1
|
Lan C, Fang G, Li X, Chen X, Chen Y, Hu T, Wang X, Cai H, Hao J, Li H, Zhang Y, Peng K, Xu Z, Yang D, Kang X, Xin Q, Yang Y. SerpinB1 targeting safeguards against pathological cardiac hypertrophy and remodelling by suppressing cardiomyocyte pyroptosis and inflammation initiation. Cardiovasc Res 2025; 121:113-127. [PMID: 39688818 DOI: 10.1093/cvr/cvae241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 07/06/2024] [Accepted: 09/15/2024] [Indexed: 12/18/2024] Open
Abstract
AIMS While the pivotal role of inflammation in pathological cardiac hypertrophy and remodelling is widely acknowledged, the mechanisms triggering inflammation initiation remain largely obscure. This study aims to elucidate the role and mechanism of serpin family B member 1 (SerpinB1) in pro-inflammatory cardiomyocyte pyroptosis, heart inflammation, and cardiac remodelling. METHODS AND RESULTS C57BL/6J wild-type, inducible cardiac-specific SerpinB1 overexpression or knockout mice underwent transverse aortic constriction (TAC) surgery. Cardiac hypertrophy and remodelling were assessed through echocardiography and histology. Cardiomyocyte pyroptosis and heart inflammation were monitored. Adeno-associated virus 9 -mediated gene manipulations and molecular assays were employed to explore the mechanisms through which SerpinB1 regulates cardiomyocyte pyroptosis and heart inflammation. Finally, recombinant mouse SerpinB1 protein (rSerpinB1) was administrated both in vivo through osmotic minipump delivery and in vitro to investigate the therapeutic potential of SerpinB1 in cardiac remodelling. Myocardial SerpinB1 overexpression was up-regulated shortly upon TAC or phenylephrine challenge, with no further elevation during prolonged hypertrophic stimuli. It is important to note that cardiac-specific overexpression of SerpinB1 markedly attenuated TAC-induced cardiac remodelling, while deletion of SerpinB1 exacerbated it. At the mechanistic level, SerpinB1 gain-of-function inhibited cardiomyocyte pyroptosis and inflammation in hypertrophic hearts; the protective effect was nullified by overexpression of either cleaved N-terminal gasdermin D or cleaved caspase-1. Co-immunoprecipitation and confocal assays confirmed that SerpinB1 directly interacts with caspase-1 in cardiomyocytes. Remarkably, rSerpinB1 replicated the cardioprotective effect against cardiac hypertrophy and remodelling. CONCLUSION SerpinB1 safeguards against pathological cardiac hypertrophy and remodelling by impeding cardiomyocyte pyroptosis to suppress inflammation initiation, achieved through interaction with caspase-1 to inhibit its activation. Targeting SerpinB1 could represent a novel therapeutic strategy for treating pathological cardiac hypertrophy and remodelling.
Collapse
MESH Headings
- Animals
- Pyroptosis/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/drug effects
- Serpins/genetics
- Serpins/metabolism
- Ventricular Remodeling/drug effects
- Mice, Knockout
- Mice, Inbred C57BL
- Disease Models, Animal
- Male
- Signal Transduction
- Cells, Cultured
- Myocarditis/metabolism
- Myocarditis/pathology
- Myocarditis/prevention & control
- Myocarditis/genetics
- Myocarditis/physiopathology
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/prevention & control
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/physiopathology
- Ventricular Function, Left
- Cardiomegaly/metabolism
- Cardiomegaly/prevention & control
- Cardiomegaly/pathology
- Cardiomegaly/genetics
- Inflammation Mediators/metabolism
- Caspase 1/metabolism
Collapse
Affiliation(s)
- Cong Lan
- Department of Cardiology, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu , Sichuan 610083, P.R. China
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu, Sichuan 610083, P.R.China
- College of Medicine, Southwest Jiaotong University, No. 144, Jiaoda Road, Jinniu District, Chengdu, Sichuan 610031, P.R. China
| | - Guangyao Fang
- Department of Cardiology, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu , Sichuan 610083, P.R. China
| | - Xiuchuan Li
- Department of Cardiology, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu , Sichuan 610083, P.R. China
| | - Xiao Chen
- Department of General Practice, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu, Sichuan 610083, P.R. China
| | - Yingmei Chen
- Department of Cardiology, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu , Sichuan 610083, P.R. China
| | - Tao Hu
- Department of Cardiology, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu , Sichuan 610083, P.R. China
| | - Xuenan Wang
- School of Clinical Medicine, Southwest Medical University, No. 319, Zhongshan Road, Jiangyang District, Luzhou, Sichuan 646000, P.R. China
| | - Huiling Cai
- School of Clinical Medicine, Southwest Medical University, No. 319, Zhongshan Road, Jiangyang District, Luzhou, Sichuan 646000, P.R. China
| | - Jiajin Hao
- Department of Cardiology, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu , Sichuan 610083, P.R. China
- College of Medicine, Southwest Jiaotong University, No. 144, Jiaoda Road, Jinniu District, Chengdu, Sichuan 610031, P.R. China
| | - Haoran Li
- Department of Cardiology, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu , Sichuan 610083, P.R. China
- College of Medicine, Southwest Jiaotong University, No. 144, Jiaoda Road, Jinniu District, Chengdu, Sichuan 610031, P.R. China
| | - Yan Zhang
- Department of Cardiology, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu , Sichuan 610083, P.R. China
| | - Ke Peng
- Department of Cardiology, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu , Sichuan 610083, P.R. China
| | - Zaicheng Xu
- Department of Cancer Center, Second Affiliated Hospital, Chongqing Medical University, No. 288, Tianwen Road, Nanan District, Chongqing 400072, P.R. China
| | - Dachun Yang
- Department of Cardiology, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu , Sichuan 610083, P.R. China
| | - Xia Kang
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu, Sichuan 610083, P.R.China
| | - Qian Xin
- Department of Cardiology, Sixth Medical Center of Chinese PLA General Hospital, No. 6, Fucheng Road, Haidian District, Beijing 100048, P.R. China
| | - Yongjian Yang
- Department of Cardiology, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu , Sichuan 610083, P.R. China
- College of Medicine, Southwest Jiaotong University, No. 144, Jiaoda Road, Jinniu District, Chengdu, Sichuan 610031, P.R. China
- School of Clinical Medicine, Southwest Medical University, No. 319, Zhongshan Road, Jiangyang District, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
2
|
Xu J, Sun Z, Li J, Li Y, Huang H, Yuan F, Liu M, Fang Z. Qian Yang Yu Yin Granule prevents hypertensive cardiac remodeling by inhibiting NLRP3 inflammasome activation via Nrf2. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118820. [PMID: 39278297 DOI: 10.1016/j.jep.2024.118820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/29/2024] [Accepted: 09/09/2024] [Indexed: 09/18/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qian Yang Yu Yin Granule (QYYYG), a traditional Chinese poly-herbal formulation, has been validated in clinical trials to mitigate cardiac remodeling (CR), and cardiac damage in patients with hypertension. However, the specific mechanism remains unclear. AIM OF THE STUDY This study explored the potential effects and potential mechanisms of QYYYG on hypertensive CR by combining various experimental approaches. MATERIALS AND METHODS Spontaneously hypertensive rats (SHRs) were used as a model of hypertensive CR, followed by QYYYG interventions. Blood pressure, cardiac function and structure, histopathological changes, and myocardial inflammation and oxidative stress were tested to assess the efficacy of QYYYG in SHRs. For in vitro experiments, a cell model of myocardial hypertrophy and injury was constructed with isoprenaline. Cardiomyocyte hypertrophy, oxidative stress, and death were examined after treatment with different concentrations of QYYYG, and transcriptomics analyses were performed to explore the underlying mechanism. Nrf2 and the ROS/NF-κB/NLRP3 inflammasome pathway were detected. Thereafter, ML385 and siRNAs were used to inhibit Nrf2 in cardiomyocytes, so as to verify whether QYYYG negatively regulates the NLRP3 inflammasome by targeting Nrf2, thereby ameliorating the associated phenotypes. Finally, high performance liquid chromatography (HPLC) was conducted to analyze the active ingredients in QYYYG, and molecular docking was utilized to preliminarily screen the compounds with modulatory effects on Nrf2 activities. RESULTS QYYYG improved blood pressure, cardiac function, and structural remodeling and attenuated myocardial inflammation, oxidative stress, and cell death in SHRs. The transcriptomics results showed that the inflammatory response might be crucial in pathological CR and that Nrf2, which potentially negatively regulates the process, was upregulated by QYYYG treatment. Furthermore, QYYYG indeed facilitated Nrf2 activation and negatively regulated the ROS/NF-κB/NLRP3 inflammasome pathway, therefore ameliorating the associated phenotypes. In vitro inhibition or knockdown of Nrf2 weakened or even reversed the repressive effect of QYYYG on ISO-induced inflammation, oxidative stress, pyroptosis, and the NLRP3 inflammasome activation. Based on the results of HPLC and molecular docking, 30 compounds, including cafestol, genistein, hesperetin, and formononetin, have binding sites to Keap1-Nrf2 protein and might affect the activity or stability of Nrf2. CONCLUSION In conclusion, the alleviatory effect of QYYYG on hypertensive CR is related to its regulation of Nrf2 activation. Specifically, QYYYG blocks the activation of the NLRP3 inflammasome by boosting Nrf2 signaling and depressing myocardial inflammation, oxidative stress, and pyroptosis, thereby effectively ameliorating hypertensive CR.
Collapse
Affiliation(s)
- Junyao Xu
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| | - Zeqi Sun
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| | - Jie Li
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| | - Yin Li
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| | - Hong Huang
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| | - Fang Yuan
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| | - Ming Liu
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China; Institute of Hypertension, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| | - Zhuyuan Fang
- Institute of Hypertension, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| |
Collapse
|
3
|
Bahrami P, Aromolaran KA, Aromolaran AS. Mechanistic Relevance of Ventricular Arrhythmias in Heart Failure with Preserved Ejection Fraction. Int J Mol Sci 2024; 25:13423. [PMID: 39769189 PMCID: PMC11677834 DOI: 10.3390/ijms252413423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/05/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is increasing at an alarming rate worldwide, with limited effective therapeutic interventions in patients. Sudden cardiac death (SCD) and ventricular arrhythmias present substantial risks for the prognosis of these patients. Obesity is a risk factor for HFpEF and life-threatening arrhythmias. Obesity and its associated metabolic dysregulation, leading to metabolic syndrome, are an epidemic that poses a significant public health problem. More than one-third of the world population is overweight or obese, leading to an enhanced risk of incidence and mortality due to cardiovascular disease (CVD). Obesity predisposes patients to atrial fibrillation and ventricular and supraventricular arrhythmias-conditions that are caused by dysfunction in the electrical activity of the heart. To date, current therapeutic options for the cardiomyopathy of obesity are limited, suggesting that there is considerable room for the development of therapeutic interventions with novel mechanisms of action that will help normalize sinus rhythms in obese patients. Emerging candidates for modulation by obesity are cardiac ion channels and Ca-handling proteins. However, the underlying molecular mechanisms of the impact of obesity on these channels and Ca-handling proteins remain incompletely understood. Obesity is marked by the accumulation of adipose tissue, which is associated with a variety of adverse adaptations, including dyslipidemia (or abnormal systemic levels of free fatty acids), increased secretion of proinflammatory cytokines, fibrosis, hyperglycemia, and insulin resistance, which cause electrical remodeling and, thus, predispose patients to arrhythmias. Furthermore, adipose tissue is also associated with the accumulation of subcutaneous and visceral fat, which is marked by distinct signaling mechanisms. Thus, there may also be functional differences in the effects of the regional distribution of fat deposits on ion channel/Ca-handling protein expression. Evaluating alterations in their functional expression in obesity will lead to progress in the knowledge of the mechanisms responsible for obesity-related arrhythmias. These advances are likely to reveal new targets for pharmacological modulation. Understanding how obesity and related mechanisms lead to cardiac electrical remodeling is likely to have a significant medical and economic impact. Nevertheless, substantial knowledge gaps remain regarding HFpEF treatment, requiring further investigations to identify potential therapeutic targets. The objective of this study is to review cardiac ion channel/Ca-handling protein remodeling in the predisposition to metabolic HFpEF and arrhythmias. This review further highlights interleukin-6 (IL-6) as a potential target, cardiac bridging integrator 1 (cBIN1) as a promising gene therapy agent, and leukotriene B4 (LTB4) as an underappreciated pathway in future HFpEF management.
Collapse
Affiliation(s)
- Pegah Bahrami
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, 95 S 2000 E, Salt Lake City, UT 84112, USA; (P.B.); (K.A.A.)
| | - Kelly A. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, 95 S 2000 E, Salt Lake City, UT 84112, USA; (P.B.); (K.A.A.)
| | - Ademuyiwa S. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, 95 S 2000 E, Salt Lake City, UT 84112, USA; (P.B.); (K.A.A.)
- Department of Surgery, Division of Cardiothoracic Surgery, Nutrition & Integrative Physiology, Biochemistry & Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| |
Collapse
|
4
|
Zhang J, Sun J, Gu X, Shen Y, Sun H. Transcriptome sequencing analysis reveals the molecular regulatory mechanism of myocardial hypertrophy induced by angiotensin II. Biochem Pharmacol 2024; 229:116532. [PMID: 39270943 DOI: 10.1016/j.bcp.2024.116532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 08/26/2024] [Accepted: 09/09/2024] [Indexed: 09/15/2024]
Abstract
The pathogenesis of myocardial hypertrophy remains incompletely understood, highlighting the critical need for in-depth investigation into its pathogenesis and pathophysiology to develop innovative strategies for preventing and treating heart diseases. In this study, a model of angiotensin II (Ang II)-induced myocardial hypertrophy was established using subcutaneous administration with a micropump. Echocardiography, wheat germ agglutinin staining, and western blot analysis were used to evaluate the myocardial hypertrophy model after 5, 10, and 15 days of Ang II treatment. RNA-seq was employed to analyze the differential expression profile of mRNA, followed by bioinformatics analysis. Subsequently, the anti-inflammatory drug meloxicam was utilized to explore its impact on cardiac hypertrophy in mice. The findings demonstrated that mice developed myocardial hypertrophy following subcutaneous administration of Ang II. Transcriptomic analysis revealed significant changes in gene expression in the myocardium induced by Ang II, with the most pronounced differences observed at day 10. Functional analysis and verification of differentially expressed genes indicated that Ang II triggered an inflammatory response in the myocardium, leading to up-regulation of genes associated with fibrosis and apoptosis while decreasing energy metabolism; alterations were also observed in genes related to oxidative stress and calcium ion binding. Treatment with meloxicam improved Ang II-induced myocardial hypertrophy. This study not only elucidated the molecular regulatory mechanism underlying mouse myocardial hypertrophy at a transcriptional level but also provided new insights into clinical prevention and treatment strategies for cardiac diseases such as dilated cardiomyopathy and heart failure.
Collapse
Affiliation(s)
- Jingjing Zhang
- Laboratory of General Surgery, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province 226001, PR China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Jiacheng Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province 226001, PR China.
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province 226001, PR China.
| |
Collapse
|
5
|
Mahmoud LM, Mageed AAAA, Saadallah JM, Youssef MF, Rashed LA, Ammar HI. Interleukin 1β receptor blocker (Anakinra) and regenerative stem cell therapy: two novel approaches effectively ameliorating diabetic cardiomyopathy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8023-8041. [PMID: 38775851 PMCID: PMC11450109 DOI: 10.1007/s00210-024-03152-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/06/2024] [Indexed: 10/04/2024]
Abstract
Diabetic cardiomyopathy (DCM) is a serious common complication of diabetes. Unfortunately, there is no satisfied treatment for those patients and more studies are in critical need to cure them. Therefore, we aimed to carry out our current research to explore the role of two novel therapeutic approaches: one a biological drug aimed to block inflammatory signaling of the IL 1beta (IL1β) axis, namely, anakinra; the other is provision of anti-inflammatory regenerative stem cells. Wistar male rats were allocated into four groups: control group: type 2 diabetes mellitus (DM) induced by 6-week high-fat diet (HFD) followed by a single-dose streptozotocin (STZ) 35 mg/kg i.p., then rats were allocated into: DM: untreated; DM BM-MSCs: received a single dose of BM-MSCs (1 × 106 cell/rat) into rat tail vein; DM-Anak received Anak 0.5 μg/kg/day i.p. for 2 weeks. Both therapeutic approaches improved cardiac performance, fibrosis, and hypertrophy. In addition, blood glucose and insulin resistance decreased, while the antioxidant parameter, nuclear factor erythroid 2-related factor 2 (Nrf2) and interleukin 10 (IL10), and anti-inflammatory agent increased. Furthermore, there is a significant reduction in tumor necrosis factor alpha (TNFα), IL1β, caspase1, macrophage marker CD 11b, inducible nitric oxide synthase (iNOS), and T-cell marker CD 8. Both Anak and BM-MSCs effectively ameliorated inflammatory markers and cardiac performance as compared to non-treated diabetics. Improvement is mostly due to anti-inflammatory, antioxidant, anti-apoptotic properties, and regulation of TNFα/IL1β/caspase1 and Nrf2/IL10 pathways.
Collapse
Affiliation(s)
| | | | | | | | - Liala Ahmed Rashed
- Department of Biochemistry, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Hania Ibrahim Ammar
- Department of Physiology, Faculty of Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
6
|
Moreyra C, Moreyra E, Rozich JD. Heart Failure With Preserved Ejection Fraction: Will Cardiac Magnetic Imaging Impact on Diagnosis, Treatment, and Outcomes?: Explaining the Need for Advanced Imaging to Clinical Stakeholders. Cardiol Rev 2024; 32:371-377. [PMID: 36576375 DOI: 10.1097/crd.0000000000000494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Clinicians frequently equate symptoms of volume overload to heart failure (HF) but such generalization may preclude diagnostic or etiologic precision essential to optimizing outcomes. HF itself must be specified as the disparate types of cardiac pathology have been traditionally surmised by examination of left ventricular (LV) ejection fraction (EF) as either HF with preserved LVEF (HFpEF-LVEF >50%) or reduced LVEF of (HFrEF-LVEF <40%). More recent data support a third, potentially transitional HF subtype, but therapy, assessment, and prognosis have been historically dictated within the corresponding LV metrics determined by echocardiography. The present effort asks whether this historically dominant role of echocardiography is now shifting slightly, becoming instead a shared if not complimentary test. Will there be a gradual increasing profile for cardiac magnetic resonance as the attempt to further refine our understanding, diagnostic accuracy, and outcomes for HFpEF is attempted?
Collapse
Affiliation(s)
- Camila Moreyra
- From the Cardiology Department, Sanatorium Allende, Córdoba, Argentina
| | - Eduardo Moreyra
- From the Cardiology Department, Sanatorium Allende, Córdoba, Argentina
| | | |
Collapse
|
7
|
Di Vincenzo A, Crescenzi M, Granzotto M, Vecchiato M, Fioretto P, Vettor R, Rossato M. Treatment with dapagliflozin increases FGF-21 gene expression and reduces triglycerides content in myocardial tissue of genetically obese mice. J Endocrinol Invest 2024; 47:1777-1786. [PMID: 38194168 DOI: 10.1007/s40618-023-02273-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 12/05/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND The association between obesity and some cardiovascular complications such as heart failure (HF) is well established, and drugs affecting adiposity are supposed to be promising treatments for these conditions. The sodium-glucose cotransporter-2 inhibitors (SGLT2i) are antidiabetic drugs showing benefits in patients with HF, despite the underlying mechanisms have not been completely understood yet. SGLT2i are supposed to promote systemic effects, such as triglycerides mobilization, through the enhancement of fibroblast growth factor-21 (FGF-21) activity. So, in this study, we evaluated the effects of dapagliflozin treatment on FGF-21 and related receptors (FGF-Rs) gene expression and on lipid content in myocardial tissue in an animal model of genetically induced obesity to unravel possible metabolic mechanisms accounting for the cardioprotection of SGLT2i. METHODS Six-week-old C57BL/6J wild-type mice and B6.V-LEP (ob/ob) mice were randomly assigned to the control or treatment group (14 animals/group). Treatment was based on the administration of dapagliflozin 0.15 mg/kg/day for 4 weeks. The gene expression of FGF-21 and related receptors (FGF-R1, FGF-R3, FGF-R4, and β-klotho co-receptor) was assessed at baseline and after treatment by real-time PCR. Similarly, cardiac triglycerides concentration was measured in the control group and treated animals. RESULTS At baseline, FGF-21 mRNA expression in the heart did not differ between lean and obese ob/ob mice. Dapagliflozin administration significantly increased heart FGF-21 gene expression, but only in ob/ob mice (p < 0.005). Consistently, when measuring the amount of triglycerides in the cardiac tissue, SGLT2i treatment reduced the lipid content in obese ob/ob mice, while no significant effects were observed in treated lean animals (p < 0.001). The overall expression of the FGF-21 receptors was only minimally affected by dapagliflozin treatment both in obese ob/ob mice and in lean controls. CONCLUSIONS Dapagliflozin administration increases FGF-21gene expression and reduces triglyceride content in myocardial tissue of ob/ob mice, while no significant effect was observed in lean controls. These results might help understand the cardiometabolic effects of SGLT2i inducing increased FGF-21 synthesis while reducing lipid content in cardiomyocytes as a possible expression of the switch to different energy substrates. This mechanism could represent a potential target of SGLT2i in obesity-related heart diseases.
Collapse
Affiliation(s)
- A Di Vincenzo
- Internal Medicine 3, Department of Medicine, University-Hospital of Padova, Padua, Italy.
| | - M Crescenzi
- Internal Medicine 3, Department of Medicine, University-Hospital of Padova, Padua, Italy
| | - M Granzotto
- Internal Medicine 3, Department of Medicine, University-Hospital of Padova, Padua, Italy
| | - M Vecchiato
- Sports and Exercise Medicine Division, Department of Medicine, University-Hospital of Padova, Padua, Italy
| | - P Fioretto
- Internal Medicine 3, Department of Medicine, University-Hospital of Padova, Padua, Italy
| | - R Vettor
- Internal Medicine 3, Department of Medicine, University-Hospital of Padova, Padua, Italy
| | - M Rossato
- Internal Medicine 3, Department of Medicine, University-Hospital of Padova, Padua, Italy
| |
Collapse
|
8
|
Haybar H, Sarbazjoda E, Purrahman D, Mahmoudian-Sani MR, Saki N. The prognostic potential of long noncoding RNA XIST in cardiovascular diseases: a review. Per Med 2024; 21:257-269. [PMID: 38889283 DOI: 10.1080/17410541.2024.2360380] [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: 08/26/2023] [Accepted: 05/23/2024] [Indexed: 06/20/2024]
Abstract
There is a significant mortality rate associated with cardiovascular disease despite advances in treatment. long Non-coding RNAs (lncRNAs) play a critical role in many biological processes and their dysregulation is associated with a wide range of diseases in which their downstream pathways are disrupted. A lncRNA X-inactive specific transcript (XIST) is well known as a factor that regulates the physiological process of chromosome dosage compensation for females. According to recent studies, lncRNA XIST is involved in a variety of cellular processes, including apoptosis, proliferation, invasion, metastasis, oxidative stress and inflammation, through molecular networks with microRNAs and their downstream targets in neoplastic and non-neoplastic diseases. Because these cellular processes play a role in the pathogenesis of cardiovascular diseases, we aim to investigate the role that lncRNA XIST plays in this process. Additionally, we wish to determine whether it is a prognostic factor or a potential therapeutic target in these diseases.
Collapse
Affiliation(s)
- Habib Haybar
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ehsan Sarbazjoda
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz,Iran
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Daryush Purrahman
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz,Iran
| | - Mohammad Reza Mahmoudian-Sani
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz,Iran
| | - Najmaldin Saki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz,Iran
| |
Collapse
|
9
|
Zhang L, Xie F, Zhang F, Lu B. The potential roles of exosomes in pathological cardiomyocyte hypertrophy mechanisms and therapy: A review. Medicine (Baltimore) 2024; 103:e37994. [PMID: 38669371 PMCID: PMC11049793 DOI: 10.1097/md.0000000000037994] [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: 01/07/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
Pathological cardiac hypertrophy, characterized by the enlargement of cardiac muscle cells, leads to serious cardiac conditions and stands as a major global health issue. Exosomes, comprising small lipid bilayer vesicles, are produced by various cell types and found in numerous bodily fluids. They play a pivotal role in intercellular communication by transferring bioactive cargos to recipient cells or activating signaling pathways in target cells. Exosomes from cardiomyocytes, endothelial cells, fibroblasts, and stem cells are key in regulating processes like cardiac hypertrophy, cardiomyocyte survival, apoptosis, fibrosis, and angiogenesis within the context of cardiovascular diseases. This review delves into exosomes' roles in pathological cardiac hypertrophy, first elucidating their impact on cell communication and signaling pathways. It then advances to discuss how exosomes affect key hypertrophic processes, including metabolism, fibrosis, oxidative stress, and angiogenesis. The review culminates by evaluating the potential of exosomes as biomarkers and their significance in targeted therapeutic strategies, thus emphasizing their critical role in the pathophysiology and management of cardiac hypertrophy.
Collapse
Affiliation(s)
- Lijun Zhang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Fang Xie
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Fengmei Zhang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Beiyao Lu
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Liu W, Higashikuni Y. Self-DNA sensing in cigarette smoke-induced vascular inflammation: the role of mitochondrial DNA release in vascular endothelial cells. Hypertens Res 2024; 47:799-802. [PMID: 38114653 DOI: 10.1038/s41440-023-01545-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 11/14/2023] [Indexed: 12/21/2023]
Affiliation(s)
- Wenhao Liu
- Department of Cardiovascular Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yasutomi Higashikuni
- Department of Cardiovascular Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
11
|
Zhang H, Dhalla NS. The Role of Pro-Inflammatory Cytokines in the Pathogenesis of Cardiovascular Disease. Int J Mol Sci 2024; 25:1082. [PMID: 38256155 PMCID: PMC10817020 DOI: 10.3390/ijms25021082] [Citation(s) in RCA: 98] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
With cardiovascular disease (CVD) being a primary source of global morbidity and mortality, it is crucial that we understand the molecular pathophysiological mechanisms at play. Recently, numerous pro-inflammatory cytokines have been linked to several different CVDs, which are now often considered an adversely pro-inflammatory state. These cytokines most notably include interleukin-6 (IL-6),tumor necrosis factor (TNF)α, and the interleukin-1 (IL-1) family, amongst others. Not only does inflammation have intricate and complex interactions with pathophysiological processes such as oxidative stress and calcium mishandling, but it also plays a role in the balance between tissue repair and destruction. In this regard, pre-clinical and clinical evidence has clearly demonstrated the involvement and dynamic nature of pro-inflammatory cytokines in many heart conditions; however, the clinical utility of the findings so far remains unclear. Whether these cytokines can serve as markers or risk predictors of disease states or act as potential therapeutic targets, further extensive research is needed to fully understand the complex network of interactions that these molecules encompass in the context of heart disease. This review will highlight the significant advances in our understanding of the contributions of pro-inflammatory cytokines in CVDs, including ischemic heart disease (atherosclerosis, thrombosis, acute myocardial infarction, and ischemia-reperfusion injury), cardiac remodeling (hypertension, cardiac hypertrophy, cardiac fibrosis, cardiac apoptosis, and heart failure), different cardiomyopathies as well as ventricular arrhythmias and atrial fibrillation. In addition, this article is focused on discussing the shortcomings in both pathological and therapeutic aspects of pro-inflammatory cytokines in CVD that still need to be addressed by future studies.
Collapse
Affiliation(s)
- Hannah Zhang
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
12
|
Xu Z, Zhou H, Zhang Y, Cheng Z, Wan M, Qin W, Li P, Feng J, Shao S, Xue W, Guo H, Liu B. Recent pharmacological advances in the treatment of cardiovascular events with Astragaloside IV. Biomed Pharmacother 2023; 168:115752. [PMID: 37875045 DOI: 10.1016/j.biopha.2023.115752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 10/26/2023] Open
Abstract
Cardiovascular disease (CVD) remains the leading cause of death and disability globally. A wide range of CVDs have been reported, each of which diverges significantly, exhibiting sophisticated types of pathogenesis (e.g., inflammatory, oxidative stress, and disorders in cardiomyocyte metabolism). Compared with conventional treatments in modern medicine, traditional Chinese medicine (TCM) can exhibit comparative advantages in the treatment of CVDs. TCM can be utilized to develop effective strategies for addressing the challenges of CVD, with fewer side effects and higher therapeutic efficiency. Astragaloside IV (AS-IV) has been confirmed as one of the major active ingredients found in Astragalus membranaceus (a Chinese herbal medicine that has been extensively employed clinically for the treatments of CVDs). Since recent studies have shown that AS-IV in CVD treatments has achieved promising results, the substance has aroused great attention and further discussions in the field. The present review aims to summarize the recent pharmacological advances in employing AS-IV in the treatment of CVDs.
Collapse
Affiliation(s)
- Zehui Xu
- Department of Anatomy, College of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Houle Zhou
- Department of Anatomy, College of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yihan Zhang
- Department of Anatomy, College of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ziji Cheng
- Department of Anatomy, College of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Melisandre Wan
- Department of Anatomy, College of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wanting Qin
- Department of Anatomy, College of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Peiyu Li
- Department of Anatomy, College of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiaming Feng
- Department of Anatomy, College of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shuijin Shao
- Department of Anatomy, College of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenlong Xue
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| | - Haidong Guo
- Department of Anatomy, College of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Baonian Liu
- Department of Anatomy, College of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
13
|
Yang C, Pan Q, Ji K, Tian Z, Zhou H, Li S, Luo C, Li J. Review on the protective mechanism of astragaloside IV against cardiovascular diseases. Front Pharmacol 2023; 14:1187910. [PMID: 37251311 PMCID: PMC10213926 DOI: 10.3389/fphar.2023.1187910] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/03/2023] [Indexed: 05/31/2023] Open
Abstract
Cardiovascular disease is a global health problem. Astragaloside IV (AS-IV) is a saponin compound extracted from the roots of the Chinese herb Astragalus. Over the past few decades, AS-IV has been shown to possess various pharmacological properties. It can protect the myocardium through antioxidative stress, anti-inflammatory effects, regulation of calcium homeostasis, improvement of myocardial energy metabolism, anti-apoptosis, anti-cardiomyocyte hypertrophy, anti-myocardial fibrosis, regulation of myocardial autophagy, and improvement of myocardial microcirculation. AS-IV exerts protective effects on blood vessels. For example, it can protect vascular endothelial cells through antioxidative stress and anti-inflammatory pathways, relax blood vessels, stabilize atherosclerotic plaques, and inhibit the proliferation and migration of vascular smooth muscle cells. Thus, the bioavailability of AS-IV is low. Toxicology indicates that AS-IV is safe, but should be used cautiously in pregnant women. In this paper, we review the mechanisms of AS-IV prevention and treatment of cardiovascular diseases in recent years to provide a reference for future research and drug development.
Collapse
Affiliation(s)
- Chunkun Yang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qingquan Pan
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Kui Ji
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Zhuang Tian
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Hongyuan Zhou
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Shuanghong Li
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Chuanchao Luo
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Jun Li
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
14
|
Qian J, Liang S, Wang Q, Xu J, Huang W, Wu G, Liang G. Toll-like receptor-2 in cardiomyocytes and macrophages mediates isoproterenol-induced cardiac inflammation and remodeling. FASEB J 2023; 37:e22740. [PMID: 36583707 DOI: 10.1096/fj.202201345r] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/28/2022] [Accepted: 12/16/2022] [Indexed: 12/31/2022]
Abstract
Heart failure (HF) is the leading cause of morbidity and mortality worldwide. Activation of the innate immune system initiates an inflammatory response during cardiac remodeling induced by isoproterenol (ISO). Here, we investigated whether Toll-like receptor-2 (TLR2) mediates ISO-induced inflammation, hypertrophy, and fibrosis. TLR2 was found to be increased in the heart tissues of mouse with HF under ISO challenge. Further, cardiomyocytes and macrophages were identified as the main cellular sources of the increased TLR2 levels in the model under ISO stimulation. The effect of TLR2 deficiency on ISO-induced cardiac remodeling was determined using TLR2 knockout mice and bone marrow transplantation models. In vitro studies involving ISO-treated cultured cardiomyocytes and macrophages showed that TLR2 knockdown significantly decreased ISO-induced cell inflammation and remodeling via MAPKs/NF-κB signaling. Mechanistically, ISO significantly increased the TLR2-MyD88 interaction in the above cells in a TLR1-dependent manner. Finally, DAMPs, such as HSP70 and fibronectin 1 (FN1), were found to be released from the cells under ISO stimulation, which further activated TLR1/2-Myd88 signaling and subsequently activated pro-inflammatory cytokine expression and cardiac remodeling. In summary, our findings suggest that TLR2 may be a target for the alleviation of chronic adrenergic stimulation-associated HF. In addition, this paper points out the possibility of TLR2 as a new target for heart failure under ISO stimulation.
Collapse
Affiliation(s)
- Jinfu Qian
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shiqi Liang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Qinyan Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jiachen Xu
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weijian Huang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Gaojun Wu
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guang Liang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
15
|
Higashikuni Y, Liu W, Numata G, Tanaka K, Fukuda D, Tanaka Y, Hirata Y, Imamura T, Takimoto E, Komuro I, Sata M. NLRP3 Inflammasome Activation Through Heart-Brain Interaction Initiates Cardiac Inflammation and Hypertrophy During Pressure Overload. Circulation 2023; 147:338-355. [PMID: 36440584 DOI: 10.1161/circulationaha.122.060860] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Mechanical stress on the heart, such as high blood pressure, initiates inflammation and causes hypertrophic heart disease. However, the regulatory mechanism of inflammation and its role in the stressed heart remain unclear. IL-1β (interleukin-1β) is a proinflammatory cytokine that causes cardiac hypertrophy and heart failure. Here, we show that neural signals activate the NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing 3) inflammasome for IL-1β production to induce adaptive hypertrophy in the stressed heart. METHODS C57BL/6 mice, knockout mouse strains for NLRP3 and P2RX7 (P2X purinoceptor 7), and adrenergic neuron-specific knockout mice for SLC17A9, a secretory vesicle protein responsible for the storage and release of ATP, were used for analysis. Pressure overload was induced by transverse aortic constriction. Various animal models were used, including pharmacological treatment with apyrase, lipopolysaccharide, 2'(3')-O-(4-benzoylbenzoyl)-ATP, MCC950, anti-IL-1β antibodies, clonidine, pseudoephedrine, isoproterenol, and bisoprolol, left stellate ganglionectomy, and ablation of cardiac afferent nerves with capsaicin. Cardiac function and morphology, gene expression, myocardial IL-1β and caspase-1 activity, and extracellular ATP level were assessed. In vitro experiments were performed using primary cardiomyocytes and fibroblasts from rat neonates and human microvascular endothelial cell line. Cell surface area and proliferation were assessed. RESULTS Genetic disruption of NLRP3 resulted in significant loss of IL-1β production, cardiac hypertrophy, and contractile function during pressure overload. A bone marrow transplantation experiment revealed an essential role of NLRP3 in cardiac nonimmune cells in myocardial IL-1β production and cardiac phenotype. Pharmacological depletion of extracellular ATP or genetic disruption of the P2X7 receptor suppressed myocardial NLRP3 inflammasome activity during pressure overload, indicating an important role of ATP/P2X7 axis in cardiac inflammation and hypertrophy. Extracellular ATP induced hypertrophic changes of cardiac cells in an NLRP3- and IL-1β-dependent manner in vitro. Manipulation of the sympathetic nervous system suggested sympathetic efferent nerves as the main source of extracellular ATP. Depletion of ATP release from sympathetic efferent nerves, ablation of cardiac afferent nerves, or a lipophilic β-blocker reduced cardiac extracellular ATP level, and inhibited NLRP3 inflammasome activation, IL-1β production, and adaptive cardiac hypertrophy during pressure overload. CONCLUSIONS Cardiac inflammation and hypertrophy are regulated by heart-brain interaction. Controlling neural signals might be important for the treatment of hypertensive heart disease.
Collapse
Affiliation(s)
- Yasutomi Higashikuni
- Department of Cardiovascular Medicine (Y.H., W.L., G.N., K. Tanaka, T.I., E.T., I.K.), The University of Tokyo, Japan
| | - Wenhao Liu
- Department of Cardiovascular Medicine (Y.H., W.L., G.N., K. Tanaka, T.I., E.T., I.K.), The University of Tokyo, Japan
| | - Genri Numata
- Department of Cardiovascular Medicine (Y.H., W.L., G.N., K. Tanaka, T.I., E.T., I.K.), The University of Tokyo, Japan
| | - Kimie Tanaka
- Department of Cardiovascular Medicine (Y.H., W.L., G.N., K. Tanaka, T.I., E.T., I.K.), The University of Tokyo, Japan.,Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan (K. Tanaka)
| | - Daiju Fukuda
- Department of Cardiovascular Medicine, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan (D.F.)
| | - Yu Tanaka
- Department of Pediatrics (Y. Tanaka, Y.H.), The University of Tokyo, Japan
| | - Yoichiro Hirata
- Department of Pediatrics (Y. Tanaka, Y.H.), The University of Tokyo, Japan
| | - Teruhiko Imamura
- Department of Cardiovascular Medicine (Y.H., W.L., G.N., K. Tanaka, T.I., E.T., I.K.), The University of Tokyo, Japan.,Second Department of Medicine, University of Toyama, Japan (T.I.)
| | - Eiki Takimoto
- Department of Cardiovascular Medicine (Y.H., W.L., G.N., K. Tanaka, T.I., E.T., I.K.), The University of Tokyo, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine (Y.H., W.L., G.N., K. Tanaka, T.I., E.T., I.K.), The University of Tokyo, Japan
| | - Masataka Sata
- Department of Cardiovascular Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Japan (M.S.)
| |
Collapse
|
16
|
Zhang Y, Wu J, Dong E, Wang Z, Xiao H. Toll-like receptors in cardiac hypertrophy. Front Cardiovasc Med 2023; 10:1143583. [PMID: 37113698 PMCID: PMC10126280 DOI: 10.3389/fcvm.2023.1143583] [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: 01/13/2023] [Accepted: 03/24/2023] [Indexed: 04/29/2023] Open
Abstract
Toll-like receptors (TLRs) are a family of pattern recognition receptors (PRRs) that can identify pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). TLRs play an important role in the innate immune response, leading to acute and chronic inflammation. Cardiac hypertrophy, an important cardiac remodeling phenotype during cardiovascular disease, contributes to the development of heart failure. In previous decades, many studies have reported that TLR-mediated inflammation was involved in the induction of myocardium hypertrophic remodeling, suggesting that targeting TLR signaling might be an effective strategy against pathological cardiac hypertrophy. Thus, it is necessary to study the mechanisms underlying TLR functions in cardiac hypertrophy. In this review, we summarized key findings of TLR signaling in cardiac hypertrophy.
Collapse
Affiliation(s)
- Yanan Zhang
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University Third Hospital, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, China
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Department of Clinical Laboratory, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Jimin Wu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University Third Hospital, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, China
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Erdan Dong
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University Third Hospital, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, China
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhanli Wang
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
- Department of Clinical Laboratory, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Correspondence: Zhanli Wang Han Xiao
| | - Han Xiao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University Third Hospital, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, China
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Correspondence: Zhanli Wang Han Xiao
| |
Collapse
|
17
|
Nakamura S, Numata G, Yamaguchi T, Tokiwa H, Higashikuni Y, Nomura S, Sasano T, Takimoto E, Komuro I. Endoplasmic reticulum stress-activated nuclear factor-kappa B signaling pathway induces the upregulation of cardiomyocyte dopamine D1 receptor in heart failure. Biochem Biophys Res Commun 2022; 637:247-253. [DOI: 10.1016/j.bbrc.2022.11.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022]
|
18
|
Wei Y, Cai J, Zhu R, Xu K, Li H, Li J. Function and therapeutic potential of transient receptor potential ankyrin 1 in fibrosis. Front Pharmacol 2022; 13:1014041. [PMID: 36278189 PMCID: PMC9582847 DOI: 10.3389/fphar.2022.1014041] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/26/2022] [Indexed: 11/18/2022] Open
Abstract
The transient receptor potential (TRP) protein superfamily is a special group of cation channels expressed in different cell types and signaling pathways. In this review, we focus on TRPA1 (transient receptor potential ankyrin 1), an ion channel in this family that exists in the cell membrane and shows a different function from other TRP channels. TRPA1 usually has a special activation effect that can induce cation ions, especially calcium ions, to flow into activated cells. In this paper, we review the role of TRPA1 in fibroblasts. To clarify the relationship between fibroblasts and TRPA1, we have also paid special attention to the interactions between TRPA1 and inflammatory factors leading to fibroblast activation. TRPA1 has different functions in the fibrosis process in different organs, and there have also been interesting discussions of the mechanism of TRPA1 in fibroblasts. Therefore, this review aims to describe the function of TRP channels in controlling fibrosis through fibroblasts in different organ inflammatory and immune-mediated diseases. We attempt to prove that TRPA1 is a target for fibrosis. In fact, some clinical trials have already proven that TRPA1 is a potential adjuvant therapy for treating fibrosis.
Collapse
Affiliation(s)
- Yicheng Wei
- Third Affiliated Hospital of Shanghai University/Wenzhou People’s Hospital, Wenzhou, China
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jialuo Cai
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Ruiqiu Zhu
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ke Xu
- Musculoskeletal Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- Wenzhou Institute of Shanghai University, Wenzhou, China
- *Correspondence: Ke Xu, , ; Hongchang Li, ; Jianxin Li,
| | - Hongchang Li
- Department of General Surgery, Institute of Fudan–Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
- *Correspondence: Ke Xu, , ; Hongchang Li, ; Jianxin Li,
| | - Jianxin Li
- Third Affiliated Hospital of Shanghai University/Wenzhou People’s Hospital, Wenzhou, China
- *Correspondence: Ke Xu, , ; Hongchang Li, ; Jianxin Li,
| |
Collapse
|
19
|
Belmadani S, Matrougui K. Role of High Mobility Group Box 1 in Cardiovascular Diseases. Inflammation 2022; 45:1864-1874. [PMID: 35386038 PMCID: PMC11145736 DOI: 10.1007/s10753-022-01668-3] [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: 10/13/2021] [Revised: 02/27/2022] [Accepted: 03/28/2022] [Indexed: 11/05/2022]
Abstract
High Mobility Group Box 1 (HMGB1) is a ubiquitous, highly conserved nuclear and cytosolic protein that has diverse biological roles depending on its cellular location and posttranslational modifications. The HMGB1 is localized in the nucleus but can be translocated to the cytoplasm to modulate the intracellular signaling and eventually secreted outside the cells. It is widely established that HMGB1 plays a key role in inflammation; however, the role of HMGB1 in the cardiovascular diseases is not well understood. In this review, we will discuss the latest reports on the pathophysiological link between HMGB1 and cardiovascular complications, with special emphasis on the inflammation. Thus, the understanding of the role of HMGB1 may provide new insights into developing new HMGB1-based therapies.
Collapse
Affiliation(s)
- Souad Belmadani
- Department of Physiological Sciences, EVMS, Norfolk, Virginia, 23501, USA
| | - Khalid Matrougui
- Department of Physiological Sciences, EVMS, Norfolk, Virginia, 23501, USA.
| |
Collapse
|
20
|
Tumor Necrosis Factor-α-Induced Protein 8-Like 2 Ameliorates Cardiac Hypertrophy by Targeting TLR4 in Macrophages. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9469143. [PMID: 35528518 PMCID: PMC9072033 DOI: 10.1155/2022/9469143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/04/2022] [Indexed: 11/28/2022]
Abstract
Background Tumor necrosis factor-α-induced protein 8-like 2 (TIPE2), a novel immunoregulatory protein, has been reported to regulate inflammation and apoptosis. The role of TIPE2 in cardiovascular disease, especially cardiac hypertrophy, has not been elucidated. Thus, the aim of the present study was to explore the role of TIPE2 in cardiac hypertrophy. Methods Mice were subjected to aortic banding (AB) to induce an adverse hypertrophic model. To overexpress TIPE2, mice were injected with a lentiviral vector expressing TIPE2. Echocardiographic and hemodynamic analyses were used to evaluate cardiac function. Neonatal rat cardiomyocytes (NRCMs) and mouse peritoneal macrophages (MPMs) were isolated and stimulated with angiotensin II. NRCMs and MPM were also cocultured and stimulated with angiotensin II. Cells were transfected with Lenti-TIPE2 to overexpress TIPE2. Results TIPE2 expression levels were downregulated in hypertrophic mouse hearts and in macrophages in heart tissue. TIPE2 overexpression attenuated pressure overload-induced cardiac hypertrophy, fibrosis, and cardiac dysfunction. Moreover, we found that TIPE2 overexpression in neonatal cardiomyocytes did not relieve the angiotensin II-induced hypertrophic response in vitro. Furthermore, TIPE2 overexpression downregulated TLR4 and NF-κB signaling in macrophages but not in cardiomyocytes, which led to diminished inflammation in macrophages and consequently reduced the activation of hypertrophic Akt signaling in cardiomyocytes. TLR4 inhibition by TAK-242 did not enhance the antihypertrophic effect of TIPE2 overexpression. Conclusions The present study indicated that TIPE2 represses macrophage activation by targeting TLR4, subsequently inhibiting cardiac hypertrophy.
Collapse
|
21
|
Theall B, Alcaide P. The heart under pressure: immune cells in fibrotic remodeling. CURRENT OPINION IN PHYSIOLOGY 2022; 25:100484. [PMID: 35224321 PMCID: PMC8881013 DOI: 10.1016/j.cophys.2022.100484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The complex syndrome of heart failure (HF) is characterized by increased left ventricular pressures. Cardiomyocytes increase in size, cardiac fibroblasts transform and make extracellular matrix, and leukocytes infiltrate the cardiac tissue and alter cardiomyocyte and cardiac fibroblast function. Here we review recent advances in our understanding of the cellular composition of the heart during homeostasis and in response to cardiac pressure overload, with an emphasis on immune cell communication with cardiac fibroblasts and its consequences in cardiac remodeling.
Collapse
Affiliation(s)
- Brandon Theall
- Department of Immunology, Tufts University School of Medicine, Boston, MA,Immunology Program, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA
| | - Pilar Alcaide
- Department of Immunology, Tufts University School of Medicine, Boston, MA,Immunology Program, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA
| |
Collapse
|
22
|
Wang Y, Wu J, Wang D, Yang R, Liu Q. Traditional Chinese Medicine Targeting Heat Shock Proteins as Therapeutic Strategy for Heart Failure. Front Pharmacol 2022; 12:814243. [PMID: 35115946 PMCID: PMC8804377 DOI: 10.3389/fphar.2021.814243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 12/21/2021] [Indexed: 11/13/2022] Open
Abstract
Heart failure (HF) is the terminal stage of multifarious heart diseases and is responsible for high hospitalization rates and mortality. Pathophysiological mechanisms of HF include cardiac hypertrophy, remodeling and fibrosis resulting from cell death, inflammation and oxidative stress. Heat shock proteins (HSPs) can ameliorate folding of proteins, maintain protein structure and stability upon stress, protect the heart from cardiac dysfunction and ameliorate apoptosis. Traditional Chinese medicine (TCM) regulates expression of HSPs and has beneficial therapeutic effect in HF. In this review, we summarized the function of HSPs in HF and the role of TCM in regulating expression of HSPs. Studying the regulation of HSPs by TCM will provide novel ideas for the study of the mechanism and treatment of HF.
Collapse
Affiliation(s)
- Yanchun Wang
- Shenyang the Tenth People’s Hospital, Shenyang, China
| | - Junxuan Wu
- Shunde Hospital of Guangzhou University of Chinese Medicine, Foshan, China
| | - Dawei Wang
- Shunde Hospital of Guangzhou University of Chinese Medicine, Foshan, China
- *Correspondence: Qing Liu, ; Dawei Wang, ; Rongyuan Yang,
| | - Rongyuan Yang
- The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, Zhuhai, China
- *Correspondence: Qing Liu, ; Dawei Wang, ; Rongyuan Yang,
| | - Qing Liu
- The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, Zhuhai, China
- *Correspondence: Qing Liu, ; Dawei Wang, ; Rongyuan Yang,
| |
Collapse
|
23
|
Higashikuni Y, Liu W, Sata M. Give a Leg Up: Screening for Peripheral Artery Disease after Acute Myocardial Infarction. J Atheroscler Thromb 2021; 29:989-991. [PMID: 34853214 PMCID: PMC9252614 DOI: 10.5551/jat.ed186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
| | - Wenhao Liu
- Department of Cardiovascular Medicine, The University of Tokyo
| | - Masataka Sata
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| |
Collapse
|
24
|
Kim YJ, Granstein RD. Roles of calcitonin gene-related peptide in the skin, and other physiological and pathophysiological functions. Brain Behav Immun Health 2021; 18:100361. [PMID: 34746878 PMCID: PMC8551410 DOI: 10.1016/j.bbih.2021.100361] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 10/01/2021] [Accepted: 10/02/2021] [Indexed: 01/05/2023] Open
Abstract
Skin immunity is regulated by many mediator molecules. One is the neuropeptide calcitonin gene-related peptide (CGRP). CGRP has roles in regulating the function of components of the immune system including T cells, B cells, dendritic cells (DCs), endothelial cells (ECs), and mast cells (MCs). Herein we discuss actions of CGRP in mediating inflammatory and vascular effects in various cutaneous models and disorders. CGRP can help to recruit immune cells through endothelium-dependent vasodilation. CGRP plays an important role in the pathogenesis of neurogenic inflammation. Functions of many components in the immune system are influenced by CGRP. CGRP regulates various inflammatory processes in human skin by affecting different cell-types.
Collapse
Affiliation(s)
- Yee Jung Kim
- Department of Dermatology, Weill Cornell Medicine, 1305 York Avenue, WGC9, New York, NY, 10021, USA
| | - Richard D Granstein
- Department of Dermatology, Weill Cornell Medicine, 1305 York Avenue, WGC9, New York, NY, 10021, USA
| |
Collapse
|
25
|
Higashikuni Y, Liu W, Obana T, Sata M. Pathogenic Basis of Thromboinflammation and Endothelial Injury in COVID-19: Current Findings and Therapeutic Implications. Int J Mol Sci 2021; 22:ijms222112081. [PMID: 34769508 PMCID: PMC8584434 DOI: 10.3390/ijms222112081] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a global pandemic with a great impact on social and economic activities, as well as public health. In most patients, the symptoms of COVID-19 are a high-grade fever and a dry cough, and spontaneously resolve within ten days. However, in severe cases, COVID-19 leads to atypical bilateral interstitial pneumonia, acute respiratory distress syndrome, and systemic thromboembolism, resulting in multiple organ failure with high mortality and morbidity. SARS-CoV-2 has immune evasion mechanisms, including inhibition of interferon signaling and suppression of T cell and B cell responses. SARS-CoV-2 infection directly and indirectly causes dysregulated immune responses, platelet hyperactivation, and endothelial dysfunction, which interact with each other and are exacerbated by cardiovascular risk factors. In this review, we summarize current knowledge on the pathogenic basis of thromboinflammation and endothelial injury in COVID-19. We highlight the distinct contributions of dysregulated immune responses, platelet hyperactivation, and endothelial dysfunction to the pathogenesis of COVID-19. In addition, we discuss potential therapeutic strategies targeting these mechanisms.
Collapse
Affiliation(s)
- Yasutomi Higashikuni
- Department of Cardiovascular Medicine, The University of Tokyo, Tokyo 113-8655, Japan; (W.L.); (T.O.)
- Correspondence: (Y.H.); (M.S.)
| | - Wenhao Liu
- Department of Cardiovascular Medicine, The University of Tokyo, Tokyo 113-8655, Japan; (W.L.); (T.O.)
| | - Takumi Obana
- Department of Cardiovascular Medicine, The University of Tokyo, Tokyo 113-8655, Japan; (W.L.); (T.O.)
| | - Masataka Sata
- Department of Cardiovascular Medicine, The University of Tokushima, Tokushima 770-8503, Japan
- Correspondence: (Y.H.); (M.S.)
| |
Collapse
|
26
|
Zhao M, Zhang J, Xu Y, Liu J, Ye J, Wang Z, Ye D, Feng Y, Xu S, Pan W, Wang M, Wan J. Selective Inhibition of NLRP3 Inflammasome Reverses Pressure Overload-Induced Pathological Cardiac Remodeling by Attenuating Hypertrophy, Fibrosis, and Inflammation. Int Immunopharmacol 2021; 99:108046. [PMID: 34435581 DOI: 10.1016/j.intimp.2021.108046] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 07/25/2021] [Accepted: 08/01/2021] [Indexed: 12/24/2022]
Abstract
Activation of the NLRP3 inflammasome promotes pathological cardiac remodeling induced by pressure overload. However, the therapeutic effects of NLRP3 inhibition after cardiac remodeling remain unknown. The present study aimed to investigate whether the selective NLRP3 inhibitor, MCC950, could reverse transverse aortic constriction (TAC)-induced cardiac remodeling. Mice were divided into four groups based on the treatment given: sham, sham + MCC950, TAC, and TAC + MCC950. MCC950 (10 mg/kg, intraperitoneal injection, once per day) was administered from two weeks after TAC or sham surgery for four weeks. Echocardiography, histological analysis, RT-PCR, and Western blotting were performed to explore the function of MCC950 after TAC. We found that MCC950 reversed cardiac dysfunction after TAC. MCC950 attenuated cardiac hypertrophy by down-regulating calcineurin expression and inhibiting MAPK activation. Further, it also alleviated cardiac fibrosis post-TAC by inhibiting the TGF-β/Smad4 pathway, and reduced cardiac inflammation and macrophage infiltration post-TAC, including both M1 and M2 macrophages. Taken together, MCC950 can attenuate cardiac remodeling due to pressure overload by inhibiting hypertrophy, fibrosis, and inflammation. Our study provides a basis for the clinical application of NLRP3 inhibitors in the treatment of non-ischemic heart failure.
Collapse
Affiliation(s)
- Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Zhen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Yongqi Feng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Shuwan Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Wei Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China.
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China.
| |
Collapse
|
27
|
Ye S, Lin K, Wu G, Xu MJ, Shan P, Huang W, Wang Y, Liang G. Toll-like receptor 2 signaling deficiency in cardiac cells ameliorates Ang II-induced cardiac inflammation and remodeling. Transl Res 2021; 233:62-76. [PMID: 33652137 DOI: 10.1016/j.trsl.2021.02.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/05/2021] [Accepted: 02/23/2021] [Indexed: 11/28/2022]
Abstract
Activation of the innate immune system represents a vital step in inflammation during cardiac remodeling induced by the angiotensin II (Ang II). This study aimed to explore the role of Toll-like receptors 2 (TLR2) in Ang II-induced cardiac remodeling. We investigated the effect of TLR2 deficiency on Ang II-induced cardiac remodeling by utilizing TLR2 knockout mice, bone marrow transplantation models, and H9C2 cells. Though TLR2 deficiency had no effect on body weight change, cardiac Ang II content and blood pressure, it significantly ameliorated cardiac hypertrophy, fibrosis and inflammation, as well as improved heart function. Further bone marrow transplantation studies showed that TLR2-deficiency in cardiac cells but not bone marrow-derived cells prevented Ang II-induced cardiac remodeling and cardiac dysfunction. The underlying mechanism may involve increased TLR2-MyD88 interaction. Further in vitro studies in Ang II-treated H9C2 cells showed that TLR2 knockdown by siRNA significantly decreased Ang II-induced cell hypertrophy, fibrosis and inflammation. Moreover, Ang II significantly increased TLR2-MyD88 interaction in H9C2 cells in a TLR4-independent manner. TLR2 deficiency in cardiac cells prevents Ang II-induced cardiac remodeling, inflammation and dysfunction through reducing the formation of TLR2-MyD88 complexes. Inhibition of TLR2 pathway may be a therapeutic strategy of hypertensive heart failure.
Collapse
Affiliation(s)
- Shiju Ye
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ke Lin
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gaojun Wu
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ming-Jiang Xu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Peiren Shan
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weijian Huang
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhuji Biomedicine Institute, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhuji, Zhejiang, China.
| | - Guang Liang
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhuji Biomedicine Institute, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhuji, Zhejiang, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
28
|
de Souza DMS, Silva MC, Farias SEB, Menezes APDJ, Milanezi CM, Lúcio KDP, Paiva NCN, de Abreu PM, Costa DC, Pinto KMDC, Costa GDP, Silva JS, Talvani A. Diet Rich in Lard Promotes a Metabolic Environment Favorable to Trypanosoma cruzi Growth. Front Cardiovasc Med 2021; 8:667580. [PMID: 34113663 PMCID: PMC8185140 DOI: 10.3389/fcvm.2021.667580] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 04/15/2021] [Indexed: 01/24/2023] Open
Abstract
Background: Trypanosoma cruzi is a protozoan parasite that causes Chagas disease and affects 6-7 million people mainly in Latin America and worldwide. Here, we investigated the effects of hyperlipidic diets, mainly composed of olive oil or lard on experimental T. cruzi infection. C57BL/6 mice were fed two different dietary types in which the main sources of fatty acids were either monounsaturated (olive oil diet) or saturated (lard diet). Methods: After 60 days on the diet, mice were infected with 50 trypomastigote forms of T. cruzi Colombian strain. We evaluated the systemic and tissue parasitism, tissue inflammation, and the redox status of mice after 30 days of infection. Results: Lipid levels in the liver of mice fed with the lard diet increased compared with that of the mice fed with olive oil or normolipidic diets. The lard diet group presented with an increased parasitic load in the heart and adipose tissues following infection as well as an increased expression of Tlr2 and Tlr9 in the heart. However, no changes were seen in the survival rates across the dietary groups. Infected mice receiving all diets presented comparable levels of recruited inflammatory cells at 30 days post-infection but, at this time, we observed lard diet inducing an overproduction of CCL2 in the cardiac tissue and its inhibition in the adipose tissue. T. cruzi infection altered liver antioxidant levels in mice, with the lard diet group demonstrating decreased catalase (CAT) activity compared with that of other dietary groups. Conclusions: Our data demonstrated that T. cruzi growth is more favorable on tissue of mice subjected to the lard diet. Our findings supported our hypothesis of a relationship between the source of dietary lipids and parasite-induced immunopathology.
Collapse
Affiliation(s)
- Débora Maria Soares de Souza
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil.,Biological Science Post-graduate Program, Federal University of Ouro Preto, Ouro Preto, Brazil.,Health and Nutrition Post-graduate Program, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Maria Cláudia Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Silvia Elvira Barros Farias
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Ana Paula de J Menezes
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Cristiane Maria Milanezi
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Karine de P Lúcio
- Laboratory of Metabolic Biochemistry, Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Nívia Carolina N Paiva
- Center of Research in Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Paula Melo de Abreu
- Biological Science Post-graduate Program, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Daniela Caldeira Costa
- Health and Nutrition Post-graduate Program, Federal University of Ouro Preto, Ouro Preto, Brazil.,Laboratory of Metabolic Biochemistry, Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Kelerson Mauro de Castro Pinto
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil.,School of Physical Education, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Guilherme de Paula Costa
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil.,Health and Nutrition Post-graduate Program, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - João Santana Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil.,Fiocruz-Bi-Institutional Translational Medicine Plataform, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - André Talvani
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil.,Health and Nutrition Post-graduate Program, Federal University of Ouro Preto, Ouro Preto, Brazil.,Health Science, Infectology and Tropical Medicine Post-graduate Program, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
29
|
Ngwenyama N, Kirabo A, Aronovitz M, Velázquez F, Carrillo-Salinas F, Salvador AM, Nevers T, Amarnath V, Tai A, Blanton RM, Harrison DG, Alcaide P. Isolevuglandin-Modified Cardiac Proteins Drive CD4+ T-Cell Activation in the Heart and Promote Cardiac Dysfunction. Circulation 2021; 143:1242-1255. [PMID: 33463362 PMCID: PMC7987774 DOI: 10.1161/circulationaha.120.051889] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/11/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Despite the well-established association between T-cell-mediated inflammation and nonischemic heart failure, the specific mechanisms triggering T-cell activation during the progression of heart failure and the antigens involved are poorly understood. We hypothesized that myocardial oxidative stress induces the formation of isolevuglandin (IsoLG)-modified proteins that function as cardiac neoantigens to elicit CD4+ T-cell receptor (TCR) activation and promote heart failure. METHODS We used transverse aortic constriction in mice to trigger myocardial oxidative stress and T-cell infiltration. We profiled the TCR repertoire by mRNA sequencing of intramyocardial activated CD4+ T cells in Nur77GFP reporter mice, which transiently express GFP on TCR engagement. We assessed the role of antigen presentation and TCR specificity in the development of cardiac dysfunction using antigen presentation-deficient MhcII-/- mice and TCR transgenic OTII mice that lack specificity for endogenous antigens. We detected IsoLG protein adducts in failing human hearts. We also evaluated the role of reactive oxygen species and IsoLGs in eliciting T-cell immune responses in vivo by treating mice with the antioxidant TEMPOL and the IsoLG scavenger 2-hydroxybenzylamine during transverse aortic constriction, and ex vivo in mechanistic studies of CD4+ T-cell proliferation in response to IsoLG-modified cardiac proteins. RESULTS We discovered that TCR antigen recognition increases in the left ventricle as cardiac dysfunction progresses and identified a limited repertoire of activated CD4+ T-cell clonotypes in the left ventricle. Antigen presentation of endogenous antigens was required to develop cardiac dysfunction because MhcII-/- mice reconstituted with CD4+ T cells and OTII mice immunized with their cognate antigen were protected from transverse aortic constriction-induced cardiac dysfunction despite the presence of left ventricle-infiltrated CD4+ T cells. Scavenging IsoLGs with 2-hydroxybenzylamine reduced TCR activation and prevented cardiac dysfunction. Mechanistically, cardiac pressure overload resulted in reactive oxygen species-dependent dendritic cell accumulation of IsoLG protein adducts, which induced robust CD4+ T-cell proliferation. CONCLUSIONS Our study demonstrates an important role of reactive oxygen species-induced formation of IsoLG-modified cardiac neoantigens that lead to TCR-dependent CD4+ T-cell activation within the heart.
Collapse
Affiliation(s)
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Mark Aronovitz
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA
| | | | | | | | - Tania Nevers
- Department of Immunology, Tufts University, Boston, MA
| | - Venkataraman Amarnath
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN
| | - Albert Tai
- Department of Immunology, Tufts University, Boston, MA
| | - Robert M. Blanton
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA
| | - David G. Harrison
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Pilar Alcaide
- Department of Immunology, Tufts University, Boston, MA
| |
Collapse
|
30
|
Li W, Zhang Z, Li X, Cai J, Li D, Du J, Zhang B, Xiang D, Li N, Li Y. CGRP derived from cardiac fibroblasts is an endogenous suppressor of cardiac fibrosis. Cardiovasc Res 2021; 116:1335-1348. [PMID: 31504241 DOI: 10.1093/cvr/cvz234] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 07/31/2019] [Accepted: 08/26/2019] [Indexed: 12/28/2022] Open
Abstract
AIMS Aberrant activation of cardiac fibroblasts leads to cardiac fibrosis, and evolving evidences suggest that endogenous bioactive substances derived from cardiac fibroblasts regulate cardiac fibroblasts activation in an autocrine/paracrine manner. Here we first presented evidence that cardiac fibroblasts can synthesize and secrete calcitonin gene-related peptide (CGRP), therefore, this study aimed to investigate the role of cardiac fibroblasts-derived CGRP in cardiac fibroblasts activation and its regulative mechanism. METHODS AND RESULTS The abundantly expression of CGRP in rat, mouse, and human myocardium allowed us to explore the cellular origin of CGRP, and found that the cardiac CGRP was mainly derived from cardiac fibroblasts. Activating TRPA1 with a specific agonist allyl isothiocyanate promoted the synthesis and secretion of CGRP, as well as intracellular Ca2+. These effects were reversed by TRPA1-specific antagonist HC030031 and Ca2+ chelator BAPTA-AM. TGF-β1 was applied to induce the activation of cardiac fibroblasts, and found that TGF-β1 can increase the mRNA expression and secretion levels of CGRP in cardiac fibroblasts. Either CGRP8-37 (CGRP receptor antagonist) or α-CGRP small interfering RNA (siRNA) aggravated TGF-β1-induced proliferation, differentiation, collagen production, and instigated inflammation in cardiac fibroblasts. Moreover, TGF-β1-induced NF-κB activation including IκBα phosphorylation and p65 nuclear translocation were also promoted by CGRP8-37 and α-CGRP siRNA. NF-κB inhibitor pyrrolidinedithiocarbamate ammonium (PDTC) reversed the effects of CGRP8-37 on NF-κB activation. The promotive effects of CGRP8-37 on TGF-β1-induced activation of cardiac fibroblasts were all reversed by PDTC. Monocrotaline (MCT) induces pulmonary arterial hypertension, progressively leading to right ventricular fibrosis. This model of cardiac fibrosis was developed here to test the potentially beneficial effects of TRPA1 activation in vivo. The non-toxic TRPA1 agonist Cinnamaldehyde (CA) inhibited MCT-induced elevation in right ventricle systolic pressure, RV/LV + S, and right ventricular collagen accumulation, as well as down-regulation of CGRP. CA increased the synthesis and secretion of CGRP, and inhibited TGF-β1-induced activation in cardiac fibroblasts. CONCLUSION Our data suggested an autocrine role for cardiac fibroblasts-derived CGRP in suppressing activation of cardiac fibroblasts through inhibiting NF-κB activation. Increasing autocrine CGRP by activating TRPA1 can ameliorate cardiac fibrosis. These findings support the notion that CGRP derived from cardiac fibroblasts is an endogenous suppressor of cardiac fibrosis.
Collapse
Affiliation(s)
- Wenqun Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, No. 172 Tongzipo Road, Changsha, Hunan 410078, China.,Department of Pharmacy, The Second Xiangya Hospital, Central South University, No. 139 Renmin Middle Road, Changsha, Hunan 410011, China
| | - Zheng Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, No. 172 Tongzipo Road, Changsha, Hunan 410078, China
| | - Xiaohui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, No. 172 Tongzipo Road, Changsha, Hunan 410078, China
| | - Jifeng Cai
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, No. 172 Tongzipo Road, Changsha, Hunan 410013, China
| | - Dai Li
- Department of Pharmacy, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, Hunan 410078, China
| | - Jie Du
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, No. 172 Tongzipo Road, Changsha, Hunan 410078, China.,Department of Pharmacy, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, Hunan 410078, China
| | - Bikui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, No. 139 Renmin Middle Road, Changsha, Hunan 410011, China
| | - Daxiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, No. 139 Renmin Middle Road, Changsha, Hunan 410011, China
| | - Niansheng Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, No. 172 Tongzipo Road, Changsha, Hunan 410078, China
| | - Yuanjian Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, No. 172 Tongzipo Road, Changsha, Hunan 410078, China
| |
Collapse
|
31
|
Liu X, Feng Y, Liu W, Li H, Hu Z, Hu S, Ke J, Long X. Toll‐like receptor 2 mediates the degeneration of cartilage in experimental inflammatory TMJOA. Oral Dis 2020. [DOI: 10.1111/odi.13744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Xin Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of Education School & Hospital of Stomatology Wuhan University Wuhan China
| | - Yaping Feng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of Education School & Hospital of Stomatology Wuhan University Wuhan China
| | - Wen Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of Education School & Hospital of Stomatology Wuhan University Wuhan China
| | - Huimin Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of Education School & Hospital of Stomatology Wuhan University Wuhan China
| | - Zhihui Hu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of Education School & Hospital of Stomatology Wuhan University Wuhan China
| | - Shiyu Hu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of Education School & Hospital of Stomatology Wuhan University Wuhan China
| | - Jin Ke
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of Education School & Hospital of Stomatology Wuhan University Wuhan China
| | - Xing Long
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of Education School & Hospital of Stomatology Wuhan University Wuhan China
- Department of Oral and Maxillofacial Surgery School & Hospital of Stomatology Wuhan University Wuhan China
| |
Collapse
|
32
|
Toko H, Morita H, Katakura M, Hashimoto M, Ko T, Bujo S, Adachi Y, Ueda K, Murakami H, Ishizuka M, Guo J, Zhao C, Fujiwara T, Hara H, Takeda N, Takimoto E, Shido O, Harada M, Komuro I. Omega-3 fatty acid prevents the development of heart failure by changing fatty acid composition in the heart. Sci Rep 2020; 10:15553. [PMID: 32968201 PMCID: PMC7512019 DOI: 10.1038/s41598-020-72686-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/04/2020] [Indexed: 12/22/2022] Open
Abstract
Some clinical trials showed that omega-3 fatty acid (FA) reduced cardiovascular events, but it remains unknown whether omega-3 FA supplementation changes the composition of FAs and their metabolites in the heart and how the changes, if any, exert beneficial effects on cardiac structure and function. To clarify these issues, we supplied omega-3 FA to mice exposed to pressure overload, and examined cardiac structure and function by echocardiography and a proportion of FAs and their metabolites by gas chromatography and liquid chromatography-tandem mass spectrometry, respectively. Pressure overload induced cardiac hypertrophy and dysfunction, and reduced concentration of all FAs’ components and increased free form arachidonic acid and its metabolites, precursors of pro-inflammatory mediators in the heart. Omega-3 FA supplementation increased both total and free form of eicosapentaenoic acid, a precursor of pro-resolution mediators and reduced free form arachidonic acid in the heart. Omega-3 FA supplementation suppressed expressions of pro-inflammatory cytokines and the infiltration of inflammatory cells into the heart and ameliorated cardiac dysfunction and fibrosis. These results suggest that omega-3 FA-induced changes of FAs composition in the heart have beneficial effects on cardiac function via regulating inflammation.
Collapse
Affiliation(s)
- Haruhiro Toko
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan. .,Department of Advanced Translational Research and Medicine in Management of Pulmonary Hypertension, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Hiroyuki Morita
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Masanori Katakura
- Department of Environmental Physiology, Faculty of Medicine, Shimane University, 89-1 Enyacho, Izumo, Shimane, 693-8501, Japan.,Laboratory of Nutritional Physiology, Department of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Michio Hashimoto
- Department of Environmental Physiology, Faculty of Medicine, Shimane University, 89-1 Enyacho, Izumo, Shimane, 693-8501, Japan
| | - Toshiyuki Ko
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Satoshi Bujo
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yusuke Adachi
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kazutaka Ueda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Haruka Murakami
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Masato Ishizuka
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Jiaxi Guo
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Chunxia Zhao
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Takayuki Fujiwara
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hironori Hara
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Norifumi Takeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Eiki Takimoto
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Osamu Shido
- Department of Environmental Physiology, Faculty of Medicine, Shimane University, 89-1 Enyacho, Izumo, Shimane, 693-8501, Japan
| | - Mutsuo Harada
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.,Department of Advanced Clinical Science and Therapeutics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
33
|
Inhibition of the ROS-EGFR Pathway Mediates the Protective Action of Nox1/4 Inhibitor GKT137831 against Hypertensive Cardiac Hypertrophy via Suppressing Cardiac Inflammation and Activation of Akt and ERK1/2. Mediators Inflamm 2020; 2020:1078365. [PMID: 32831633 PMCID: PMC7424508 DOI: 10.1155/2020/1078365] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/08/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress, inflammation, and hypertension constitute a self-perpetuating vicious circle to exacerbate hypertension and subsequent hypertensive cardiac hypertrophy. NADPH oxidase (Nox) 1/4 inhibitor GKT137831 alleviates hypertensive cardiac hypertrophy in models of secondary hypertension; however, it remains unclear about its effect on hypertensive cardiac hypertrophy in models of essential hypertension. This study is aimed at determining the beneficial role of GKT137831 in hypertensive cardiac hypertrophy in spontaneously hypertensive rats (SHRs) and its mechanisms of action. Treating with GKT137831 prevented cardiac hypertrophy in SHRs. Likewise, decreasing production of reactive oxygen species (ROS) with GKT137831 reduced epidermal growth factor receptor (EGFR) activity in the left ventricle of SHRs. Additionally, EGFR inhibition also reduced ROS production in the left ventricle and blunted hypertensive cardiac hypertrophy in SHRs. Moreover, inhibition of the ROS-EGFR pathway with Nox1/4 inhibitor GKT137831 or selective EGFR inhibitor AG1478 reduced protein and mRNA levels of proinflammatory cytokines tumor necrosis factor α (TNF-α), interleukin 6 (IL-6), and interleukin 1β (IL-1β), as well as the activities of Akt and extracellular signal-regulated kinase (ERK) 1/2 in the left ventricle of SHRs. In summary, GKT137831 prevents hypertensive cardiac hypertrophy in SHRs, Nox-deprived ROS regulated EGFR activation through positive feedback in the hypertrophic myocardium, and inhibition of the ROS-EGFR pathway mediates the protective role of GKT137831 in hypertensive cardiac hypertrophy via repressing cardiac inflammation and activation of Akt and ERK1/2. This research will provide additional details for GKT137831 to prevent hypertensive cardiac hypertrophy.
Collapse
|
34
|
Hu D, Cui YX, Wu MY, Li L, Su LN, Lian Z, Chen H. Cytosolic DNA sensor cGAS plays an essential pathogenetic role in pressure overload-induced heart failure. Am J Physiol Heart Circ Physiol 2020; 318:H1525-H1537. [PMID: 32383996 DOI: 10.1152/ajpheart.00097.2020] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Growing evidence shows that activation of inflammation in the heart provokes left ventricular (LV) remodeling and dysfunction in humans and experimental animals with heart failure (HF). Moreover, recent studies found that cyclic GMP-AMP synthase (cGAS), serving as a cytosolic DNA sensor, was essential for activating innate immunity against infection and cellular damage by initiating the STING-IRFs-type I IFN signaling cascade, which played important roles in regulating the inflammatory response. However, the pathophysiological role of cGAS in pressure overload-induced HF is unclear. Wild-type C57BL/6J mice and cGAS inhibition mice were subjected to transverse aortic constriction (TAC) to induce HF or sham operation. Inhibition of cGAS in the murine heart was performed using adeno-associated virus 9 (AAV9). Alterations of the cGAS/STING pathway were examined by qPCR and Western blotting. Cardiac remodeling was assessed by echocardiography as well as histological and molecular phenotyping. Compared with sham-operated mice, the cGAS/STING pathway was activated in LV tissues in TAC mice. Whereas TAC mice exhibited significant pathological cardiac remodeling and LV dysfunction, inhibition of cGAS improved early survival rates after TAC, preserved LV contractile function, and blunted pathological remodeling, including cardiac hypertrophy, fibrosis, and apoptosis. Furthermore, downregulation of cGAS diminished early inflammatory cell infiltration and inflammatory cytokine expression in response to TAC. These results demonstrated that cGAS played an essential pathogenetic role in pressure overload-induced HF to promote pathological cardiac remodeling and dysfunction. Our results suggest that inhibition of cGAS may be a novel therapeutic approach for HF.NEW & NOTEWORTHY In this study, we first revealed a novel role of cGAS in the regulation of pathological cardiac remodeling and dysfunction upon pressure overload. We found that the cGAS/STING pathway was activated during pressure overload. Moreover, we also demonstrated that inhibition of the cGAS/STING pathway alleviated pathological cardiac remodeling and downregulated the early inflammatory response during pressure overload-induced HF. Together, these findings will provide a new therapeutic target for HF.
Collapse
Affiliation(s)
- Dan Hu
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Yu-Xia Cui
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Man-Yan Wu
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Long Li
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Li-Na Su
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Zheng Lian
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Hong Chen
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| |
Collapse
|
35
|
Ovchinnikov AG, Arefieva TI, Potekhina AV, Filatova AY, Ageev FT, Boytsov SA. The Molecular and Cellular Mechanisms Associated with a Microvascular Inflammation in the Pathogenesis of Heart Failure with Preserved Ejection Fraction. Acta Naturae 2020; 12:40-51. [PMID: 32742726 PMCID: PMC7385098 DOI: 10.32607/actanaturae.10990] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a severe disease with an often unfavorable outcome. The prevalence of HFpEF continues to increase, while effective treatment options remain elusive. All the medical strategies used to improve the outcome in a heart failure with reduced ejection fraction proved ineffective in HFpEF, which was probably due to the different mechanisms of development of these two types of heart failure and the diversity of the HFpEF phenotypes. According to the current paradigm of HFpEF development, a chronic mild pro-inflammatory state causes a coronary microvascular endothelial inflammation, with further myocardial fibrosis and diastolic dysfunction progression. This inflammatory paradigm of HFpEF has been confirmed with some evidence, and suppressing the inflammation may become a novel strategy for treating and managing HFpEF. This review summarizes current concepts about a microvascular inflammation in hypertrophied myocardium and provides a translational perspective of the anti-inflammatory and immunomodulatory approaches in HFpEF.
Collapse
Affiliation(s)
- A. G. Ovchinnikov
- National Medical Research Center of Cardiology, Moscow, 121552 Russia
| | - T. I. Arefieva
- National Medical Research Center of Cardiology, Moscow, 121552 Russia
| | - A. V. Potekhina
- National Medical Research Center of Cardiology, Moscow, 121552 Russia
| | - A. Yu. Filatova
- National Medical Research Center of Cardiology, Moscow, 121552 Russia
| | - F. T. Ageev
- National Medical Research Center of Cardiology, Moscow, 121552 Russia
| | - S. A. Boytsov
- National Medical Research Center of Cardiology, Moscow, 121552 Russia
| |
Collapse
|
36
|
Complement component C3 and the TLR co-receptor CD14 are not involved in angiotensin II induced cardiac remodelling. Biochem Biophys Res Commun 2020; 523:867-873. [PMID: 31955888 DOI: 10.1016/j.bbrc.2020.01.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 01/05/2020] [Indexed: 11/23/2022]
Abstract
Inflammation is centrally involved in the development of cardiac hypertrophy and the processes of remodelling. The complement system and Toll-like receptor (TLR) family, two upstream arms of the innate immune system, have previously been reported to be involved in cardiac remodelling. However, the role of complement component 3 (C3), TLR co-receptor CD14 and the synergy between them have not been addressed during pressure overload-induced cardiac remodelling. Here, we examined angiotensin II-induced cardiac hypertrophy and remodelling for 7 days in male C57Bl/6 J mice deficient in C3, CD14, or both (C3CD14), and WT controls. Angiotensin II infusion induced a mild concentric hypertrophic phenotype in WT mice with increased left ventricle weight, wall thicknesses and reduced ventricular internal diameter, associated with increased cardiac fibrosis. However, there were no differences between WT mice and mice deficient for C3, CD14 or C3CD14, as systolic blood pressure, cardiac function and structure and levels of fibrosis were comparable between WT mice and the three other genotypes. C5a did not change in angiotensin II treated mice, whereas Mac2 levels were increased in angiotensin II treated mice, but did not differ between genotypes. The inflammatory IL-6 response was comparable between WT and C3 deficient mice, however, it was decreased in CD14 and C3CD14 deficient mice. We conclude that deficiency in C3, CD14 or C3CD14 had no effect on cardiac remodelling following angiotensin II-induced pressure overload. This suggests that C3 and CD14 are not involved in angiotensin II-induced adverse cardiac remodelling.
Collapse
|
37
|
Rhee AJ, Lavine KJ. New Approaches to Target Inflammation in Heart Failure: Harnessing Insights from Studies of Immune Cell Diversity. Annu Rev Physiol 2019; 82:1-20. [PMID: 31658002 DOI: 10.1146/annurev-physiol-021119-034412] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Despite mounting evidence implicating inflammation in cardiovascular diseases, attempts at clinical translation have shown mixed results. Recent preclinical studies have reenergized this field and provided new insights into how to favorably modulate cardiac macrophage function in the context of acute myocardial injury and chronic disease. In this review, we discuss the origins and roles of cardiac macrophage populations in the steady-state and diseased heart, focusing on the human heart and mouse models of ischemia, hypertensive heart disease, and aortic stenosis. Specific attention is given to delineating the roles of tissue-resident and recruited monocyte-derived macrophage subsets. We also highlight emerging concepts of monocyte plasticity and heterogeneity among monocyte-derived macrophages, describe possible mechanisms by which infiltrating monocytes acquire unique macrophage fates, and discuss the putative impact of these populations on cardiac remodeling. Finally, we discuss strategies to target inflammatory macrophage populations.
Collapse
Affiliation(s)
- Aaron J Rhee
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| | - Kory J Lavine
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA; .,Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA.,Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|
38
|
Zhang L, Yang X, Jiang G, Yu Y, Wu J, Su Y, Sun A, Zou Y, Jiang H, Ge J. HMGB1 enhances mechanical stress-induced cardiomyocyte hypertrophy in vitro via the RAGE/ERK1/2 signaling pathway. Int J Mol Med 2019; 44:885-892. [PMID: 31524228 PMCID: PMC6657962 DOI: 10.3892/ijmm.2019.4276] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 06/26/2019] [Indexed: 01/13/2023] Open
Abstract
Pressure overload-induced cardiac hypertrophy is associated with a complex spectrum of pathophysiological mechanisms, including the inflammation response. High mobility group box-1 (HMGB1), a pro-inflammatory cytokine, is not only increased in myocardium under pressure overload, but also exacerbates pressure overload-induced cardiac hypertrophy and dysfunction; however, the underlying mechanisms have remained elusive. In the present study, cultured cardiomyocytes were stimulated by mechanical stress and/or HMGB1 for various durations to examine the role of HMGB1 in cardiomyocyte hypertrophy, and to detect the expression of receptor for advanced glycation end products (RAGE), toll-like receptor 4 (TLR-4) and the activation status of mitogen-activated protein kinases (MAPKs) and Janus kinase 2 (JAK2)/STAT3. The results indicated that HMGB1 aggravated mechanical stress-induced cardiomyocyte hypertrophy. Furthermore, mechanical stress and HMGB1 stimulation activated extracellular signal-regulated kinase 1/2 (ERK1/2), P38 and JAK2/STAT3 signaling in cardiomyocytes, but an additive effect of the combined stimuli was only observed on the activation of ERK1/2. In addition, mechanical stress caused a prompt upregulation of the expression of RAGE and TLR-4 in cardiomyocytes, while the activation of ERK1/2 by HMGB1 was inhibited by blockage of RAGE, but not by blockage of TLR-4. In summary, the present results indicated that extracellular HMGB1 enhanced mechanical stress-induced cardiomyocyte hypertrophy in vitro, at least partially via the RAGE/ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Shanghai 200032, P.R. China
| | - Xue Yang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Shanghai 200032, P.R. China
| | - Guoliang Jiang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Shanghai 200032, P.R. China
| | - Ying Yu
- Department of General Practice, Zhongshan Hospital, Shanghai Medical College of Fudan University, Shanghai 200032, P.R. China
| | - Jian Wu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Shanghai 200032, P.R. China
| | - Yangang Su
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Shanghai 200032, P.R. China
| | - Aijun Sun
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Shanghai 200032, P.R. China
| | - Yunzeng Zou
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Shanghai 200032, P.R. China
| | - Hong Jiang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Shanghai 200032, P.R. China
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
39
|
Zhao Y, Wang C, Hong X, Miao J, Liao Y, Hou FF, Zhou L, Liu Y. Wnt/β-catenin signaling mediates both heart and kidney injury in type 2 cardiorenal syndrome. Kidney Int 2019; 95:815-829. [PMID: 30770217 PMCID: PMC6431558 DOI: 10.1016/j.kint.2018.11.021] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 10/19/2018] [Accepted: 11/02/2018] [Indexed: 12/13/2022]
Abstract
In type 2 cardiorenal syndrome, chronic heart failure is thought to cause or promote chronic kidney disease; however, the underlying mechanisms remain poorly understood. We investigated the role of Wnt signaling in heart and kidney injury in a mouse model of cardiac hypertrophy and heart failure induced by transverse aortic constriction (TAC). At 8 weeks after TAC, cardiac hypertrophy, inflammation, and fibrosis were prominent, and echocardiography confirmed impaired cardiac function. The cardiac lesions were accompanied by upregulation of multiple Wnt ligands and activation of β-catenin, as well as activation of the renin-angiotensin system (RAS). Wnt3a induced multiple components of the RAS in primary cardiomyocytes and cardiac fibroblasts in vitro. TAC also caused proteinuria and kidney fibrosis, accompanied by klotho depletion and β-catenin activation in the kidney. Pharmacologic blockade of β-catenin with a small molecule inhibitor or the RAS with losartan ameliorated cardiac injury, restored heart function, and mitigated the renal lesions. Serum from TAC mice was sufficient to activate β-catenin and trigger tubular cell injury in vitro, indicating a role for circulating factors. Multiple inflammatory cytokines were upregulated in the circulation of TAC mice, and tumor necrosis factor-α was able to inhibit klotho, induce β-catenin activation, and cause tubular cell injury in vitro. These studies identify Wnt/β-catenin signaling as a common pathogenic mediator of heart and kidney injury in type 2 cardiorenal syndrome after TAC. Targeting this pathway could be a promising therapeutic strategy to protect both organs in cardiorenal syndrome.
Collapse
Affiliation(s)
- Yue Zhao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Cong Wang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xue Hong
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinhua Miao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yulin Liao
- Division of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fan Fan Hou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China; Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
40
|
Wang W, Wu L, Du X, Zhang F, Ullah SH, Lei T, Li D, Yan X. Anti-Toll-like receptor 2 antibody inhibits nuclear factor kappa B activation and attenuates cardiac damage in high-fat-feeding rats. Acta Biochim Biophys Sin (Shanghai) 2019; 51:347-355. [PMID: 30877771 DOI: 10.1093/abbs/gmz009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 01/10/2019] [Accepted: 01/15/2019] [Indexed: 01/07/2023] Open
Abstract
Long-time consumption of high-fat food is a direct cause of cardiovascular diseases, and high-fat-related inflammation plays an important role in it. Toll-like receptors (TLRs), especially TLR2 and TLR4, play important roles in high-fat-related inflammation. However, the impact of TLR2 on high-fat-associated cardiovascular complications is still unknown. In this study, we try to investigate the relationship between TLR2 and high-fat-related cardiac injury. SD rats were allocated to either a control group which were fed with normal diet or a high-fat group which were fed with high-fat diet for 5 months. At the last month, rats fed with high-fat diet were intraperitoneally injected with control normal mouse IgG or anti-TLR2 antibody. Heart tissues were collected for further analysis. RT-qPCR and western blot analysis results revealed that TLR2 expression was increased in the heart tissues from rats fed with high-fat diet and anti-TLR2 antibody had no effect on TLR2 expression. However, anti-TLR2 antibody alleviated masson staining area, levels of TGF-β1 and Collagen I mRNA, and decreased TUNEL-positive myocardial cells and caspase-3 activity, suggesting that anti-TLR2 antibody protected cardiac cells against high-fat-induced cardiac fibrosis and cell apoptosis. By using immunohistochemistry, RT-qPCR and ELISA, we found that anti-TLR2 antibody blocked NF-κB activation, inhibited the expression of inflammatory factors such as TNF-α, IL-1β, IL-6 and IL-18 in the heart tissues from rats fed with high-fat diet. These results hinted that anti-TLR2 antibody might exert its protective effect via inhibition of the TLR2/NF-κB/inflammation pathway. Our findings suggest that anti-TLR2 antibody has a preventive function against high-fat-induced deleterious effects in the heart, and anti-TLR2 antibody may be used as an attractive therapeutic option for high-fat-induced cardiac injury.
Collapse
Affiliation(s)
- Wanzheng Wang
- Department of Pathology, Medical College of Xi’an Jiaotong University, Xi’an, China
- The Hanjiang River Hospital Affiliated of Xi’an Medical University, Hanzhong, China
| | - Litao Wu
- Department of Biochemistry and Molecular Biology, Medical College of Xi’an Jiaotong University, Xi’an, China
| | - Xiaojuan Du
- Department of Biochemistry and Molecular Biology, Medical College of Xi’an Jiaotong University, Xi’an, China
| | - Fujun Zhang
- Department of Biochemistry and Molecular Biology, Medical College of Xi’an Jiaotong University, Xi’an, China
| | - Sayyed Hanif Ullah
- Department of Biochemistry and Molecular Biology, Medical College of Xi’an Jiaotong University, Xi’an, China
| | - Ting Lei
- The Hanjiang River Hospital Affiliated of Xi’an Medical University, Hanzhong, China
| | - Dongming Li
- Department of Biochemistry and Molecular Biology, Medical College of Xi’an Jiaotong University, Xi’an, China
| | - Xiaofei Yan
- Department of Biochemistry and Molecular Biology, Medical College of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
41
|
Recent advances in understanding the roles of T cells in pressure overload-induced cardiac hypertrophy and remodeling. J Mol Cell Cardiol 2019; 129:293-302. [DOI: 10.1016/j.yjmcc.2019.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 01/08/2019] [Accepted: 01/09/2019] [Indexed: 12/15/2022]
|
42
|
Embryonic programming of heart disease in response to obesity during pregnancy. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165402. [PMID: 30759362 DOI: 10.1016/j.bbadis.2019.01.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 01/19/2019] [Accepted: 01/28/2019] [Indexed: 12/20/2022]
Abstract
Obesity during pregnancy programs adult-onset heart disease in the offspring. Clinical studies indicate that exposure to an adverse environment in utero during early, as compared to late, gestation leads to a higher prevalence of adult-onset heart disease. This suggests that the early developing heart is particularly sensitive to an adverse environment. Accordingly, growing evidence from clinical studies and animal models demonstrates that obesity during pregnancy alters the function of the fetal heart, programming a higher risk of cardiovascular disease later in life. Moreover, gene expression patterns and signaling pathways that promote initiation and progression of cardiovascular disease are altered in the hearts in offspring born to obese mothers. However, the mechanisms mediating the long-term effects of an adverse environment in utero on the developing heart leading to adult-onset disease are not clear. Here, we review clinical and experimental evidence documenting the effects of maternal obesity during pregnancy on the fetal and post-natal heart and emphasize on the potential mechanisms of disease programming.
Collapse
|
43
|
Xu M, Liu PP, Li H. Innate Immune Signaling and Its Role in Metabolic and Cardiovascular Diseases. Physiol Rev 2019; 99:893-948. [PMID: 30565509 DOI: 10.1152/physrev.00065.2017] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The innate immune system is an evolutionarily conserved system that senses and defends against infection and irritation. Innate immune signaling is a complex cascade that quickly recognizes infectious threats through multiple germline-encoded cell surface or cytoplasmic receptors and transmits signals for the deployment of proper countermeasures through adaptors, kinases, and transcription factors, resulting in the production of cytokines. As the first response of the innate immune system to pathogenic signals, inflammatory responses must be rapid and specific to establish a physical barrier against the spread of infection and must subsequently be terminated once the pathogens have been cleared. Long-lasting and low-grade chronic inflammation is a distinguishing feature of type 2 diabetes and cardiovascular diseases, which are currently major public health problems. Cardiometabolic stress-induced inflammatory responses activate innate immune signaling, which directly contributes to the development of cardiometabolic diseases. Additionally, although the innate immune elements are highly conserved in higher-order jawed vertebrates, lower-grade jawless vertebrates lack several transcription factors and inflammatory cytokine genes downstream of the Toll-like receptors (TLRs) and retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs) pathways, suggesting that innate immune signaling components may additionally function in an immune-independent way. Notably, recent studies from our group and others have revealed that innate immune signaling can function as a vital regulator of cardiometabolic homeostasis independent of its immune function. Therefore, further investigation of innate immune signaling in cardiometabolic systems may facilitate the discovery of new strategies to manage the initiation and progression of cardiometabolic disorders, leading to better treatments for these diseases. In this review, we summarize the current progress in innate immune signaling studies and the regulatory function of innate immunity in cardiometabolic diseases. Notably, we highlight the immune-independent effects of innate immune signaling components on the development of cardiometabolic disorders.
Collapse
Affiliation(s)
- Meng Xu
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan , China ; Medical Research Center, Zhongnan Hospital of Wuhan University , Wuhan , China ; Animal Experiment Center, Wuhan University , Wuhan , China ; Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario , Canada
| | - Peter P Liu
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan , China ; Medical Research Center, Zhongnan Hospital of Wuhan University , Wuhan , China ; Animal Experiment Center, Wuhan University , Wuhan , China ; Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario , Canada
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan , China ; Medical Research Center, Zhongnan Hospital of Wuhan University , Wuhan , China ; Animal Experiment Center, Wuhan University , Wuhan , China ; Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario , Canada
| |
Collapse
|
44
|
Nox1/4 dual inhibitor GKT137831 attenuates hypertensive cardiac remodelling associating with the inhibition of ADAM17-dependent proinflammatory cytokines-induced signalling pathways in the rats with abdominal artery constriction. Biomed Pharmacother 2019; 109:1907-1914. [DOI: 10.1016/j.biopha.2018.11.077] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/10/2018] [Accepted: 11/19/2018] [Indexed: 11/21/2022] Open
|
45
|
Brenes-Castro D, Castillo EC, Vázquez-Garza E, Torre-Amione G, García-Rivas G. Temporal Frame of Immune Cell Infiltration during Heart Failure Establishment: Lessons from Animal Models. Int J Mol Sci 2018; 19:E3719. [PMID: 30467294 PMCID: PMC6321195 DOI: 10.3390/ijms19123719] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 11/13/2018] [Accepted: 11/14/2018] [Indexed: 12/16/2022] Open
Abstract
Heart failure (HF) is a cardiovascular syndrome characterized by maladaptive changes with an underlying inflammatory mediated pathogenesis. Nevertheless, current therapy is aimed at the heart workload and neurohormonal axis; thus, prognosis remains poor. To continue improving treatment, we rely on murine models for a better understanding of HF pathophysiology. Among them, pressure overload HF (PO-HF) animal models are a common strategy. Development of PO-HF is characterized by monocyte infiltration, which orchestrates a cascade of events leading to sustained inflammation and maladaptive changes. Here, we divide the PO-HF model progression into four phases and describe the inflammatory, structural, and gene expression profiles. This division is relevant due to its similarities with clinical hypertensive heart disease progression to HF. Evidence shows improvement in hemodynamic and other local parameters by altering the inflammatory response in a specific immune response at a specific point of time. Thus, it is relevant to focus on the time-dependent immune response interaction in order to provide more effective therapy. This review summarizes the pathogenesis of PO-HF murine models, highlighting the inflammatory events in a time frame view. By this approach, we expect to provide researchers with a better understanding of the intertwining time-dependent events that occur in PO-HF.
Collapse
Affiliation(s)
- David Brenes-Castro
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Cátedra de Cardiología y Medicina Vascular, Monterrey, Nuevo León 64849, Mexico.
| | - Elena C Castillo
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Cátedra de Cardiología y Medicina Vascular, Monterrey, Nuevo León 64849, Mexico.
| | - Eduardo Vázquez-Garza
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Cátedra de Cardiología y Medicina Vascular, Monterrey, Nuevo León 64849, Mexico.
| | - Guillermo Torre-Amione
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Cátedra de Cardiología y Medicina Vascular, Monterrey, Nuevo León 64849, Mexico.
- Tecnologico de Monterrey, Hospital Zambrano Hellion, TecSalud, Centro de Investigación Biomédica, San Pedro Garza García, Nuevo León 66278, Mexico.
- Methodist DeBakey Heart & Vascular Center, The Methodist Research Institute, Weill Cornell Medical College, Houston, TX 77030, USA.
| | - Gerardo García-Rivas
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Cátedra de Cardiología y Medicina Vascular, Monterrey, Nuevo León 64849, Mexico.
- Tecnologico de Monterrey, Hospital Zambrano Hellion, TecSalud, Centro de Investigación Biomédica, San Pedro Garza García, Nuevo León 66278, Mexico.
| |
Collapse
|
46
|
Błyszczuk P. Toll-like receptor 2: On the crossroad between immune response, cardiac fibrosis and liquorice candy. Int J Cardiol 2018; 267:163-164. [DOI: 10.1016/j.ijcard.2018.05.071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 05/21/2018] [Indexed: 11/27/2022]
|
47
|
NLRP1 promotes TGF-β1-induced myofibroblast differentiation in neonatal rat cardiac fibroblasts. J Mol Histol 2018; 49:509-518. [PMID: 30120609 DOI: 10.1007/s10735-018-9789-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 07/27/2018] [Indexed: 12/15/2022]
Abstract
Nuclear localization leucine-rich-repeat protein 1 (NLRP1) is a member of Nod-like receptors (NLRs) family. Recent studies have reported that NLRP1 is involved in various diseases, especially in cardiovascular diseases. However, the effect of NLRP1 on cardiac fibrosis remains unclear. In this study, NLRP1 overexpression and NLRP1 silencing constructs were transfected into neonatal rat cardiac fibroblasts induced by TGF-β1 for 48 h to investigate the effect of NLRP1 in cardiac fibrosis and its molecular mechanisms. Cardiac fibroblasts were transfected with NLRP1 and then cultured in the presence and absence of TGF-β1and Smad3 inhibitor (SIS3). Our data indicated that NLRP1 not only promoted fibroblast activation and myofibroblast differentiation, but also upregulated the mRNA and protein levels of α-SMA in the TGF-β1-treated neonatal rat cardiac fibroblasts. Overexpressing NLRP1 in TGF-β1-induced cardiac fibroblasts upregulated the mRNA and protein levels of Collagen I, Collagen III, and connective tissue growth factor. Moreover, NLRP1 upregulated the protein levels of Smad2, Smad3, and Smad4 in nuclei of fibroblasts, and attenuated levels of phosphorylated Smad2 and Smad3 in the cytoplasm of fibroblasts induced by TGF-β1. In addition, the increase in fibrotic genes and Smad proteins was significantly reduced in the presence of SIS3. Our findings illustrated that NLRP1 promoted myofibroblast differentiation and excessive ECM production in TGF-β1-induced neonatal cardiac fibroblasts through directly targeting TGF-β1/Smad signaling pathways.
Collapse
|
48
|
Spurthi KM, Sarikhani M, Mishra S, Desingu PA, Yadav S, Rao S, Maity S, Tamta AK, Kumar S, Majumdar S, Jain A, Raghuraman A, Khan D, Singh I, Samuel RJ, Ramachandra SG, Nandi D, Sundaresan NR. Toll-like receptor 2 deficiency hyperactivates the FoxO1 transcription factor and induces aging-associated cardiac dysfunction in mice. J Biol Chem 2018; 293:13073-13089. [PMID: 29929978 DOI: 10.1074/jbc.ra118.001880] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 06/09/2018] [Indexed: 12/17/2022] Open
Abstract
Toll-like receptors (TLRs) are a family of pattern-recognition receptors involved in innate immunity. Previous studies have shown that TLR2 inhibition protects the heart from acute stress, including myocardial infarction and doxorubicin-induced cardiotoxicity in animal models. However, the role of TLR2 in the development of aging-associated heart failure is not known. In this work, we studied aging-associated changes in structure and function of TLR2-deficient mice hearts. Whereas young TLR2-KO mice did not develop marked cardiac dysfunction, 8- and 12-month-old TLR2-KO mice exhibited spontaneous adverse cardiac remodeling and cardiac dysfunction in an age-dependent manner. The hearts of the 8-month-old TLR2-KO mice had increased fibrosis, cell death, and reactivation of fetal genes. Moreover, TLR2-KO hearts displayed reduced infiltration by macrophages, increased numbers of myofibroblasts and atrophic cardiomyocytes, and higher levels of the atrophy-related ubiquitin ligases MuRF-1 and atrogin-1. Mechanistically, TLR2 deficiency impaired the PI3K/Akt signaling pathway, leading to hyperactivation of the transcription factor Forkhead box protein O1 (FoxO1) and, in turn, to elevated expression of FoxO target genes involved in the regulation of muscle wasting and cell death. AS1842856-mediated chemical inhibition of FoxO1 reduced the expression of the atrophy-related ubiquitin ligases and significantly reversed the adverse cardiac remodeling while improving the contractile functions in the TLR2-KO mice. Interestingly, TLR2 levels decreased in hearts of older mice, and the activation of TLR1/2 signaling improved cardiac functions in these mice. These findings suggest that TLR2 signaling is essential for protecting the heart against aging-associated adverse remodeling and contractile dysfunction in mice.
Collapse
Affiliation(s)
- Kondapalli Mrudula Spurthi
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Mohsen Sarikhani
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Sneha Mishra
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Perumal Arumugam Desingu
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Shikha Yadav
- the Department of Biochemistry, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Swathi Rao
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Sangeeta Maity
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Ankit Kumar Tamta
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Shweta Kumar
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Shamik Majumdar
- the Department of Biochemistry, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Aditi Jain
- the Centre for Biosystems Science and Engineering, Indian Institute of Science, Bengaluru, Karnataka 560012, India, and
| | - Aishwarya Raghuraman
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Danish Khan
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Ishwar Singh
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Rosa J Samuel
- the Central Animal Facility, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Subbaraya G Ramachandra
- the Central Animal Facility, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Dipankar Nandi
- the Department of Biochemistry, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Nagalingam R Sundaresan
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India,
| |
Collapse
|
49
|
Abdullah M, Berthiaume JM, Willis MS. Tumor necrosis factor receptor-associated factor 6 as a nuclear factor kappa B-modulating therapeutic target in cardiovascular diseases: at the heart of it all. Transl Res 2018; 195:48-61. [PMID: 29175266 PMCID: PMC5898986 DOI: 10.1016/j.trsl.2017.10.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/17/2017] [Accepted: 10/30/2017] [Indexed: 02/06/2023]
Abstract
Inflammatory and immune signaling has been documented as a root cause of many cardiovascular pathologies. In this review, we explore the emerging role of tumor necrosis factor receptor-associated factor 6 (TRAF6)-nuclear factor kappa B (NF-κB) signaling axis in atherosclerosis, ischemic heart disease, pathologic cardiac hypertrophy or heart failure, myocarditis, and sepsis-induced cardiomyopathy. We discuss the current understanding of cardiac inflammation in heart disease, present the TRAF6 signaling axis in the heart, then summarize what is known about TRAF6 in pathophysiology of heart disease including proof-of-concept studies that identify the utility of blocking TRAF6 to attenuate cardiac dysfunction, which suggests that TRAF6 is a novel, druggable target in treating cardiovascular disease incurred by inflammatory processes.
Collapse
Affiliation(s)
- Muhammad Abdullah
- Department of Biochemistry, QuaidiAzam University, Islamabad, Pakistan; Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC
| | - Jessica M Berthiaume
- Department of Physiology & Biophysics, Case Western Reserve University, Cleveland, Ohio
| | - Monte S Willis
- Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC; Department of Pharmacology, University of North Carolina, Chapel Hill, NC.
| |
Collapse
|
50
|
Acute Hyperthermia Inhibits TGF-β1-induced Cardiac Fibroblast Activation via Suppression of Akt Signaling. Sci Rep 2018; 8:6277. [PMID: 29674727 PMCID: PMC5908876 DOI: 10.1038/s41598-018-24749-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 03/06/2018] [Indexed: 12/19/2022] Open
Abstract
Transforming growth factor-β1 (TGF-β1) induces phenotypic changes in fibroblasts to become myofibroblasts with increased production of extracellular matrix (ECM) components and cytokines. It is also known that excessive activation of myofibroblasts accelerates cardiac fibrosis, remodeling, and thus cardiac dysfunction. However, no effective therapy has been established to prevent this process although recent clinical studies have demonstrated the effectiveness of hyperthermia in cardiac dysfunction. The aim of this study was to examine the molecular mechanism of hyperthermia on TGF-β1-mediated phenotypic changes in cardiac fibroblasts. TGF-β1 increased the expression of IL-6, α-smooth muscle actin (α-SMA), and collagen in human cardiac fibroblasts (HCFs). Hyperthermia (42 °C) significantly prevented these changes, i.e., increases in IL-6, α-SMA, and collagen, as induced by TGF-β1 in a time-dependent manner. Immunoblotting showed that hyperthermia decreased Akt/S6K signaling, but did not affect Smad2 and Smad3 signaling. Pharmacological inhibition of Akt signaling mimicked these effects of hyperthermia. Furthermore, hyperthermia treatment prevented cardiac fibrosis in Ang II infusion mice model. Putting together, our findings suggest that hyperthermia directly inhibits TGF-β-mediated activation of HCFs via suppressing Akt/S6K signaling.
Collapse
|