrations were estimated at 1 km resolution and assigned to patient residence. Associations between log-transformed troponin I and daily PM2.5 were evaluated using distributed lag linear mixed effects models adjusted for patient demographics, socioeconomic status and meteorology. Results A 10 µg/m3 elevation in PM2.5 3 days before troponin I measurement was associated with 0.06 ng/mL higher troponin I (95% CI=0.004 to 0.12). In stratified models, this association was strongest in patients that were men, white and living in less urban areas. Similar associations were observed when using 2-day rolling averages and were consistently strongest when using the average exposure over the 5 days prior to troponin I measurement. Conclusions Daily elevations in PM2.5 were associated with damage to cardiomyocytes, outside of the occurrence of an MI. Poor air quality may cause persistent damage to the cardiovascular system leading to increased risk of cardiovascular disease and adverse cardiovascular events.
Collapse
Affiliation(s)
- Lauren Wyatt
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency Center for Public Health and Environmental Assessment, Research Triangle Park, North Carolina, USA
| | - Gauri Kamat
- Brown University, Providence, Rhode Island, USA
| | - Joshua Moyer
- US Environmental Protection Agency Center for Public Health and Environmental Assessment, Research Triangle Park, North Carolina, USA
| | - Anne M Weaver
- US Environmental Protection Agency Center for Public Health and Environmental Assessment, Research Triangle Park, North Carolina, USA
| | - David Diaz-Sanchez
- US Environmental Protection Agency Center for Public Health and Environmental Assessment, Research Triangle Park, North Carolina, USA
| | - Robert B Devlin
- US Environmental Protection Agency Center for Public Health and Environmental Assessment, Research Triangle Park, North Carolina, USA
| | - Qian Di
- Vanke School of Public Health, Tsinghua University, Beijing, China
| | - Joel D Schwartz
- Harvard T.H. Chan School of Public Health, Department of Environmental Health, Harvard University, Boston, Massachusetts, USA
| | - Wayne E Cascio
- US Environmental Protection Agency Center for Public Health and Environmental Assessment, Research Triangle Park, North Carolina, USA
| | - Cavin K Ward-Caviness
- US Environmental Protection Agency Center for Public Health and Environmental Assessment, Research Triangle Park, North Carolina, USA
| |
Collapse
|
37
|
Zhang MX, Song Y, Xu WL, Zhang LX, Li C, Li YL. Natural Herbal Medicine as a Treatment Strategy for Myocardial Infarction through the Regulation of Angiogenesis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:8831750. [PMID: 35600953 PMCID: PMC9119779 DOI: 10.1155/2022/8831750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/25/2022] [Indexed: 11/18/2022]
Abstract
Methods We conducted a literature search on the bioactive components of medicinal plants and their effects on angiogenesis after MI. We searched for articles in Web of Science, MEDLINE, PubMed, Scopus, Google Scholar, and China National Knowledge Infrastructure databases before April 2021. Results In this article, we summarized the mechanisms by which copper ions, microRNA, Akt1, inflammation, oxidative stress, mitochondria, and pericytes are involved in angiogenesis after myocardial infarction. In addition, we reviewed the angiogenic effects of natural herbal medicines such as Salvia miltiorrhiza Bunge Bunge, Carthamus tinctorius L., Pueraria lobata, Astragalus, Panax ginseng C.A. Mey., Panax notoginseng (Burkill) F.H. Chen, Cinnamomum cassia (L.) J. Presl, Rehmannia glutinosa (Gaertn.) DC., Leonurus japonicus Houtt, Scutellaria baicalensis Georgi., and Geum macrophyllum Willd. Conclusions Some herbs have the effect of promoting angiogenesis. In the future, natural proangiogenic drugs may become candidates for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Mu-xin Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yu Song
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Wan-li Xu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Ling-xiao Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yun-lun Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Department of Cardiology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| |
Collapse
|
38
|
Hao T, Qian M, Zhang Y, Liu Q, Midgley AC, Liu Y, Che Y, Hou J, Zhao Q. An Injectable Dual-Function Hydrogel Protects Against Myocardial Ischemia/Reperfusion Injury by Modulating ROS/NO Disequilibrium. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105408. [PMID: 35319828 PMCID: PMC9130918 DOI: 10.1002/advs.202105408] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/25/2022] [Indexed: 06/14/2023]
Abstract
Acute myocardial infarction (MI) is the leading cause of death worldwide. Exogenous delivery of nitric oxide (NO) to the infarcted myocardium has proven to be an effective strategy for treating MI due to the multiple physiological functions of NO. However, reperfusion of blood flow to the ischemic tissues is accompanied by the overproduction of toxic reactive oxygen species (ROS), which can further exacerbate tissue damage and compromise the therapeutic efficacy. Here, an injectable hydrogel is synthesized from the chitosan modified by boronate-protected diazeniumdiolate (CS-B-NO) that can release NO in response to ROS stimulation and thereby modulate ROS/NO disequilibrium after ischemia/reperfusion (I/R) injury. Furthermore, administration of CS-B-NO efficiently attenuated cardiac damage and adverse cardiac remodeling, promoted repair of the heart, and ameliorated cardiac function, unlike a hydrogel that only released NO, in a mouse model of myocardial I/R injury. Mechanistically, regulation of the ROS/NO balance activated the antioxidant defense system and protected against oxidative stress induced by I/R injury via adaptive regulation of the Nrf2-Keap1 pathway. Inflammation is then reduced by inhibition of the activation of NF-κB signaling. Collectively, these results show that this dual-function hydrogel may be a promising candidate for the protection of tissues and organs after I/R injury.
Collapse
Affiliation(s)
- Tian Hao
- State key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Sustainable Chemical TransformationsKey Laboratory of Bioactive Materials (Ministry of Education)Frontiers Science Center for Cell ResponsesCollege of Life SciencesNankai UniversityTianjin300071China
| | - Meng Qian
- State key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Sustainable Chemical TransformationsKey Laboratory of Bioactive Materials (Ministry of Education)Frontiers Science Center for Cell ResponsesCollege of Life SciencesNankai UniversityTianjin300071China
| | - Yating Zhang
- State key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Sustainable Chemical TransformationsKey Laboratory of Bioactive Materials (Ministry of Education)Frontiers Science Center for Cell ResponsesCollege of Life SciencesNankai UniversityTianjin300071China
| | - Qi Liu
- State key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Sustainable Chemical TransformationsKey Laboratory of Bioactive Materials (Ministry of Education)Frontiers Science Center for Cell ResponsesCollege of Life SciencesNankai UniversityTianjin300071China
| | - Adam C. Midgley
- State key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Sustainable Chemical TransformationsKey Laboratory of Bioactive Materials (Ministry of Education)Frontiers Science Center for Cell ResponsesCollege of Life SciencesNankai UniversityTianjin300071China
| | - Yangping Liu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and DiagnosticsSchool of PharmacyTianjin Medical UniversityTianjin300070China
| | - Yongzhe Che
- School of MedicineNankai UniversityTianjin300071China
| | - Jingli Hou
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and DiagnosticsSchool of PharmacyTianjin Medical UniversityTianjin300070China
| | - Qiang Zhao
- State key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Sustainable Chemical TransformationsKey Laboratory of Bioactive Materials (Ministry of Education)Frontiers Science Center for Cell ResponsesCollege of Life SciencesNankai UniversityTianjin300071China
| |
Collapse
|
39
|
Kwan AC, Salto G, Nguyen TT, Kim EH, Luong E, Hiremath P, Ouyang D, Ebinger JE, Li D, Berman DS, Kittleson MM, Kobashigawa JA, Patel JK, Cheng S. Cardiac microstructural alterations in immune-inflammatory myocardial disease: a retrospective case-control study. Cardiovasc Ultrasound 2022; 20:9. [PMID: 35369883 PMCID: PMC8978375 DOI: 10.1186/s12947-022-00279-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 03/28/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Immune-inflammatory myocardial disease contributes to multiple chronic cardiac processes, but access to non-invasive screening is limited. We have previously developed a method of echocardiographic texture analysis, called the high-spectrum signal intensity coefficient (HS-SIC) which assesses myocardial microstructure and previously associated with myocardial fibrosis. We aimed to determine whether this echocardiographic texture analysis of cardiac microstructure can identify inflammatory cardiac disease in the clinical setting. METHODS We conducted a retrospective case-control study of 318 patients with distinct clinical myocardial pathologies and 20 healthy controls. Populations included myocarditis, atypical chest pain/palpitations, STEMI, severe aortic stenosis, acute COVID infection, amyloidosis, and cardiac transplantation with acute rejection, without current rejection but with prior rejection, and with no history of rejection. We assessed the HS-SIC's ability to differentiate between a broader diversity of clinical groups and healthy controls. We used Kruskal-Wallis tests to compare HS-SIC values measured in each of the clinical populations with those in the healthy control group and compared HS-SIC values between the subgroups of cardiac transplantation rejection status. RESULTS For the total sample of N = 338, the mean age was 49.6 ± 20.9 years and 50% were women. The mean ± standard error of the mean of HS-SIC were: 0.668 ± 0.074 for controls, 0.552 ± 0.049 for atypical chest pain/palpitations, 0.425 ± 0.058 for myocarditis, 0.881 ± 0.129 for STEMI, 1.116 ± 0.196 for severe aortic stenosis, 0.904 ± 0.116 for acute COVID, and 0.698 ± 0.103 for amyloidosis. Among cardiac transplant recipients, HS-SIC values were 0.478 ± 0.999 for active rejection, 0.594 ± 0.091 for prior rejection, and 1.191 ± 0.442 for never rejection. We observed significant differences in HS-SIC between controls and myocarditis (P = 0.0014), active rejection (P = 0.0076), and atypical chest pain or palpitations (P = 0.0014); as well as between transplant patients with active rejection and those without current or prior rejection (P = 0.031). CONCLUSIONS An echocardiographic method can be used to characterize tissue signatures of microstructural changes across a spectrum of cardiac disease including immune-inflammatory conditions.
Collapse
Affiliation(s)
- Alan C. Kwan
- grid.50956.3f0000 0001 2152 9905Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA USA
| | - Gerran Salto
- grid.50956.3f0000 0001 2152 9905Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA USA ,grid.62560.370000 0004 0378 8294Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA USA ,grid.510954.c0000 0004 0444 3861Framingham Heart Study, Framingham, MA USA
| | - Trevor-Trung Nguyen
- grid.50956.3f0000 0001 2152 9905Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA USA
| | - Elizabeth H. Kim
- grid.50956.3f0000 0001 2152 9905Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA USA
| | - Eric Luong
- grid.50956.3f0000 0001 2152 9905Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA USA
| | - Pranoti Hiremath
- grid.411935.b0000 0001 2192 2723Division of Cardiology, Johns Hopkins Hospital, Baltimore, MD USA
| | - David Ouyang
- grid.50956.3f0000 0001 2152 9905Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA USA
| | - Joseph E. Ebinger
- grid.50956.3f0000 0001 2152 9905Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA USA
| | - Debiao Li
- grid.50956.3f0000 0001 2152 9905Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA USA
| | - Daniel S. Berman
- grid.50956.3f0000 0001 2152 9905Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA USA ,grid.50956.3f0000 0001 2152 9905Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, CA USA
| | - Michelle M. Kittleson
- grid.50956.3f0000 0001 2152 9905Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA USA
| | - Jon A. Kobashigawa
- grid.50956.3f0000 0001 2152 9905Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA USA
| | - Jignesh K. Patel
- grid.50956.3f0000 0001 2152 9905Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA USA
| | - Susan Cheng
- grid.50956.3f0000 0001 2152 9905Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA USA ,grid.62560.370000 0004 0378 8294Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA USA ,grid.510954.c0000 0004 0444 3861Framingham Heart Study, Framingham, MA USA
| |
Collapse
|
40
|
Shi H, Zhou ZM, Zhu L, Chen L, Jiang ZL, Wu XT. Underlying Mechanisms and Related Diseases Behind the Complex Regulatory Role of NOD-Like Receptor X1. DNA Cell Biol 2022; 41:469-478. [PMID: 35363060 DOI: 10.1089/dna.2022.0051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Among nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs), NOD-like receptor X1 (NLRX1) is the only known NLR family member that is targeted to the mitochondria, which contains a C-terminal leucine-rich repeat domain, a central conserved nucleotide-binding domain, and an unconventional N-terminal effector domain. It is unique due to several atypical features, such as mitochondrial localization, noninflammasome forming, and relatively undefined N-terminal domain. NLRX1 has multiple functions, including negative regulation of type-I interferon signaling, attenuation of proinflammatory nuclear factor kappa B (NF-κB) signaling, autophagy induction, modulation of reactive oxygen species production, cell death regulation, and participating in cellular senescence. In addition, due to its diverse functions, NLRX1 has been associated with various human diseases, including respiratory, circulatory, motor, urinary, nervous, and digestive systems, to name but a few. However, the exact regulatory mechanisms of NLRX1 are still unclear in many related diseases since conflicting and controversial topics on NLRX1 in the previous studies remain. In this review, we review recent research advances on the underlying mechanisms and related disorders behind the complex regulatory role of NLRX1, which may provide a promising target to prevent and/or treat the corresponding diseases.
Collapse
Affiliation(s)
- Hang Shi
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhi-Min Zhou
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Lei Zhu
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Lu Chen
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zan-Li Jiang
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xiao-Tao Wu
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
41
|
Yao Y, Li F, Zhang M, Jin L, Xie P, Liu D, Zhang J, Hu X, Lv F, Shang H, Zheng W, Sun X, Duanmu J, Wu F, Lan F, Xiao RP, Zhang Y. Targeting CaMKII-δ9 Ameliorates Cardiac Ischemia/Reperfusion Injury by Inhibiting Myocardial Inflammation. Circ Res 2022; 130:887-903. [PMID: 35152717 DOI: 10.1161/circresaha.121.319478] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 02/01/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND CaMKII (Ca2+/calmodulin-dependent kinase II) plays a central role in cardiac ischemia/reperfusion (I/R) injury-an important therapeutic target for ischemic heart disease. In the heart, CaMKII-δ is the predominant isoform and further alternatively spliced into 11 variants. In humans, CaMKII-δ9 and CaMKII-δ3, the major cardiac splice variants, inversely regulate cardiomyocyte viability with the former pro-death and the latter pro-survival. However, it is unknown whether specific inhibition of the detrimental CaMKII-δ9 prevents cardiac I/R injury and, if so, what is the underlying mechanism. Here, we aim to investigate the cardioprotective effect of specific CaMKII-δ9 inhibition against myocardial I/R damage and determine the underlying mechanisms. METHODS The role and mechanism of CaMKII-δ9 in cardiac I/R injury were investigated in mice in vivo, neonatal rat ventricular myocytes, and human embryonic stem cell-derived cardiomyocytes. RESULTS We demonstrate that CaMKII-δ9 inhibition with knockdown or knockout of its feature exon, exon 16, protects the heart against I/R-elicited injury and subsequent heart failure. I/R-induced cardiac inflammation was also ameliorated by CaMKII-δ9 inhibition, and compared with the previously well-studied CaMKII-δ2, CaMKII-δ9 overexpression caused more profound cardiac inflammation. Mechanistically, in addition to IKKβ (inhibitor of NF-κB [nuclear factor-κB] kinase subunit β), CaMKII-δ9, but not δ2, directly interacted with IκBα (NF-κB inhibitor α) with its feature exon 13-16-17 combination and increased IκBα phosphorylation and consequently elicited more pronounced activation of NF-κB signaling and inflammatory response. Furthermore, the essential role of CaMKII-δ9 in myocardial inflammation and damage was confirmed in human cardiomyocytes. CONCLUSIONS We not only identified CaMKII-δ9-IKK/IκB-NF-κB signaling as a new regulator of human cardiomyocyte inflammation but also demonstrated that specifically targeting CaMKII-δ9, the most abundant CaMKII-δ splice variant in human heart, markedly suppresses I/R-induced cardiac NF-κB activation, inflammation, and injury and subsequently ameliorates myocardial remodeling and heart failure, providing a novel therapeutic strategy for various ischemic heart diseases.
Collapse
Affiliation(s)
- Yuan Yao
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.), Peking University, Beijing, China
| | - Fan Li
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.), Peking University, Beijing, China
| | - Mao Zhang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.), Peking University, Beijing, China
| | - Li Jin
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.), Peking University, Beijing, China
| | - Peng Xie
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.), Peking University, Beijing, China
| | - Dairu Liu
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.), Peking University, Beijing, China
| | - Junxia Zhang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.), Peking University, Beijing, China
| | - Xinli Hu
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.), Peking University, Beijing, China
| | - Fengxiang Lv
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.), Peking University, Beijing, China
| | - Haibao Shang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.), Peking University, Beijing, China
| | - Wen Zheng
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.), Peking University, Beijing, China
| | - Xueting Sun
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.), Peking University, Beijing, China
| | - Jiaxin Duanmu
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, School of Basic Medical Sciences, Ministry of Education, Peking University Health Science Center, Beijing, China (J.D., Y.Z.)
| | - Fujian Wu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (F.W., F. Lan)
| | - Feng Lan
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (F.W., F. Lan)
| | - Rui-Ping Xiao
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.), Peking University, Beijing, China
- Beijing City Key Laboratory of Cardiometabolic Molecular Medicine (R.-P.X.), Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Beijing, China (R.-P.X.)
- PKU-Nanjing Institute of Translational Medicine, China (R.-P.X.)
| | - Yan Zhang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.), Peking University, Beijing, China
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, School of Basic Medical Sciences, Ministry of Education, Peking University Health Science Center, Beijing, China (J.D., Y.Z.)
| |
Collapse
|
42
|
Wang H, Hua J, Chen S, Chen Y. SERPINB1 overexpression protects myocardial damage induced by acute myocardial infarction through AMPK/mTOR pathway. BMC Cardiovasc Disord 2022; 22:107. [PMID: 35291946 PMCID: PMC8925243 DOI: 10.1186/s12872-022-02454-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 01/03/2022] [Indexed: 11/23/2022] Open
Abstract
Background SERPINB1 is involved in the development of a variety of diseases. The purpose of this study was to explore the effect of SERPINB1 on acute myocardial infarction (AMI). Methods Serum SERPINB1 level of AMI patients was measured for receiver operating characteristic curve analysis. The AMI rat model was constructed to observe myocardial damage, and the H9C2 cell oxygen glucose deprivation (OGD) model was constructed to detect cell viability. Transthoracic echocardiography was used to assess the cardiac function. TTC staining and HE staining were used to detect pathologic changes of myocardial tissues. The apoptosis of myocardial tissues and cells were measured by TUNLE staining and flow cytometry assay. CCK-8 assay to measure cell viability. SERPINB1 expression was measured by qRT-PCR. Protein expression was measured by western blot. Results The serum SERPINB1 level was down-regulated in AMI patients. AMI modeling reduced the SERPINB1 expression level, induced inflammatory cells infiltrated, and myocardial apoptosis. OGD treatment inhibited cell viability and promoted apoptosis. The AMPK/mTOR pathway was inhibited in AMI rats and OGD-treated H9C2 cells. Overexpression of SERPINB1 reduced infarct size and myocardial apoptosis of AMI rats, inhibited apoptosis of H9C2 cells, and activated AMPK/mTOR pathway. However, AMPK inhibitor Dorsomorphin reversed the protective effect of SERPINB1 on myocardial cells. Conclusion SERPINB1 overexpression relieved myocardial damage induced by AMI via AMPK/mTOR pathway.
Collapse
Affiliation(s)
- Hongliang Wang
- Department of Cardiovasology, First People's Hospital of Jinan, Jinan, 250000, Shandong, People's Republic of China
| | - Jun Hua
- Department of Clinical Laboratory, Gaotang County People's Hospital, Liaocheng, 252800, Shandong, People's Republic of China
| | - Shiyuan Chen
- Department of Breast and Thyroid Surgery, Dongying People's Hospital, Dongying, 257091, Shandong, People's Republic of China
| | - Ying Chen
- Department of Clinical Laboratory, Central Hospital of Shengli Oilfield, No. 31 Jinan Road, Dongying, 257000, Shandong, People's Republic of China.
| |
Collapse
|
43
|
Ma K, Yang J, Shao Y, Li P, Guo H, Wu J, Zhu Y, Zhang H, Zhang X, Du J, Li Y. Therapeutic and Prognostic Significance of Arachidonic Acid in Heart Failure. Circ Res 2022; 130:1056-1071. [PMID: 35255710 DOI: 10.1161/circresaha.121.320548] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Accurate prediction of death is an unmet need in patients with acute decompensated heart failure (HF). Arachidonic acid (AA) metabolites play an important role in the multiple pathophysiological processes. We aimed to develop an AA score to accurately predict mortality in patients with acute decompensated HF and explore the causal relationship between the AA predictors and HF. METHODS The serum AA metabolites was measured in patients with acute decompensated HF (discovery cohort n=419; validation cohort n=386) by mass spectroscopy. We assessed the prognostic importance of AA metabolites for 1-year death using Cox regression and machine learning approaches. An machine learning-based AA score for predicting 1-year death was created and validated. We explored the mechanisms using transcriptome and functional experiments in a mouse model of early ischemic cardiomyopathy. RESULTS Among the 27 AA metabolites, elevated 14,15-DHET/14,15-EET ratio was the strongest predictor of 1-year death (hazard ratio, 2.10, P=3.1×10-6). Machine learning-based AA score using a combination of the 14,15-DHET/14,15-EET ratio, 14,15-DHET, PGD2, and 9-HETE performed best (area under the curve [AUC]: 0.85). The machine learning-based AA score provided incremental information to predict mortality beyond BNP (B-type natriuretic peptide; ΔAUC: 0.19), clinical score (ΔAUC: 0.09), and preexisting ADHERE, Organized Program to Initiate Lifesaving Treatment in Hospitalized Patients With Heart Failure, and Get With The Guidelines Heart Failure scores (ΔAUC: 0.17, 0.17, 0.15, respectively). In the validation cohort, the AA score accurately predicted mortality (AUC:0.81). False-negative and false-positive findings, as classified by the BNP threshold, were correctly reclassified by the AA score (46.2% of false-negative and 84.5% of false-positive). In a murine model, the expression and enzymatic activity of sEH (soluble epoxide hydrolase) increased after myocardial infarction. Genetic deletion of sEH improved HF and the blockade of 14,15-EET abolished this cardioprotection. We mechanistically revealed the beneficial effect of 14,15-EET by impairing the activation of monocytes/macrophages. CONCLUSIONS Our studies propose that the AA score predicts death in patients with acute decompensated HF and inhibiting sEH serves as a therapeutic target for treating HF. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT04108182.
Collapse
Affiliation(s)
- Ke Ma
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (K.M., J.Y., Y.S., P.L., H.G., J.W., J.D., Y.L.)
| | - Jie Yang
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (K.M., J.Y., Y.S., P.L., H.G., J.W., J.D., Y.L.)
| | - Yihui Shao
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (K.M., J.Y., Y.S., P.L., H.G., J.W., J.D., Y.L.)
| | - Ping Li
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (K.M., J.Y., Y.S., P.L., H.G., J.W., J.D., Y.L.)
| | - Hongchang Guo
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (K.M., J.Y., Y.S., P.L., H.G., J.W., J.D., Y.L.)
| | - Jianing Wu
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (K.M., J.Y., Y.S., P.L., H.G., J.W., J.D., Y.L.)
| | - Yi Zhu
- Tianjin Key Laboratory of Metabolic Diseases, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Center for Cardiovascular Diseases, Research Center of Basic Medical Sciences, Department of Physiology and Pathophysiology, Tianjin Medical University, China (Y.Z., X.Z.)
| | - Hui Zhang
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University (H.Z.)
| | - Xu Zhang
- Tianjin Key Laboratory of Metabolic Diseases, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Center for Cardiovascular Diseases, Research Center of Basic Medical Sciences, Department of Physiology and Pathophysiology, Tianjin Medical University, China (Y.Z., X.Z.)
| | - Jie Du
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (K.M., J.Y., Y.S., P.L., H.G., J.W., J.D., Y.L.)
| | - Yulin Li
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (K.M., J.Y., Y.S., P.L., H.G., J.W., J.D., Y.L.)
| |
Collapse
|
44
|
Feng G, Bajpai G, Ma P, Koenig A, Bredemeyer A, Lokshina I, Lai L, Förster I, Leuschner F, Kreisel D, Lavine KJ. CCL17 Aggravates Myocardial Injury by Suppressing Recruitment of Regulatory T Cells. Circulation 2022; 145:765-782. [PMID: 35113652 PMCID: PMC8957788 DOI: 10.1161/circulationaha.121.055888] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 01/07/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Recent studies have established that CCR2 (C-C chemokine receptor type 2) marks proinflammatory subsets of monocytes, macrophages, and dendritic cells that contribute to adverse left ventricle (LV) remodeling and heart failure progression. Elucidation of the effector mechanisms that mediate adverse effects of CCR2+ monocytes, macrophages, and dendritic cells will yield important insights into therapeutic strategies to suppress myocardial inflammation. METHODS We used mouse models of reperfused myocardial infarction, angiotensin II and phenylephrine infusion, and diphtheria toxin cardiomyocyte ablation to investigate CCL17 (C-C chemokine ligand 17). We used Ccl17 knockout mice, flow cytometry, RNA sequencing, biochemical assays, cell trafficking studies, and in vivo cell depletion to identify the cell types that generate CCL17, define signaling pathways that controlled its expression, delineate the functional importance of CCL17 in adverse LV remodeling and heart failure progression, and determine the mechanistic basis by which CCL17 exerts its effects. RESULTS We demonstrated that CCL17 is expressed in CCR2+ macrophages and cluster of differentiation 11b+ conventional dendritic cells after myocardial infarction, angiotensin II and phenylephrine infusion, and diphtheria toxin cardiomyocyte ablation. We clarified the transcriptional signature of CCL17+ macrophages and dendritic cells and identified granulocyte-macrophage colony-stimulating factor (GM-CSF) signaling as a key regulator of CCL17 expression through cooperative activation of STAT5 (signal transducer and activator of transcription 5) and canonical NF-κB (nuclear factor κ-light-chain-enhancer of activated B cells) signaling. Ccl17 deletion resulted in reduced LV remodeling, decreased myocardial fibrosis and cardiomyocyte hypertrophy, and improved LV systolic function after myocardial infarction and angiotensin II and phenylephrine infusion. We observed increased abundance of regulatory T cells (Tregs) in the myocardium of injured Ccl17 knockout mice. CCL17 inhibited Treg recruitment through biased activation of CCR4. CCL17 activated Gq signaling and CCL22 (C-C chemokine ligand 22) activated both Gq and ARRB (β-arrestin) signaling downstream of CCR4. CCL17 competitively inhibited CCL22 stimulated ARRB signaling and Treg migration. We provide evidence that Tregs mediated the protective effects of Ccl17 deletion on myocardial inflammation and adverse LV remodeling. CONCLUSIONS These findings identify CCL17 as a proinflammatory mediator of CCR2+ macrophages and dendritic cells and suggest that inhibition of CCL17 may serve as an effective strategy to promote Treg recruitment and suppress myocardial inflammation.
Collapse
Affiliation(s)
- Guoshuai Feng
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
| | - Geetika Bajpai
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
| | - Pan Ma
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
| | - Andrew Koenig
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
| | - Andrea Bredemeyer
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
| | - Inessa Lokshina
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
| | - Lulu Lai
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
| | | | - Florian Leuschner
- LIMES Institute, University of Bonn, Department of Internal Medicine III, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany (F.L.)
| | - Daniel Kreisel
- Department of Surgery, Washington University, Saint Louis, Missouri, USA (D.K.)
- Department of Pathology and Immunology, Washington University, Saint Louis, Missouri, USA (D.K., K.L.)
| | - Kory J. Lavine
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
- Department of Pathology and Immunology, Washington University, Saint Louis, Missouri, USA (D.K., K.L.)
- Department of Developmental Biology, Washington University, Saint Louis, Missouri, USA (K.L.)
| |
Collapse
|
45
|
El-Awaisi J, Kavanagh DP, Rink MR, Weston CJ, Drury NE, Kalia N. Targeting IL-36 improves age-related coronary microcirculatory dysfunction and attenuates myocardial ischaemia-reperfusion injury in mice. JCI Insight 2022; 7:155236. [PMID: 35113814 PMCID: PMC8983126 DOI: 10.1172/jci.insight.155236] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/28/2022] [Indexed: 11/17/2022] Open
Abstract
Following myocardial infarction (MI), elderly patients have a poorer prognosis than younger patients, which may be linked to increased coronary microvessel susceptibility to injury. Interleukin-36 (IL-36), a newly discovered proinflammatory member of the IL-1 superfamily, may mediate this injury, but its role in the injured heart is currently not known. We first demonstrated the presence of IL-36(α/β) and its receptor (IL-36R) in ischemia/reperfusion-injured (IR-injured) mouse hearts and, interestingly, noted that expression of both increased with aging. An intravital model for imaging the adult and aged IR-injured beating heart in real time in vivo was used to demonstrate heightened basal and injury-induced neutrophil recruitment, and poorer blood flow, in the aged coronary microcirculation when compared with adult hearts. An IL-36R antagonist (IL-36Ra) decreased neutrophil recruitment, improved blood flow, and reduced infarct size in both adult and aged mice. This may be mechanistically explained by attenuated endothelial oxidative damage and VCAM-1 expression in IL-36Ra–treated mice. Our findings of an enhanced age-related coronary microcirculatory dysfunction in reperfused hearts may explain the poorer outcomes in elderly patients following MI. Since targeting the IL-36/IL-36R pathway was vasculoprotective in aged hearts, it may potentially be a therapy for treating MI in the elderly population.
Collapse
Affiliation(s)
- Juma El-Awaisi
- Institute of Cardiovascular Sciences, University of Birmingham Medical School, Birmingham, United Kingdom
| | - Dean Pj Kavanagh
- Institute of Cardiovascular Sciences, University of Birmingham Medical School, Birmingham, United Kingdom
| | - Marco R Rink
- Institute of Immunology and Immunotherapy, University of Birmingham Medical School, Birmingham, United Kingdom
| | - Chris J Weston
- Institute of Immunology and Immunotherapy, University of Birmingham Medical School, Birmingham, United Kingdom
| | - Nigel E Drury
- Institute of Cardiovascular Sciences, University of Birmingham Medical School, Birmingham, United Kingdom
| | - Neena Kalia
- Institute of Cardiovascular Sciences, University of Birmingham Medical School, Birmingham, United Kingdom
| |
Collapse
|
46
|
Shan M, Yu X, Li Y, Fu C, Zhang C. Vitamin B6 Alleviates Lipopolysaccharide-induced Myocardial Injury by Ferroptosis and Apoptosis Regulation. Front Pharmacol 2022; 12:766820. [PMID: 35002705 PMCID: PMC8740299 DOI: 10.3389/fphar.2021.766820] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/04/2021] [Indexed: 01/15/2023] Open
Abstract
Vitamin B6 (VitB6) is a water-soluble vitamin and includes pyridoxine, pyridoxal, pyridoxamine, and their phosphorylated forms. In the current study, we demonstrated that VitB6 could improve lipopolysaccharide (LPS)-induced myocardial injury. We demonstrated that VitB6 can suppress LPS-induced oxidative stress and lipid peroxidation that lead to ferroptosis and apoptosis in vivo and in vitro. Moreover, we found that VitB6 can regulate the expression of iron regulatory proteins, maintaining intracellular iron homeostasis. To confirm that VitB6 could inhibit LPS-induced ferroptosis and apoptosis, we pretreated mice with ferrostatin-1 (Fer-1) and emricasan that efficiently mimicked VitB6 pharmacological effects. This improved the survival rate of mice challenged with a high LPS dose. In addition, VitB6 regulated the expression of LPS-induced apoptosis-related proteins and iron regulatory proteins. It mediated the expression of Nrf2, transcription factor NF-E2-related factor 2, which promoted the expression of antioxidant enzymes and restrained LPS-induced ferroptosis and apoptosis. Overall, our results indicated that VitB6 can be used on novel therapies to relieve LPS-induced myocardial injury.
Collapse
Affiliation(s)
- Meirong Shan
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China.,Department of Geriatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xujie Yu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| | - Yajie Li
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Changning Fu
- Department of Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
47
|
Zhang L, Wen K, Zhang Z, Ma C, Zheng N. 3,4-Dihydroxyphenylethanol ameliorates lipopolysaccharide-induced septic cardiac injury in a murine model. Open Life Sci 2022; 16:1313-1320. [PMID: 35005242 PMCID: PMC8691377 DOI: 10.1515/biol-2021-0125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022] Open
Abstract
3,4-Dihydroxyphenylethanol (DOPET) is a polyphenol found in olive oil. The present study evaluated the protective role of DOPET on LPS provoked septic cardiac injury in a murine model. Four groups were used in the study (n = 3): control, LPS, DOPET alone, and DOPET + LPS. LPS (15 mg/kg; i.p.); they were used to induce cardiac sepsis. The cardiac markers like LDH, CK-MB, and troponin-T, as well as inflammatory cytokines like TNF-α and IL-6 were measured in the serum. The antioxidants and oxidative stress parameters were measured in cardiac tissues. RT-PCR and western blot methods were done to evaluate the expression of inflammatory mediators and apoptotic markers. DOPET significantly decreased the cardiac markers (LDH, CK-MB, and troponin-T) and TNF-α and IL-6 level in the serum. DOPET effectively reduced the levels of MDA and NO in LPS intoxicated rats. DOPET also increased the levels of antioxidants like SOD, CAT, GPx, and GSH in LPS intoxicated rats. The mRNA levels of TNF-α, IL-6, and NF-κB were significantly downregulated by DOPET in cardiac tissues of LPS rats. The protein expression of Bcl-2 was upregulated, and Bax and caspase-3 were downregulated by DOPET. DOPET effectively attenuates LPS-induced cardiac dysfunction through its antioxidant, anti-inflammatory, and anti-apoptotic mechanisms.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Intensive Care Unit, The Second Hospital of Shandong University, Jinan, Shandong Province, 250033, China
| | - Kun Wen
- Department of Intensive Care Unit, The Second Hospital of Shandong University, Jinan, Shandong Province, 250033, China
| | - Zhiqiang Zhang
- Department of Intensive Care Unit, The Second Hospital of Shandong University, Jinan, Shandong Province, 250033, China
| | - Chengen Ma
- Department of Intensive Care Unit, The Second Hospital of Shandong University, Jinan, Shandong Province, 250033, China
| | - Ni Zheng
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwu Weiqi Road, Jinan, Shandong Province, 250021, China
| |
Collapse
|
48
|
Tang Y, Shi C, Qin Y, Wang S, Pan H, Chen M, Yu X, Lou Y, Fan G. Network Pharmacology-Based Investigation and Experimental Exploration of the Antiapoptotic Mechanism of Colchicine on Myocardial Ischemia Reperfusion Injury. Front Pharmacol 2022; 12:804030. [PMID: 34975499 PMCID: PMC8716846 DOI: 10.3389/fphar.2021.804030] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 11/29/2021] [Indexed: 12/11/2022] Open
Abstract
Background: The beneficial effects of colchicine on cardiovascular disease have been widely reported in recent studies. Previous research demonstrated that colchicine has a certain protective effect on ischemic myocardium and has the potential to treat myocardial ischemia reperfusion injury (MIRI). However, the potential targets and pharmacological mechanism of colchicine to treat MIRI has not been reported. Methods: In this study, we used network pharmacology and experimental verification to investigate the pharmacological mechanisms of colchicine for the treatment of MIRI. Potential targets of colchicine and MIRI related genes were screened from public databases. The mechanism of colchicine in the treatment of MIRI was determined by protein-protein interaction (PPI), gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Additionally, we evaluated the effect of colchicine on H9C2 cell activity using CCK-8 assays, observed the effect of colchicine on H9C2 cell apoptosis via flow cytometry, and further verified the expression of key targets after colchicine treated by Western blot. Results: A total of 626 target genes for colchicine and 1549 MIRI disease targets were obtained. 138 overlapping genes were determined as potential targets of colchicine in treating MIRI. the PPI network analysis demonstrated that the targets linked to MIRI were ALB, TNF, ACTB, AKT1, IL6, TP53, IL1B, CASP3 and these targets showed nice affinity with colchicine in molecular docking experiments. The results of GO analysis and KEGG pathway enrichment demonstrated that the anti-MIRI effect of colchicine involves in apoptotic signaling pathway. Further tests suggested that colchicine can protect H9C2 cell from Hypoxia/Reoxygenation (H/R) injury through anti-apoptotic effects. Western blot results demonstrated that colchicine can inhibited MIRI induced apoptosis of H9C2 cell by enhancing the decreased levels of Caspase-3 in myocardial injure model induced by H/R and activating the PI3K/AKT/eNOS pathway. Conclusions: we performed network pharmacology and experimental evaluation to reveal the pharmacological mechanism of colchicine against MIRI. The results from this study could provide a theoretical basis for the development and clinical application of colchicine.
Collapse
Affiliation(s)
- Yuanjun Tang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chenyang Shi
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yingyi Qin
- Department of Health Statistics, Naval Medical University, Shanghai, China
| | - Shuowen Wang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hui Pan
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ming Chen
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xuemei Yu
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuefen Lou
- Department of Pharmacy, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Guorong Fan
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
49
|
The nuclear receptor co-repressor 1 is a novel cardioprotective factor against acute myocardial ischemia-reperfusion injury. J Mol Cell Cardiol 2022; 166:50-62. [DOI: 10.1016/j.yjmcc.2022.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 01/01/2022] [Accepted: 01/18/2022] [Indexed: 12/31/2022]
|
50
|
Seidman MA, McManus B. Myocarditis. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00005-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|