1
|
Fahmy MI, Sadek MA, Abdou K, El-Dessouki AM, El-Shiekh RA, Khalaf SS. Orientin: a comprehensive review of a promising bioactive flavonoid. Inflammopharmacology 2025; 33:1713-1728. [PMID: 40056319 PMCID: PMC11991976 DOI: 10.1007/s10787-025-01690-5] [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: 02/06/2025] [Accepted: 02/10/2025] [Indexed: 03/10/2025]
Abstract
Medicinal herbs continue to play an important part in modern drugs and healthcare because customers think that most of them have fewer or milder side effects than traditional modern medicines. Bioactive compounds are typically isolated from plants before being used as a source of therapeutic medicines. As a result, extracting bioactive compounds from medicinal plants is an important step in developing plant-based medications. Orientin is a flavonoid C-glycoside found in many plants, is frequently used in bioactivity studies due to its numerous beneficial properties, which include antioxidants, antiaging, anti-inflammation, vasodilation and cardioprotective, neuroprotective, antidiabetic, hepatoprotective, and adaptogenic effects. In this review, the comprehensive search for the health benefits of orientin was traced. The findings reflected that orientin could be considered one of the important natural candidates as a potential nutraceutical. This underscores its promising attributes and potential applications in health and wellness. Further research may be guaranteed to fully elucidate its benefits and mechanisms of action.
Collapse
Affiliation(s)
- Mohamed I Fahmy
- Department of Pharmacology and Toxicology, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), Giza, Egypt
| | - Mohamed A Sadek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Kareem Abdou
- College of Pharmacy, Al-Ain University, Abu Dhabi, United Arab Emirates
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ahmed M El-Dessouki
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza, 12566, Egypt
| | - Riham A El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, 11562, Egypt.
| | - Samar S Khalaf
- Biochemistry Department Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| |
Collapse
|
2
|
De Masi De Luca G, Palama Z, Longo S, Barba F, Robles AG, Nesti M, Scara A, Coluccia G, Colopi M, De Masi De Luca G, Minardi S, Fusco L, Palmisano P, Accogli M, Sciarra L, Romano S. Effect of Dapagliflozin on Ventricular Arrhythmic Events in Heart Failure Patients With an Implantable Cardioverter Defibrillator. Cardiol Res 2025; 16:140-152. [PMID: 40051671 PMCID: PMC11882230 DOI: 10.14740/cr2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 01/21/2025] [Indexed: 03/09/2025] Open
Abstract
Background The aim of our study was to evaluate the effects of dapagliflozin on the ventricular arrhythmia burden (VAb) in patients with heart failure with reduced ejection fraction (HFrEF) and an implantable cardioverter defibrillator (ICD), correlating the possible reduction in arrhythmic events and ICD therapies with the basal functional capacity, as well as the remodeling parameters induced by treatment. Methods A total of 117 outpatient ICD patients with a known diagnosis of HFrEF who underwent treatment with dapagliflozin were evaluated according to a prospective observational protocol. VAb (including sustained ventricular tachycardia, non-sustained ventricular tachycardia, ventricular fibrillation, and total ventricular events) and specific ICD therapies (anti-tachycardia pacing (ATP) and ICD shocks) were extrapolated from the devices' memory (events per patient per month) by comparing events in the observation period before and after the introduction of dapagliflozin. Results The VAb was significantly reduced after dapagliflozin introduction (2.9 ± 1.8 vs. 4.5 ± 2.0, P = 0.01). The burden of appropriate ATPs was significantly reduced (0.57 ± 0.80 vs. 0.65 ± 0.91, P = 0.03), but not for ICD shocks. In patients with a more advanced functional class, a greater reduction in VAb was observed than in patients with a better initial functional capacity (2.2 ± 0.8 vs. 5.5 ± 1.8, P = 0.001 in the New York Heart Association (NYHA) III/IV group; 3.5 ± 2.1 vs. 4.5 ± 2.2, P = 0.02 in the NYHA I/II group). Considering two independent groups according to reverse remodeling (Δleft ventricular ejection fraction (LVEF) > 15%), a significant reduction in VAb was observed only in those patients who presented significant reverse remodeling (2.5 ± 1.1 vs. 5.1 ± 1.6, P = 0.01). A statistically significant interaction between the variation of total ventricular arrhythmias (VTA) and the basal NYHA class (F(1,115) = 142.25, P < 0.0001, partial η2 = 0.553), as well as between the variation of VTA and the ΔLVEF (F(1,115) = 107.678, P < 0.0001, partial η2 = 0.484) has been demonstrated using a two-way analysis of variance (ANOVA) test. Conclusions In ICD outpatients with HFrEF, dapagliflozin treatment produces a reduction in arrhythmic ventricular events. This improvement is more evident in patients who have a worse functional class and thus a more precarious hemodynamic state, and in patients who present with significant ventricular reverse remodeling. Therefore, we can hypothesize that the hemodynamic and structural improvements induced by treatment represent, at least in the short-medium term, some of the principal elements justifying the significant reduction in VAb.
Collapse
Affiliation(s)
- Gabriele De Masi De Luca
- Department of Life, Health and Environmental Science, University of L’Aquila, L’Aquila, Italy
- Cardiology Unit, Card. “G. Panico” Hospital, Tricase, Italy
- Cardiomed Medical Center, Maglie, Italy
| | - Zefferino Palama
- Department of Life, Health and Environmental Science, University of L’Aquila, L’Aquila, Italy
- Cardiology Unit, “Villa Verde” Hospital, Taranto, Italy
| | | | | | - Antonio Gianluca Robles
- Department of Life, Health and Environmental Science, University of L’Aquila, L’Aquila, Italy
- Cardiology Department, Ospedale “L. Bonomo”, Andria, Italy
| | - Martina Nesti
- Cardiology Unit, CNR Fondazione Toscana “Gabriele Monasterio”, Pisa, Italy
| | - Antonio Scara
- GVM Care and Research, “San Carlo di Nancy” Hospital, Rome, Italy
| | | | - Marzia Colopi
- Cardiology Unit, Card. “G. Panico” Hospital, Tricase, Italy
| | | | - Simona Minardi
- Department of Life, Health and Environmental Science, University of L’Aquila, L’Aquila, Italy
| | - Liuba Fusco
- Department of Life, Health and Environmental Science, University of L’Aquila, L’Aquila, Italy
| | | | | | - Luigi Sciarra
- Department of Life, Health and Environmental Science, University of L’Aquila, L’Aquila, Italy
| | - Silvio Romano
- Department of Life, Health and Environmental Science, University of L’Aquila, L’Aquila, Italy
| |
Collapse
|
3
|
Chen Z, Zhong M, Lin Y, Zhang W, Zhu Y, Chen L, Huang Z, Luo K, Lu Z, Huang Z, Yan Y. METTL7B-induced histone lactylation prevents heart failure by ameliorating cardiac remodelling. J Mol Cell Cardiol 2025; 202:64-80. [PMID: 40068772 DOI: 10.1016/j.yjmcc.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 02/13/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025]
Abstract
INTRODUCTION Lactylation is important for a variety of biological activities. It is reported that Class I histone deacetylases (HDAC1-3) are histone lysine delactylases. However, the role of lactylation in cardiac remodelling remains uncertain. OBJECTIVES To explore a novel regulator of lactylation and elucidate their functional mechanisms in cardiac remodelling and heart failure. METHODS GSE36961, GSE141910 and GSE174691 related to HCM (hypertrophic cardiomyopathy) were separately acquired from Gene expression Omnibus. Candidate genes related to both HCM and histone lactylation were determined by the intersection of DEGs (differentially expressed genes) and module genes sifted by WGCNA (Weighted Gene Co-Expression Network Analysis). METTL7B was screened out and its expression in hypertrophic myocardium was measured by qRT-PCR and western blotting. Furthermore, immunofluorescence, immunoprecipitation, and RNA pull-down assays were utilized to identify the biological functions of METTL7B. The myocardial biopsy of HCM and transverse aortic constriction (TAC) mouse model were performed to analyze the effects of METTL7B on cardiac remodelling in vivo. RESULTS We observed that the expression of METTL7B was down-regulated in hypertrophic myocardium, and the lactylation level was increased during the early stage and falling rapidly in the process of cardiac remodelling. Furthermore, we demonstrated that sodium lactate (NALA) administration fulfil a protective role on cardiac remodelling, and METTL7B alleviates cardiac remodelling and improves heart function by maintaining the activation of histone lactylation possibly at the later stage. Impressively, METTL7B suppressed the expression of USP38 via m6A dependent mRNA degradation, resulting in increasing ubiquitylation of HDAC3, which is a proven histone lysine delactylases. CONCLUSION We identifed METTL7B as a potential therapeutic target for myocardial remodelling and showed that it played a critical role in the promotion of myocardial lactylation, which is beneficial for improvement of cardiac function and attenuation of cardiac remodelling.
Collapse
Affiliation(s)
- Ziqi Chen
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Meijun Zhong
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Yuhui Lin
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Wei Zhang
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Yinghong Zhu
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Lin Chen
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Ziyao Huang
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Kaiyuan Luo
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Zhifeng Lu
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China.
| | - Zhaoqi Huang
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China.
| | - Yi Yan
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China.
| |
Collapse
|
4
|
Huang H, Huang G, Li R, Wei L, Yuan Z, Huang W. Exercise Training After Myocardial Infarction Enhances Endothelial Progenitor Cells Function via NRG-1 Signaling. Cardiovasc Toxicol 2025; 25:411-426. [PMID: 39893285 DOI: 10.1007/s12012-025-09967-5] [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: 12/10/2024] [Accepted: 01/24/2025] [Indexed: 02/04/2025]
Abstract
Vascular regeneration after myocardial infarction (MI) is essential to improve myocardial ischemia, delay post-infarction ventricular remodeling, and improve the long-term prognosis of MI. Endothelial progenitor cells (EPCs) play important roles in the functional repair and homeostatic maintenance of the vascular endothelium. Exercise training stimulates EPC mobilization and increases the number of circulating EPCs, which has beneficial effects on the restoration of vascular integrity and hemodynamic reconstitution. After post-MI exercise training, cardiac function, the myocardial infarct area, and capillary density in the peri-infarct zone were measured. Bone marrow-derived EPCs were isolated from mice to measure the proliferation, migration, and in vitro angiogenesis of EPCs after myocardial infarction exercise. The expression of NRG-1/ErbB4 signaling factor and related proteins in downstream PI3K/AKT signaling pathway were detected, and the level of autocrine NRG-1 in EPCs was detected. Post-MI resistance training, aerobic exercise training, and combined exercise training increased EPC mobilization and proliferation, migration, and tube-forming capacity, promoted myocardial vascular regeneration, improved cardiac function, and reduced infarct size. Exercise training upregulated NRG-1 expression in EPCs, and NRG-1/ErbB4 signaling activated the downstream PI3K/Akt signaling pathway. Moreover, EPCs may have a positive feedback autocrine loop with NRG-1 to improve the function of EPCs and promote vascular repair and regeneration in mice with MI. Exercise training after MI promotes the function of bone marrow-derived EPCs through NRG-1/ErbB4/PI3K/AKT signaling, thus exerting a role in angiogenesis.
Collapse
Affiliation(s)
- Huai Huang
- Department of Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Guoqiang Huang
- Department of Cardiology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, 528400, China
| | - Ruojun Li
- Department of Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Liqin Wei
- Department of Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Zhu Yuan
- Department of Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Weiqiang Huang
- Department of Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
5
|
Wei H, Guo X, Yan J, Tian X, Yang W, Cui K, Wang L, Guo B. Neuregulin-4 alleviates isoproterenol (ISO)-induced cardial remodeling by inhibiting inflammation and apoptosis via AMPK/NF-κB pathway. Int Immunopharmacol 2024; 143:113301. [PMID: 39418729 DOI: 10.1016/j.intimp.2024.113301] [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: 08/20/2024] [Revised: 09/19/2024] [Accepted: 09/29/2024] [Indexed: 10/19/2024]
Abstract
Cardiac remodeling refers to the abnormal changes in cardiac structure and function caused by various pathological conditions. It is an inevitable pathological process in the occurrence and development of heart failure and is related to a variety of cardiovascular diseases. Inflammation and apoptosis are critical pathological processes involved in cardiac remodeling. Neuregulin 4 (Nrg 4) is an adipokine produced primarily by brown adipose tissue that may play a protective role in a variety of inflammatory diseases. The aim of this study was to investigate whether Nrg4 can delay the progression of cardiac remodeling by regulating AMPK/NF-κB pathway, inhibiting inflammation and apoptosis. In our study, we established a model of cardiac remodeling in mice after 14 days of isoproterenol (ISO) intervention, and then gave Nrg4 treatment for another 4 weeks. The cardiac function, the degree of myocardial hypertrophy and myocardial fibrosis of the mice were observed. At the same time, the levels of apoptosis-related proteins (Bax,Bcl-2,Caspase-3), IL-6,IL-Iβ and TNF-α, as well as the activation level of AMPK/NF-κB signaling pathway were evaluated.Nrg4 alleviated ISO-induced cardiac dysfunction, cardiac hypertrophy and fibrosis in mice. Nrg4 also attenuated ISO-induced apoptosis and reduces levels of inflammatory factors to protect ISO-induced myocardial damage. At the same time, the effect of Nrg4 on AMPK/NF-κB pathway was measured in vivo and in vitro. The administration of an AMPK inhibitor was found to reverse the anti-hypertrophy, anti-inflammatory, and anti-apoptotic effects of Nrg4. Our findings suggest that Nrg4 may play a protective role in cardiac remodeling by inhibiting inflammation and apoptosis via AMPK/NF-κB pathway.
Collapse
Affiliation(s)
- Huiqing Wei
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Xiaohua Guo
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Jie Yan
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Xiaochao Tian
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Wenhui Yang
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Kun Cui
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Lijie Wang
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Bingyan Guo
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China; Hebei Key Laboratory of Laboratory Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China.
| |
Collapse
|
6
|
Ageev AA, Kozhevnikova MV, Tyurina DA, Korobkova EO, Kondratieva TO, Shestakova KM, Moskaleva NE, Markin PA, Khabarova NV, Appolonova SA, Belenkov YN. Left Ventricular Remodeling Predictors in Chronic Heart Failure of Ischemic Etiology. KARDIOLOGIIA 2024; 64:106-116. [PMID: 39637396 DOI: 10.18087/cardio.2024.11.n2794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
Aim To identify metabolomic and structure and function markers of remote left ventricular (LV) remodeling in patients with chronic heart failure (CHF) of ischemic etiology and LV ejection fraction (EF) <50%.Material and methods This prospective study included 56 patients with 3-4 NYHA functional class CHF of ischemic etiology (mean age, 66±7 years) and 50 patients with ischemic heart disease (IHD) without signs of CHF (69 [64; 73.7] years). Concentration of 19 amino acids, 11 products of kynurenine catabolism of tryptophan, 30 acylcarnitines with different chain lengths were measured in all participants. The metabolites that showed statistical differences between the comparison groups were then used for the analysis. Echocardiography was used to assess LV cavity remodeling at the time of the CHF patient inclusion in the study and after 6 months of follow-up. Predictors of long-term LV cavity remodeling were assessed for this cohort taking into account statistically significant echocardiographic parameters and metabolites.Results Patients with CHF of ischemic etiology, predominantly (81%) had pathological calculated types of LV remodeling (concentric and eccentric hypertrophy, 46 and 35%, respectively). However, this classification had limitations in describing this cohort. In addition, in this group, the concentrations of alanine, proline, asparagine, glycine, arginine, histidine, lysine, valine, indolyl-3-acetic acid, indolyl-3-propionic acid, C16-1-OH, and C16-OH were significantly (p<0.05) lower, and the concentrations of most medium- and long-chain acylcarnitines were higher than in patients with IHD without signs of CHF. The long-term (6 months) reverse remodeling of the LV cavity in CHF of ischemic etiology was influenced by changes in the interventricular septum thickness (hazard ratio, HR, 19.07; 95% confidence interval, CI, 1.76-206.8; p=0.006) and concentrations of anthranilic acid (HR 19.8; 95% CI 1.01-387.8; p=0.019) and asparagine (HR 8.76; 95% CI 1.07-71.4; p=0.031).Conclusion The presence of an interventricular septum thickness of more than 13.5 mm, anthranilic acid concentrations of higher than 0.235 μM/l, or an asparagine concentration of less than 135.2 μM/l in patients with CHF of ischemic etiology after 6 months of follow-up affects their achievement of LV cavity reverse remodeling.
Collapse
Affiliation(s)
- A A Ageev
- Sechenov First Moscow State Medical University, Moscow
| | | | - D A Tyurina
- Sechenov First Moscow State Medical University, Moscow
| | - E O Korobkova
- Sechenov First Moscow State Medical University, Moscow
| | | | | | - N E Moskaleva
- Sechenov First Moscow State Medical University, Moscow
| | - P A Markin
- Sechenov First Moscow State Medical University, Moscow
| | - N V Khabarova
- Sechenov First Moscow State Medical University, Moscow
| | | | - Yu N Belenkov
- Sechenov First Moscow State Medical University, Moscow
| |
Collapse
|
7
|
Kan H, Wang P, Yang Y, Jia H, Liu A, Wang M, Ouyang C, Yang X. Apigenin inhibits proliferation and differentiation of cardiac fibroblasts through AKT/GSK3β signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118518. [PMID: 38964628 DOI: 10.1016/j.jep.2024.118518] [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: 04/04/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Salvia miltiorrhiza Bunge (S. miltiorrhiza) is an important Traditional Chinese herbal Medicine (TCM) used to treat cardio-cerebrovascular diseases. Based on the pharmacodynamic substance of S. miltiorrhiza, the aim of present study was to investigate the underlying mechanism of S. miltiorrhiza against cardiac fibrosis (CF) through a systematic network pharmacology approach, molecular docking and dynamics simulation as well as experimental investigation in vitro. MATERIALS AND METHODS A systematic pharmacological analysis was conducted using the Traditional Chinese Medicine Pharmacology (TCMSP) database to screen the effective chemical components of S. miltiorrhiza, then the corresponding potential target genes of the compounds were obtained by the Swiss Target Prediction and TCMSP databases. Meanwhile, GeneCards, DisGeNET, OMIM, and TTD disease databases were used to screen CF targets, and a protein-protein interaction (PPI) network of drug-disease targets was constructed on S. miltiorrhiza/CF targets by Search Tool for the Retrieval of Interacting Genes/Proteins (STING) database. After that, the component-disease-target network was constructed by software Cytoscape 3.7. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were performed for the intersection targets between drug and disease. The relationship between active ingredient of S. miltiorrhiza and disease targets of CF was assessed via molecular docking and molecular dynamics simulation. Subsequently, the underlying mechanism of the hub compound on CF was experimentally investigated in vitro. RESULTS 206 corresponding targets to effective chemical components from S. miltiorrhiza were determined, and among them, there were 82 targets that overlapped with targets of CF. Further, through PPI analysis, AKT1 and GSK3β were the hub targets, and which were both enriched in the PI3K/AKT signaling pathway, it was the sub-pathways of the lipid and atherosclerosis pathway. Subsequently, compound-disease-genes-pathways diagram is constructed, apigenin (APi) was a top ingredients and AKT1 (51) and GSK3β (22) were the hub genes according to the degree value. The results of molecular docking and dynamics simulation showed that APi has strong affinities with AKT and GSK3β. The results of cell experiments showed that APi inhibited cells viability, proliferation, proteins expression of α-SMA and collagen I/III, phosphorylation of AKT1 and GSK3β in MCFs induced by TGFβ1. CONCLUSION Through a systematic network pharmacology approach, molecular docking and dynamics simulation, and confirmed by in vitro cell experiments, these results indicated that APi interacts with AKT and GSK3β to disrupt the phosphorylation of AKT and GSK3β, thereby inhibiting the proliferation and differentiation of MCFs induced by TGFβ1, which providing new insights into the pharmacological mechanism of S. miltiorrhiza in the treatment of CF.
Collapse
Affiliation(s)
- Hongshuang Kan
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, 437100, China.
| | - Pengyu Wang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, 437100, China.
| | - Yayuan Yang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, 437100, China.
| | - Hongyu Jia
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, 437100, China.
| | - Aimei Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437100, China.
| | - Miao Wang
- Department of Cardiovascular Medicine, Xian Ning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xian'an District, Xian Ning City, Hubei Province, China.
| | - Changhan Ouyang
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437100, China.
| | - Xiaosong Yang
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437100, China; School of Stomatology and Ophthalmology, Xianning Medical College, Hubei University of Science and Technology, 437100, China.
| |
Collapse
|
8
|
Shi M, Yuan H, Li Y, Guo Z, Wei J. Targeting Macrophage Phenotype for Treating Heart Failure: A New Approach. Drug Des Devel Ther 2024; 18:4927-4942. [PMID: 39525046 PMCID: PMC11549885 DOI: 10.2147/dddt.s486816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Heart failure (HF) is a disease with high morbidity and mortality rates worldwide and significantly affects human health. Currently, the treatment options for HF are limited, and there is an urgent need to discover new therapeutic targets and strategies. Macrophages are innate immune cells involved in the development of HF. They play a crucial role in maintaining cardiac homeostasis and regulating cardiac stress. Recently, macrophages have received increasing attention as potential targets for treating HF. With the improvement of technological means, the study of macrophages in HF has made great progress. This article discusses the biological functions of macrophage phagocytosis, immune response, and tissue repair. The polarization, pyroptosis, autophagy, and apoptosis are of macrophages, deeply involved in the pathogenesis of HF. Modulation of the phenotypic changes of macrophages can improve immune-inflammation, myocardial fibrosis, energy metabolism, apoptosis, and angiogenesis in HF.
Collapse
Affiliation(s)
- Min Shi
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- Hunan Key Laboratory of Colleges and Universities of Intelligent TCM Diagnosis and Preventive Treatment of Chronic Diseases, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Hui Yuan
- Hunan Key Laboratory of Colleges and Universities of Intelligent TCM Diagnosis and Preventive Treatment of Chronic Diseases, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- First Clinical College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 41020, People’s Republic of China
| | - Ya Li
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Zhihua Guo
- Hunan Key Laboratory of Colleges and Universities of Intelligent TCM Diagnosis and Preventive Treatment of Chronic Diseases, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- First Clinical College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 41020, People’s Republic of China
| | - Jiaming Wei
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- Hunan Key Laboratory of Colleges and Universities of Intelligent TCM Diagnosis and Preventive Treatment of Chronic Diseases, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
| |
Collapse
|
9
|
Yang Z, Li J, Song H, Wu H, Zhang S, Mei Z, Xue Y, Zhang X, Yan C, Han Y. Integrating network pharmacology and experimental verification to explore the pharmacological mechanisms of Guanxin Shutong capsule in treating heart failure. Medicine (Baltimore) 2024; 103:e40118. [PMID: 39432648 PMCID: PMC11495747 DOI: 10.1097/md.0000000000040118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 09/27/2024] [Indexed: 10/23/2024] Open
Abstract
The Guanxin Shutong capsule (GXST), a traditional Chinese medicine, is commonly used for treating cardiovascular disease, it has shown efficacy in improving symptoms and enhancing the quality of life for patients with heart failure (HF). However, the specific mechanism of action of GXST in HF remains unclear. In this study, we employed a comprehensive approach combining network pharmacology, molecular dynamics (MD) simulations, and in vitro validations to investigate the potential targets and molecular mechanisms of GXST against HF. We collected active ingredients and target genes of GXST, as well as related genes of HF, from multiple public databases. Using bioinformatics analysis, we constructed networks of ingredients-disease-targets and performed functional annotations of the core targets. MD simulations were conducted to verify the binding between the core protein-ligand complexes. In vitro evaluations, including cell proliferation, apoptosis, and protein expression in human umbilical vein endothelial cells (HUVECs) and H9C2 cells were treated with GXST, were performed for pharmacodynamics evaluation. Network analysis revealed 320 intersection genes and 74 active ingredients in the Herbs-ingredients-target genes-disease network. We identified key active ingredients and target genes that overlapped. The KEGG pathways of the intersection genes were primarily enriched in the PI3K-Akt signaling pathway and apoptosis. The protein-protein interaction network highlighted proteins such as AKT1, VEGFR2, and eNOS. MD simulations confirmed stable docking and lower binding energy between 4 identified ingredients (kaempferol, quercetin, (2R)-5,7-dihydroxy-2-(4-hydroxyphenyl) chroman-4-one, and ellagic acid) and their respective core proteins (VEGFR2, eNOS, and AKT). In vitro experiments demonstrated the protective effects of GXST against H2O2-induced apoptosis in both HUVECs and H9C2 cells. Notably, consistent with the in silico predictions, GXST effectively activates the VEGFR2/AKT/eNOS signaling pathways in HUVECs. This study provides insights into the underlying mechanism of GXST's therapeutic effects in heart failure. The involvement of the VEGFR2/AKT/eNOS signaling pathways suggests their importance in further elucidating and applying GXST in the clinical treatment of heart failure.
Collapse
Affiliation(s)
- Zheming Yang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning, China
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China
| | - Jiayin Li
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning, China
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China
| | - Haixu Song
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China
| | - Hanlin Wu
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China
| | - Shuli Zhang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning, China
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China
| | - Zhu Mei
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning, China
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China
| | - Yu Xue
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China
| | - Xiaolin Zhang
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China
| | - Chenghui Yan
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China
| | - Yaling Han
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning, China
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
10
|
Fu K, Dong Y, Wang Z, Teng J, Cheng C, Su C, Ji X, Lu H. The role of body composition in left ventricular remodeling, reverse remodeling, and clinical outcomes for heart failure with mildly reduced ejection fraction: more knowledge to the "obesity paradox". Cardiovasc Diabetol 2024; 23:334. [PMID: 39261931 PMCID: PMC11391770 DOI: 10.1186/s12933-024-02430-9] [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: 08/16/2024] [Accepted: 09/03/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND Although the "obesity paradox" is comprehensively elucidated in heart failure (HF) with reduced ejection fraction (HFrEF) and HF with preserved ejection fraction (HFpEF), the role of body composition in left ventricular (LV) remodeling, LV reverse remodeling (LVRR), and clinical outcomes is still unclear for HF with mildly reduced ejection fraction (HFmrEF). METHODS Our study is a single-centre, prospective, and echocardiography-based study. Consecutive HFmrEF patients, defined as HF patients with a left ventricular ejection fraction (LVEF) between 40 and 49%, between January 2016 to December 2021 were included. Echocardiography was re-examined at 3-, 6-, and 12-month follow-up to assess the LVRR dynamically. Body mass index (BMI), fat mass, fat-free mass, percent body fat (PBF), CUN-BAE index, and lean mass index (LMI) were adopted as anthropometric parameters in our study to assess body composition. The primary outcome was LVRR, defined as: (1) a reduction higher than 10% in LV end-diastolic diameter index (LVEDDI), or a LVEDDI < 33 mm/m2, (2) an absolute increase of LVEF higher than 10 points compared with baseline echocardiogram, or a follow-up LVEF ≥50%. The secondary outcome was a composite of re-hospitalization for HF or cardiovascular death. RESULTS A total of 240 HFmrEF patients were enrolled in our formal analysis. After 1-year follow-up based on echocardiography, 113 (47.1%) patients developed LVRR. Patients with LVRR had higher fat mass (21.7 kg vs. 19.3 kg, P = 0.034) and PBF (28.7% vs. 26.6%, P = 0.047) compared with those without. The negative correlation between anthropometric parameters and baseline LVEDDI was significant (all P < 0.05). HFmrEF patients with higher BMI, fat mass, PBF, CUN-BAE index, and LMI had more pronounced and persistent increase of LVEF and decline in LV mass index (LVMI). Univariable Cox regression analysis revealed that higher BMI (HR 1.042, 95% CI 1.002-1.083, P = 0.037) and fat mass (HR 1.019, 95% CI 1.002-1.036, P = 0.026) were each significantly associated with higher cumulative incidence of LVRR for HFmrEF patients, while this relationship vanished in the adjusted model. Mediation analysis indicated that the association between BMI and fat mass with LVRR was fully mediated by baseline LV dilation. Furthermore, higher fat mass (aHR 0.957, 95% CI 0.917-0.999, P = 0.049) and PBF (aHR 0.963, 95% CI 0.924-0.976, P = 0.043) was independently associated with lower risk of adverse clinical events. CONCLUSIONS Body composition played an important role in the LVRR and clinical outcomes for HFmrEF. For HFmrEF patients, BMI and fat mass was positively associated with the cumulative incidence of LVRR, while higher fat mass and PBF predicted lower risk of adverse clinical events but not LMI.
Collapse
Affiliation(s)
- Kang Fu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Jinan, China
- Department of Cardiology, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Shandong University, Jinan, China
| | - Youran Dong
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Jinan, China
- Department of Cardiology, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Shandong University, Jinan, China
| | - Zhiyuan Wang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Jinan, China
- Department of Cardiology, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Shandong University, Jinan, China
| | - Junlin Teng
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Jinan, China
- Department of Cardiology, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Shandong University, Jinan, China
| | - Congyi Cheng
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Jinan, China
- Department of Cardiology, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Shandong University, Jinan, China
| | - Cong Su
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Jinan, China
- Department of Cardiology, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Shandong University, Jinan, China
| | - Xiaoping Ji
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Jinan, China.
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Jinan, China.
- Department of Cardiology, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Shandong University, Jinan, China.
| | - Huixia Lu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Jinan, China.
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Jinan, China.
- Department of Cardiology, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Shandong University, Jinan, China.
| |
Collapse
|
11
|
Castiglioni L, Gelosa P, Muluhie M, Mercuriali B, Rzemieniec J, Gotti M, Fiordaliso F, Busca G, Sironi L. Fenofibrate reduces cardiac remodeling by mitochondrial dynamics preservation in a renovascular model of cardiac hypertrophy. Eur J Pharmacol 2024; 978:176767. [PMID: 38909934 DOI: 10.1016/j.ejphar.2024.176767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/20/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024]
Abstract
Fenofibrate, a PPAR-α agonist clinically used to lower serum lipid levels, reduces cardiac remodeling and improves cardiac function. However, its mechanism of action is not completely elucidated. In this study we examined the effect of fenofibrate on mitochondria in a rat model of renovascular hypertension, focusing on mediators controlling mitochondrial dynamics and autophagy. Rats with two-kidney one-clip (2K1C) hypertension were treated with fenofibrate 150 mg/kg/day (2K1C-FFB) or vehicle (2K1C-VEH) for 8 weeks. Systolic blood pressure and cardiac functional were in-vivo assessed, while cardiomyocyte size and protein expression of mediators of cardiac hypertrophy and mitochondrial dynamics were ex-vivo examined by histological and Western blot analyses. Fenofibrate treatment counteracted the development of hypertension and the increase of left ventricular mass, relative wall thickness and cross-sectional area of cardiomyocytes. Furthermore, fenofibrate re-balanced the expression Mfn2, Drp1 and Parkin, regulators of fusion, fission, mitophagy respectively. Regarding autophagy, the LC3-II/LC3-I ratio was increased in 2K1C-VEH and 2K1C-FFB, whereas the autophagy was increased only in 2K1C-FFB. In cultured H9C2 cardiomyoblasts, fenofibrate reversed the Ang II-induced mRNA up-regulation of hypertrophy markers Nppa and Myh7, accumulation of reactive oxygen species and depolarization of the mitochondrial membrane exerting protection mediated by up-regulation of the Uncoupling protein 2. Our results indicate that fenofibrate acts directly on cardiomyocytes and counteracts the pressure overload-induced cardiac maladaptive remodeling. This study reveals a so far hidden mechanism involving mitochondrial dynamics in the beneficial effects of fenofibrate, support its repurposing for the treatment of cardiac hypertrophy and provide new potential targets for its pharmacological function.
Collapse
Affiliation(s)
- Laura Castiglioni
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Paolo Gelosa
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Majeda Muluhie
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | | | - Joanna Rzemieniec
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Marco Gotti
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Fabio Fiordaliso
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Giuseppe Busca
- Azienda "Polo Veterinario di Lodi", University of Milan, Milan, Italy
| | - Luigi Sironi
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy.
| |
Collapse
|
12
|
Fatima M, Andleeb H, Rehman T, Gul O, Azeezz S, Rehman H, Nawaz H. Effect of alantolactones on cardiac parameters of animals under artificially induced oxidative stress. Biomedicine (Taipei) 2024; 14:12-22. [PMID: 39386184 PMCID: PMC11460572 DOI: 10.37796/2211-8039.1457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 10/12/2024] Open
Abstract
Purpose Phytochemicals have been found effective in reducing the oxidative stress and damage to cardiovascular and other tissues. In this study, the effects of alantolactone (AL) on cardiac parameters in rabbits exposed to artificially-induced oxidative stress were investigated. Method The oxidative stress was induced in a group of White New Zealand rabbits by injecting 40% hydrogen peroxide solution (1 ml/kg body weight) thrice with an interval of 72 h. The hydrogen peroxide-treated animals were orally treated with AL extracted from the roots of Inula helenium (1 ml/kg repeated thrice after 72 h). Blood samples were taken before and after the hydrogen peroxide and AL treatments, and the sera were subjected to analysis of oxidative damage in terms of malondialdehyde content (MDA), total antioxidant activity (TAOA), linoleic acid reduction capacity (LARC), hydroxyl radical scavenging capacity (HRSC), 2,2-diphenyl-1-picrylhydrazyl radical scavenging capacity (DPPH RSC), superoxide dismutase activity (SOD) and catalase activity, and cardiac parameters including troponin-I content (Trop-I), creatine kinase-MB (CKMB), aspartate transaminase (AST). Results The hydrogen peroxide treatment substantially enhanced MDA content and SOD activity and decreased LARC, HRSC, DPPH, and catalase activity. The AL treatment significantly decreased MDA content, TAOA, Trop-I, CK-MB, and AST levels and increased LARC, DPPH RSC, HRSC, and catalase activity. Conclusion The observed effect of AL treatment on the animals' oxidative stress, antioxidant status, and cardiac biomarkers emphasizes that AL may effectively manage oxidative stress and cardiac damage.
Collapse
Affiliation(s)
- Mishal Fatima
- Department of Biochemistry, Bahauddin Zakariya University, Multan, 60800,
Pakistan
| | - Hina Andleeb
- Department of Biochemistry, Bahauddin Zakariya University, Multan, 60800,
Pakistan
| | - Tanzila Rehman
- Department of Chemistry, The Women University Multan, Multan,
Pakistan
| | - Ouz Gul
- Department of Biochemistry, Bahauddin Zakariya University, Multan, 60800,
Pakistan
| | - Shanza Azeezz
- Department of Biochemistry, Bahauddin Zakariya University, Multan, 60800,
Pakistan
| | - Huzaifa Rehman
- Department of Biochemistry, Bahauddin Zakariya University, Multan, 60800,
Pakistan
| | - Haq Nawaz
- Department of Biochemistry, Bahauddin Zakariya University, Multan, 60800,
Pakistan
| |
Collapse
|
13
|
McClendon LK, Lanz RB, Panigrahi A, Gomez K, Bolt MJ, Liu M, Stossi F, Mancini MA, Dacso CC, Lonard DM, O'Malley BW. Transcriptional coactivation of NRF2 signaling in cardiac fibroblasts promotes resistance to oxidative stress. J Mol Cell Cardiol 2024; 194:70-84. [PMID: 38969334 DOI: 10.1016/j.yjmcc.2024.07.001] [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: 11/06/2023] [Revised: 06/17/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
We recently discovered that steroid receptor coactivators (SRCs) SRCs-1, 2 and 3, are abundantly expressed in cardiac fibroblasts (CFs) and their activation with the SRC small molecule stimulator MCB-613 improves cardiac function and dramatically lowers pro-fibrotic signaling in CFs post-myocardial infarction. These findings suggest that CF-derived SRC activation could be beneficial in the mitigation of chronic heart failure after ischemic insult. However, the cardioprotective mechanisms by which CFs contribute to cardiac pathological remodeling are unclear. Here we present studies designed to identify the molecular and cellular circuitry that governs the anti-fibrotic effects of an MCB-613 derivative, MCB-613-10-1, in CFs. We performed cytokine profiling and whole transcriptome and proteome analyses of CF-derived signals in response to MCB-613-10-1. We identified the NRF2 pathway as a direct MCB-613-10-1 therapeutic target for promoting resistance to oxidative stress in CFs. We show that MCB-613-10-1 promotes cell survival of anti-fibrotic CFs exposed to oxidative stress by suppressing apoptosis. We demonstrate that an increase in HMOX1 expression contributes to CF resistance to oxidative stress-mediated apoptosis via a mechanism involving SRC co-activation of NRF2, hence reducing inflammation and fibrosis. We provide evidence that MCB-613-10-1 acts as a protectant against oxidative stress-induced mitochondrial damage. Our data reveal that SRC stimulation of the NRF2 transcriptional network promotes resistance to oxidative stress and highlights a mechanistic approach toward addressing pathologic cardiac remodeling.
Collapse
Affiliation(s)
- Lisa K McClendon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America.
| | - Rainer B Lanz
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America.
| | - Anil Panigrahi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America.
| | - Kristan Gomez
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America.
| | - Michael J Bolt
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America.
| | - Min Liu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America.
| | - Fabio Stossi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America.
| | - Michael A Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America.
| | - Clifford C Dacso
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America.
| | - David M Lonard
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America.
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America.
| |
Collapse
|
14
|
Jang YJ, Kim HY, Na SW, Hong MH, Yoon JJ, Lee HS, Kang DG. The Cardioprotective Potential of Herbal Formulas in Myocardial Infarction-Induced Heart Failure through Inhibition of JAK/STAT3 Signaling and Improvement of Cardiac Function. Pharmaceuticals (Basel) 2024; 17:1132. [PMID: 39338297 PMCID: PMC11434789 DOI: 10.3390/ph17091132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/20/2024] [Accepted: 08/24/2024] [Indexed: 09/30/2024] Open
Abstract
Myocardial infarction (MI) is a leading cause of heart failure, characterized by adverse cardiac remodeling. This study evaluated the cardioprotective potential of Dohongsamul-tang (DHT), a traditional Korean herbal formula, in a rat model of MI-induced heart failure. Rats underwent left anterior descending (LAD) artery ligation and were treated with either 100 mg/kg or 200 mg/kg of DHT daily for 8 weeks. DHT treatment significantly improved cardiac function, as evidenced by increased ejection fraction (EF) from 62.1% to 70.1% (100 mg/kg) and fractional shortening (FS) from 32.3% to 39.4% (200 mg/kg) compared to the MI control group. Additionally, DHT reduced infarct size by approximately 63.3% (from 60.0% to 22.0%) and heart weight by approximately 16.7% (from 3.6 mg/g to 3.0 mg/g), and significantly decreased levels of heart failure biomarkers: LDH was reduced by 37.6% (from 1409.1 U/L to 879.1 U/L) and CK-MB by 47.6% (from 367.3 U/L to 192.5 U/L). Histological analysis revealed a reduction in left ventricle (LV) fibrosis by approximately 50% (from 24.0% to 12.0%). At the molecular level, DHT inhibited the expression of phospho-JAK by 75% (from 2-fold to 0.5-fold), phospho-STAT3 by 30.8% (from 1.3-fold to 0.9-fold), Bax/Bcl-2 by 56.3% (from 3.2-fold to 1.4-fold), and caspase-3 by 46.3% (from 1.23-fold to 0.66-fold). These results suggest that DHT exerts cardioprotective effects by modulating the JAK/STAT3 signaling pathway, highlighting its potential as a therapeutic option for heart failure.
Collapse
Affiliation(s)
- Youn-Jae Jang
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan 54538, Republic of Korea; (Y.-J.J.); (H.-Y.K.); (S.-W.N.); (M.-H.H.); (J.-J.Y.)
- College of Oriental Medicine, Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, Republic of Korea
| | - Hye-Yoom Kim
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan 54538, Republic of Korea; (Y.-J.J.); (H.-Y.K.); (S.-W.N.); (M.-H.H.); (J.-J.Y.)
| | - Se-Won Na
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan 54538, Republic of Korea; (Y.-J.J.); (H.-Y.K.); (S.-W.N.); (M.-H.H.); (J.-J.Y.)
- College of Oriental Medicine, Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, Republic of Korea
| | - Mi-Hyeon Hong
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan 54538, Republic of Korea; (Y.-J.J.); (H.-Y.K.); (S.-W.N.); (M.-H.H.); (J.-J.Y.)
| | - Jung-Joo Yoon
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan 54538, Republic of Korea; (Y.-J.J.); (H.-Y.K.); (S.-W.N.); (M.-H.H.); (J.-J.Y.)
| | - Ho-Sub Lee
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan 54538, Republic of Korea; (Y.-J.J.); (H.-Y.K.); (S.-W.N.); (M.-H.H.); (J.-J.Y.)
- College of Oriental Medicine, Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, Republic of Korea
| | - Dae-Gill Kang
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan 54538, Republic of Korea; (Y.-J.J.); (H.-Y.K.); (S.-W.N.); (M.-H.H.); (J.-J.Y.)
- College of Oriental Medicine, Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, Republic of Korea
| |
Collapse
|
15
|
Witmer NH, McLendon JM, Stein CS, Yoon JY, Berezhnaya E, Elrod JW, London BL, Boudreau RL. Upstream alternative polyadenylation in SCN5A produces a short transcript isoform encoding a mitochondria-localized NaV1.5 N-terminal fragment that influences cardiomyocyte respiration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.09.607406. [PMID: 39211120 PMCID: PMC11360925 DOI: 10.1101/2024.08.09.607406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
SCN5A encodes the cardiac voltage-gated Na+ channel, NaV1.5, that initiates action potentials. SCN5A gene variants cause arrhythmias and increased heart failure risk. Mechanisms controlling NaV1.5 expression and activity are not fully understood. We recently found a well-conserved alternative polyadenylation (APA) signal downstream of the first SCN5A coding exon. This yields a SCN5A-short transcript isoform expressed in several species (e.g. human, pig, and cat), though rodents lack this upstream APA. Reanalysis of transcriptome-wide cardiac APA-seq and mRNA-seq data shows reductions in both upstream APA usage and short/full-length SCN5A mRNA ratios in failing hearts. Knock-in of the human SCN5A APA sequence into mice is sufficient to enable expression of SCN5A -short transcript, while significantly decreasing expression of full-length SCN5A mRNA. Notably, SCN5A -short transcript encodes a novel protein (NaV1.5-NT), composed of an N-terminus identical to NaV1.5 and a unique C-terminus derived from intronic sequence. AAV9 constructs were able to achieve stable NaV1.5-NT expression in mouse hearts, and western blot of human heart tissues showed bands co-migrating with NaV1.5-NT transgene-derived bands. NaV1.5-NT is predicted to contain a mitochondrial targeting sequence and localizes to mitochondria in cultured cardiomyocytes and in mouse hearts. NaV1.5-NT expression in cardiomyocytes led to elevations in basal oxygen consumption rate, ATP production, and mitochondrial ROS, while depleting NADH supply. Native PAGE analyses of mitochondria lysates revealed that NaV1.5-NT expression resulted in increased levels of disassembled complex V subunits and accumulation of complex I-containing supercomplexes. Overall, we discovered that APA-mediated regulation of SCN5A produces a short transcript encoding NaV1.5-NT. Our data support that NaV1.5-NT plays a multifaceted role in influencing mitochondrial physiology: 1) by increasing basal respiration likely through promoting complex V conformations that enhance proton leak, and 2) by increasing overall respiratory efficiency and NADH consumption by enhancing formation and/or stability of complex I-containing respiratory supercomplexes, though the specific molecular mechanisms underlying each of these remain unresolved.
Collapse
|
16
|
Cao H, Zhao L, Yuan Y, Liao C, Zeng W, Li A, Huang Q, Zhao Y, Fan Y, Jiang L, Song D, Li S, Zhang B. Lipoamide Attenuates Hypertensive Myocardial Hypertrophy Through PI3K/Akt-Mediated Nrf2 Signaling Pathway. J Cardiovasc Transl Res 2024; 17:910-922. [PMID: 38334841 PMCID: PMC11371882 DOI: 10.1007/s12265-024-10488-9] [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: 09/21/2023] [Accepted: 01/29/2024] [Indexed: 02/10/2024]
Abstract
The process of myocardial hypertrophy in hypertension can lead to excessive activation of oxidative stress. Lipoamide (ALM) has significant antioxidant and anti-inflammatory effects. This study aimed to investigate the effects of ALM on hypertension-induced cardiac hypertrophy, as well as explore its underlying mechanisms. We evaluated the effects of ALM on spontaneously hypertensive rats and rat cardiomyocytes treated with Ang II. We found that ALM was not effective in lowering blood pressure in SHR, but it attenuated hypertension-mediated cardiac fibrosis, oxidative stress, inflammation, and hypertrophy in rats. After that, in cultured H9C2 cells stimulated with Ang II, ALM increased the expression of antioxidant proteins that were decreased in the Ang II group. ALM also alleviated cell hypertrophy and the accumulation of ROS, while LY294002 partially abrogated these effects. Collectively, these results demonstrate that ALM could alleviate oxidative stress in cardiac hypertrophy, potentially through the activation of the PI3K/Akt-mediated Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Hongjuan Cao
- Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Lina Zhao
- Guizhou Medical University, Guiyang, Guizhou Province, China
- Department of Ultrasound Center, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Yao Yuan
- Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Chunyan Liao
- Guizhou Medical University, Guiyang, Guizhou Province, China
- Department of Ultrasound Center, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Weidan Zeng
- Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Aiyue Li
- Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Quanfeng Huang
- Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Yueyao Zhao
- Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Yubing Fan
- Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Liu Jiang
- Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Dandan Song
- Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Sha Li
- Guizhou Medical University, Guiyang, Guizhou Province, China
- Department of Ultrasound Center, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Bei Zhang
- Guizhou Medical University, Guiyang, Guizhou Province, China.
- Department of Ultrasound Center, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China.
| |
Collapse
|
17
|
Forte M, D'Ambrosio L, Schiattarella GG, Salerno N, Perrone MA, Loffredo FS, Bertero E, Pilichou K, Manno G, Valenti V, Spadafora L, Bernardi M, Simeone B, Sarto G, Frati G, Perrino C, Sciarretta S. Mitophagy modulation for the treatment of cardiovascular diseases. Eur J Clin Invest 2024; 54:e14199. [PMID: 38530070 DOI: 10.1111/eci.14199] [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: 01/07/2024] [Revised: 03/15/2024] [Accepted: 03/16/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Defects of mitophagy, the selective form of autophagy for mitochondria, are commonly observed in several cardiovascular diseases and represent the main cause of mitochondrial dysfunction. For this reason, mitophagy has emerged as a novel and potential therapeutic target. METHODS In this review, we discuss current evidence about the biological significance of mitophagy in relevant preclinical models of cardiac and vascular diseases, such as heart failure, ischemia/reperfusion injury, metabolic cardiomyopathy and atherosclerosis. RESULTS Multiple studies have shown that cardiac and vascular mitophagy is an adaptive mechanism in response to stress, contributing to cardiovascular homeostasis. Mitophagy defects lead to cell death, ultimately impairing cardiac and vascular function, whereas restoration of mitophagy by specific compounds delays disease progression. CONCLUSIONS Despite previous efforts, the molecular mechanisms underlying mitophagy activation in response to stress are not fully characterized. A comprehensive understanding of different forms of mitophagy active in the cardiovascular system is extremely important for the development of new drugs targeting this process. Human studies evaluating mitophagy abnormalities in patients at high cardiovascular risk also represent a future challenge.
Collapse
Affiliation(s)
| | - Luca D'Ambrosio
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Gabriele G Schiattarella
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Nadia Salerno
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Marco Alfonso Perrone
- Division of Cardiology and CardioLab, Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
- Clinical Pathways and Epidemiology Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Francesco S Loffredo
- Division of Cardiology, Department of Translational Medical Sciences, University of Campania "L. Vanvitelli", Naples, Italy
| | - Edoardo Bertero
- Department of Internal Medicine, University of Genova, Genoa, Italy
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino-Italian IRCCS Cardiology Network, Genoa, Italy
| | - Kalliopi Pilichou
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Girolamo Manno
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Valentina Valenti
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- ICOT Istituto Marco Pasquali, Latina, Italy
| | | | - Marco Bernardi
- Department of Clinical, Internal Medicine, Anesthesiology and Cardiovascular Sciences, Sapienza University, Rome, Italy
| | | | | | - Giacomo Frati
- IRCCS Neuromed, Pozzilli, Italy
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Cinzia Perrino
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - Sebastiano Sciarretta
- IRCCS Neuromed, Pozzilli, Italy
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| |
Collapse
|
18
|
Wang X, Sun L. REEP5 mediates the function of CLEC5A to alleviate myocardial infarction by inhibiting endoplasmic reticulum stress-induced apoptosis. BMC Cardiovasc Disord 2024; 24:382. [PMID: 39044150 PMCID: PMC11265427 DOI: 10.1186/s12872-024-04018-3] [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: 12/01/2023] [Accepted: 06/27/2024] [Indexed: 07/25/2024] Open
Abstract
MI (myocardial infarction) often triggers severe heart failure and is one of the leading causes of death worldwide. Receptor expression-enhancing protein 5 (REEP5), a member of REEPs, acts as regulators of endoplasmic reticulum (ER) affecting cardiac functions. Based on GSE114695 profile data, REEP5 was decreased in the left ventricle of MI mice. However, its role and potential mechanism in MI remain to be investigated. In the present study, the mouse MI model was established by ligation of the left anterior descending artery. REEP5 expression was downregulated in the infarct penumbra area of MI mice. Next, its role during MI was explored by gain-of-function. Interestingly, REEP5 overexpression improved left ventricular function of mice with MI, accompanied with reduced infarct size. In cardiomyocytes, REEP5 overexpression inhibited ER stress, accompanied with repressive phosphorylation of PERK and IRE1α, and the decreased nuclear translocation of ATF6. Subsequently, REEP5 overexpression downregulated the levels of Chop and cleaved caspase-12, further alleviating ER stress-induced apoptosis, which was consistent with the in vivo results. Moreover, REEP5 was found to bind to C-type lectin member 5 A (CLEC5A), a protein that triggers cardiac dysfunction. CLEC5A, whose expression was elevated in hypoxia-induced cell models, led to cardiomyocyte apoptosis. Noteworthily, REEP5 overexpression markedly abolished the effects of CLEC5A on ER stress-induced apoptosis. Taken together, REEP5 mediated the function of CLEC5A to relieve MI via inhibiting ER stress-induced apoptosis in vivo and in vitro. REEP5 may be a promising target for treating MI.
Collapse
Affiliation(s)
- Xin Wang
- Department of Cardiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Limin Sun
- Department of General Practice, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China.
| |
Collapse
|
19
|
Islam KN, Nguyen ID, Islam R, Pirzadah H, Malik H. Roles of Hydrogen Sulfide (H2S) as a Potential Therapeutic Agent in Cardiovascular Diseases: A Narrative Review. Cureus 2024; 16:e64913. [PMID: 39156383 PMCID: PMC11330631 DOI: 10.7759/cureus.64913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2024] [Indexed: 08/20/2024] Open
Abstract
Cardiovascular disease (CVD) stands as one of the leading causes of morbidity and mortality worldwide, and the continued search for novel therapeutics is vital for addressing this global health challenge. Over the past decade, hydrogen sulfide (H₂S) has garnered significant attention in the field of medical research, as it has been proven to be a cardioprotective gaseous signaling molecule. It joins nitric oxide and carbon monoxide as endogenously produced gasotransmitters. As for its mechanism, H₂S functions through the posttranslational addition of a sulfur group to cysteine residues on target proteins in a process called sulfhydration. As a result, the observed physiological effects of H₂S can include vasodilation, anti-apoptosis, anti-inflammation, antioxidant effects, and regulation of ion channels. Various studies have observed the cardioprotective benefits of H₂S in diseases such as myocardial infarction, ischemia-reperfusion injury, cardiac remodeling, heart failure, arrhythmia, and atherosclerosis. In this review, we discuss the mechanisms and therapeutic potential of H₂S in various CVDs.
Collapse
Affiliation(s)
- Kazi N Islam
- Department of Agricultural Research Development Program, Central State University, Wilberforce, USA
| | - Ivan D Nguyen
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA
| | - Rahib Islam
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA
| | - Humza Pirzadah
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA
| | - Hassan Malik
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA
| |
Collapse
|
20
|
Wang X, Zhao X, Wang X, Cao L, Lu B, Wang Z, Zhang W, Ti Y, Zhong M. Effect of levosimendan on ventricular remodelling in patients with left ventricular systolic dysfunction: a meta-analysis. ESC Heart Fail 2024; 11:1352-1376. [PMID: 38419326 PMCID: PMC11098670 DOI: 10.1002/ehf2.14714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/10/2024] [Accepted: 01/18/2024] [Indexed: 03/02/2024] Open
Abstract
Heart failure is the final stage of several cardiovascular diseases, and the key to effectively treating heart failure is to reverse or delay ventricular remodelling. Levosimendan is a novel inotropic and vasodilator agent used in heart failure, whereas the impact of levosimendan on ventricular remodelling is still unclear. This study aims to investigate the impact of levosimendan on ventricular remodelling in patients with left ventricular systolic dysfunction. Electronic databases were searched to identify eligible studies. A total of 66 randomized controlled trials involving 7968 patients were included. Meta-analysis results showed that levosimendan increased left ventricular ejection fraction [mean difference (MD) = 3.62, 95% confidence interval (CI) (2.88, 4.35), P < 0.00001] and stroke volume [MD = 6.59, 95% CI (3.22, 9.96), P = 0.0001] and significantly reduced left ventricular end-systolic volume [standard mean difference (SMD) = -0.52, 95% CI (-0.67, -0.37), P < 0.00001], left ventricular end-diastolic volume index [SMD = -1.24, 95% CI (-1.61, -0.86), P < 0.00001], and left ventricular end-systolic volume index [SMD = -1.06, 95% CI (-1.43, -0.70), P < 0.00001]. In terms of biomarkers, levosimendan significantly reduced the level of brain natriuretic peptide [SMD = -1.08, 95% CI (-1.60, -0.56), P < 0.0001], N-terminal pro-brain natriuretic peptide [SMD = -0.99, 95% CI (-1.41, -0.56), P < 0.00001], and interleukin-6 [SMD = -0.61, 95% CI (-0.86, -0.35), P < 0.00001]. Meanwhile, levosimendan may increase the incidence of hypotension [risk ratio (RR) = 1.24, 95% CI (1.12, 1.39), P < 0.0001], hypokalaemia [RR = 1.57, 95% CI (1.08, 2.28), P = 0.02], headache [RR = 1.89, 95% CI (1.50, 2.39), P < 0.00001], atrial fibrillation [RR = 1.31, 95% CI (1.12, 1.52), P = 0.0005], and premature ventricular complexes [RR = 1.86, 95% CI (1.27, 2.72), P = 0.001]. In addition, levosimendan reduced all-cause mortality [RR = 0.83, 95% CI (0.74, 0.94), P = 0.002]. In conclusion, our study found that levosimendan might reverse ventricular remodelling when applied in patients with left ventricular systolic dysfunction, especially in patients undergoing cardiac surgery, decompensated heart failure, and septic shock.
Collapse
Affiliation(s)
- Xia Wang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of CardiologyQilu Hospital of Shandong UniversityChina
| | - Xiu‐Zhi Zhao
- Department of CardiologyPeople's Hospital of Lixia District of JinanJinanShandongChina
| | - Xi‐Wen Wang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of CardiologyQilu Hospital of Shandong UniversityChina
| | - Lu‐Ying Cao
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of CardiologyQilu Hospital of Shandong UniversityChina
| | - Bin Lu
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of CardiologyQilu Hospital of Shandong UniversityChina
| | - Zhi‐Hao Wang
- Department of Geriatric MedicineShandong Key Laboratory of Cardiovascular Proteomics, Qilu Hospital, Cheeloo College of Medicine, Shandong UniversityJinanChina
| | - Wei Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of CardiologyQilu Hospital of Shandong UniversityChina
| | - Yun Ti
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of CardiologyQilu Hospital of Shandong UniversityChina
| | - Ming Zhong
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of CardiologyQilu Hospital of Shandong UniversityChina
| |
Collapse
|
21
|
Pate BS, Smiley CE, Harrington EN, Bielicki BH, Davis JM, Reagan LP, Grillo CA, Wood SK. Voluntary wheel running as a promising strategy to promote autonomic resilience to social stress in females: Vagal tone lies at the heart of the matter. Auton Neurosci 2024; 253:103175. [PMID: 38677130 PMCID: PMC11173375 DOI: 10.1016/j.autneu.2024.103175] [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: 12/13/2023] [Revised: 03/06/2024] [Accepted: 04/09/2024] [Indexed: 04/29/2024]
Abstract
Social stress is a major risk factor for comorbid conditions including cardiovascular disease and depression. While women exhibit 2-3× the risk for these stress-related disorders compared to men, the mechanisms underlying heightened stress susceptibility among females remain largely unknown. Due to a lack in understanding of the pathophysiology underlying stress-induced comorbidities among women, there has been a significant challenge in developing effective therapeutics. Recently, a causal role for inflammation has been established in the onset and progression of comorbid cardiovascular disease/depression, with women exhibiting increased sensitivity to stress-induced immune signaling. Importantly, reduced vagal tone is also implicated in stress susceptibility, through a reduction in the vagus nerve's well-recognized anti-inflammatory properties. Thus, examining therapeutic strategies that stabilize vagal tone during stress may shed light on novel targets for promoting stress resilience among women. Recently, accumulating evidence has demonstrated that physical activity exerts cardio- and neuro-protective effects by enhancing vagal tone. Based on this evidence, this mini review provides an overview of comorbid cardiovascular and behavioral dysfunction in females, the role of inflammation in these disorders, how stress may impart its negative effects on the vagus nerve, and how exercise may act as a preventative. Further, we highlight a critical gap in the literature with regard to the study of females in this field. This review also presents novel data that are the first to demonstrate a protective role for voluntary wheel running over vagal tone and biomarkers of cardiac dysfunction in the face of social stress exposure in female rats.
Collapse
Affiliation(s)
- Brittany S Pate
- Department of Exercise Science, University of South Carolina, Columbia, SC, United States of America; Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States of America; Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Cora E Smiley
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States of America; Columbia VA Health Care System, Columbia, SC, United States of America
| | - Evelynn N Harrington
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States of America; Columbia VA Health Care System, Columbia, SC, United States of America
| | - B Hunter Bielicki
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States of America; Columbia VA Health Care System, Columbia, SC, United States of America
| | - J Mark Davis
- Department of Exercise Science, University of South Carolina, Columbia, SC, United States of America
| | - Lawrence P Reagan
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States of America; Columbia VA Health Care System, Columbia, SC, United States of America
| | - Claudia A Grillo
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States of America; Columbia VA Health Care System, Columbia, SC, United States of America
| | - Susan K Wood
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States of America; Columbia VA Health Care System, Columbia, SC, United States of America; USC Institute for Cardiovascular Disease Research, Columbia, SC, United States of America.
| |
Collapse
|
22
|
Ostrowska-Leśko M, Herbet M, Pawłowski K, Korga-Plewko A, Poleszak E, Dudka J. Pathological Changes and Metabolic Adaptation in the Myocardium of Rats in Response to Chronic Variable Mild Stress. Int J Mol Sci 2024; 25:5899. [PMID: 38892086 PMCID: PMC11172974 DOI: 10.3390/ijms25115899] [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: 04/12/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Chronic variable mild stress (CVS) in rats is a well-established paradigm for inducing depressive-like behaviors and has been utilized extensively to explore potential therapeutic interventions for depression. While the behavioral and neurobiological effects of CVS have been extensively studied, its impact on myocardial function remains largely unexplored. To induce the CVS model, rats were exposed to various stressors over 40 days. Behavioral assessments confirmed depressive-like behavior. Biochemical analyses revealed alterations in myocardial metabolism, including changes in NAD+ and NADP+, and NADPH concentrations. Free amino acid analysis indicated disturbances in myocardial amino acid metabolism. Evaluation of oxidative DNA damage demonstrated an increased number of abasic sites in the DNA of rats exposed to CVS. Molecular analysis showed significant changes in gene expression associated with glucose metabolism, oxidative stress, and cardiac remodeling pathways. Histological staining revealed minor morphological changes in the myocardium of CVS-exposed rats, including increased acidophilicity of cells, collagen deposition surrounding blood vessels, and glycogen accumulation. This study provides novel insights into the impact of chronic stress on myocardial function and metabolism, highlighting potential mechanisms linking depression and cardiovascular diseases. Understanding these mechanisms may aid in the development of targeted therapeutic strategies to mitigate the adverse cardiovascular effects of depression.
Collapse
Affiliation(s)
- Marta Ostrowska-Leśko
- Department of Toxicology, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland; (M.H.); (J.D.)
| | - Mariola Herbet
- Department of Toxicology, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland; (M.H.); (J.D.)
| | - Kamil Pawłowski
- Department of Toxicology, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland; (M.H.); (J.D.)
| | - Agnieszka Korga-Plewko
- Independent Medical Biology Unit, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland
| | - Ewa Poleszak
- Department of Applied Pharmacy, Medical University of Lublin, 1 Chodźko Street, 20-093 Lublin, Poland
| | - Jarosław Dudka
- Department of Toxicology, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland; (M.H.); (J.D.)
| |
Collapse
|
23
|
Ciampi CM, Sultana A, Ossola P, Farina A, Fragasso G, Spoladore R. Current experimental and early investigational agents for cardiac fibrosis: where are we at? Expert Opin Investig Drugs 2024; 33:389-404. [PMID: 38426439 DOI: 10.1080/13543784.2024.2326024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/28/2024] [Indexed: 03/02/2024]
Abstract
INTRODUCTION Myocardial fibrosis (MF) is induced by factors activating pro-fibrotic pathways such as acute and prolonged inflammation, myocardial ischemic events, hypertension, aging process, and genetically-linked cardiomyopathies. Dynamics and characteristics of myocardial fibrosis development are very different. The broad range of myocardial fibrosis presentations suggests the presence of multiple potential targets. AREA COVERED Heart failure treatment involves medications primarily aimed at counteracting neurohormonal activation. While these drugs have demonstrated efficacy against MF, not all specifically target inflammation or fibrosis progression with some exceptions such as RAAS inhibitors. Consequently, new therapies are being developed to address this issue. This article is aimed to describe anti-fibrotic drugs currently employed in clinical practice and emerging agents that target specific pathways, supported by evidence from both preclinical and clinical studies. EXPERT OPINION Despite various preclinical findings suggesting the potential utility of new drugs and molecules for treating cardiac fibrosis in animal models, there is a notable scarcity of clinical trials investigating these effects. However, the pathology of damage and repair in the heart muscle involves a complex network of interconnected inflammatory pathways and various types of immune cells. Our comprehension of the positive and negative roles played by specific immune cells and cytokines is an emerging area of research.
Collapse
Affiliation(s)
- Claudio M Ciampi
- Health Science Department, University of Milan Bicocca, Milano, Italy
| | - Andrea Sultana
- Health Science Department, University of Milan Bicocca, Milano, Italy
| | - Paolo Ossola
- Health Science Department, University of Milan Bicocca, Milano, Italy
| | - Andrea Farina
- Division of Cardiology, Alessandro Manzoni Hospital, ASST- Lecco, Italy
| | - Gabriele Fragasso
- Heart Failure Unit Head, Division of Cardiology, IRCCS Vita-Salute San Raffaele University Hospital, Milan, Italy
| | - Roberto Spoladore
- Division of Cardiology, Alessandro Manzoni Hospital, ASST- Lecco, Italy
| |
Collapse
|
24
|
Chen H, Cheng Z, Wang M, Huang Q, Zheng D, Huang Q, Cai K. Circ_0020887 Silencing Combats Hypoxic-Induced Cardiomyocyte Injury in an MiR-370-3p/CYP1B1-Dependent Manner. Int Heart J 2024; 65:308-317. [PMID: 38479850 DOI: 10.1536/ihj.23-325] [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] [Indexed: 04/02/2024]
Abstract
Targeting circular RNA has been a novel approach to preventing and limiting acute myocardial infarction (AMI). Here, we planned to investigate the role and mechanism of circ_0020887 in AMI progression.Hypoxic injury in human cardiomyocytes (AC16) was measured using cell counting kit-8 assay, 5-ethynyl-2'-deoxyuridine assay, flow cytometry, and colorimetric assay kits. RNA and protein expressions were determined using real-time quantitative PCR and western blotting. Direct interplay between RNAs was determined using dual-luciferase reporter, RNA pull-down, and RIP assays.In the plasma and hypoxia-induced AC16 cells of patients with AMI, circ_0020887 and miR-370-3p were upregulated and downregulated, respectively, concomitant with the upregulation of cytochrome P450 1B1 (CYP1B1). Circ_0020887 interference could inhibit hypoxia-induced AC16 cell apoptosis, oxidative stress, and inflammatory response. Circ_0020887 could sponge miR-370-3p, and miR-370-3p could target CYP1B1. The inhibition effect of circ_0020887 knockdown on hypoxia-induced AC16 cell injury could be reversed by the miR-370-3p inhibitor. Besides, CYP1B1 overexpression also overturned the suppressive effect of miR-370-3p on hypoxia-induced AC16 cell apoptosis, oxidative stress, and inflammatory response.In conclusion, circ_0020887 regulated the miR-370-3p/CYP1B1 axis to regulate hypoxia-induced cardiomyocyte injury, confirming that circ_0020887 might promote cardiomyocyte injury.
Collapse
Affiliation(s)
- Huiqin Chen
- Department of Basic Medical, Quanzhou Medical College
| | - Zhendong Cheng
- Department of Cardiovascular, The Second Affiliated Hospital of Fujian Medical University
| | - Meiai Wang
- Department of Basic Medical, Quanzhou Medical College
| | - Qian Huang
- Department of Basic Medical, Quanzhou Medical College
| | - Dandan Zheng
- Department of Basic Medical, Quanzhou Medical College
| | - Qiuhong Huang
- Department of Basic Medical, Quanzhou Medical College
| | - Kefeng Cai
- Department of Cardiovascular, The Second Affiliated Hospital of Fujian Medical University
| |
Collapse
|
25
|
Bao H, Wang X, Zhou H, Zhou W, Liao F, Wei F, Yang S, Luo Z, Li W. PCSK9 regulates myofibroblast transformation through the JAK2/STAT3 pathway to regulate fibrosis after myocardial infarction. Biochem Pharmacol 2024; 220:115996. [PMID: 38154546 DOI: 10.1016/j.bcp.2023.115996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 11/20/2023] [Accepted: 12/18/2023] [Indexed: 12/30/2023]
Abstract
Cardiac fibrosis is pivotal in the progression of numerous cardiovascular diseases. This phenomenon is hallmarked by an excessive deposition of ECM protein secreted by myofibroblasts, leading to increased myocardial stiffness. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a serine protease that belongs to the proprotein-converting enzyme family. It has emerged as a viable therapeutic target for reducing plasma low-density lipoprotein cholesterol. However, the exact mechanism via which PCSK9 impacts cardiac fibrosis remains unclear. In the present research, an increase in circulating PCSK9 protein levels was observed in individuals with myocardial infarction and rat models of myocardial infarction. Moreover, the inhibition of circulating PCSK9 in rats was found to reduce post-infarction fibrosis. In vitro experiments further demonstrated that overexpression of PCSK9 or stimulation by extracellular PCSK9 recombinant protein enhanced the transformation of cardiac fibroblasts to myofibroblasts. This process also elevated collagen Ⅰ, and Ⅲ, as well as α-SMA protein levels. However, these effects were countered when co-incubated with the STAT3 inhibitor S3I-201. This study suggests that PCSK9 may function as a novel regulator of myocardial fibrosis, primarily via the JAK2/STAT3 pathway.
Collapse
Affiliation(s)
- Hailong Bao
- Department of Cardiovascular Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, China; Department of Cardiovascular Medicine, Gui Qian International General Hospital, Guiyang 550018, Guizhou, China
| | - Xu Wang
- Department of Cardiovascular Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, China; The Key Laboratory of Myocardial Remodeling Research, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, China
| | - Haiyan Zhou
- Department of Cardiovascular Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, China; The Key Laboratory of Myocardial Remodeling Research, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, China
| | - Wei Zhou
- Department of Cardiovascular Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, China; The Key Laboratory of Myocardial Remodeling Research, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, China
| | - Fujun Liao
- Department of Cardiovascular Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, China; The Key Laboratory of Myocardial Remodeling Research, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, China
| | - Fang Wei
- Department of Cardiovascular Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, China; The Key Laboratory of Myocardial Remodeling Research, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, China
| | - Shiyu Yang
- Department of Cardiovascular Medicine, Gui Qian International General Hospital, Guiyang 550018, Guizhou, China
| | - Zhenhua Luo
- Department of Central Lab, Guizhou Provincial People's Hospital, Guiyang 550002, Guizhou, China.
| | - Wei Li
- Department of Cardiovascular Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, China; The Key Laboratory of Myocardial Remodeling Research, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, China.
| |
Collapse
|
26
|
Chen Y, Peng W, Pang M, Zhu B, Liu H, Hu D, Luo Y, Wang S, Wu S, He J, Yang Y, Peng D. The effects of psychiatric disorders on the risk of chronic heart failure: a univariable and multivariable Mendelian randomization study. Front Public Health 2024; 12:1306150. [PMID: 38299073 PMCID: PMC10827915 DOI: 10.3389/fpubh.2024.1306150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 01/04/2024] [Indexed: 02/02/2024] Open
Abstract
Background Substantial evidence suggests an association between psychiatric disorders and chronic heart failure. However, further investigation is needed to confirm the causal relationship between these psychiatric disorders and chronic heart failure. To address this, we evaluated the potential effects of five psychiatric disorders on chronic heart failure using two-sample Mendelian Randomization (MR). Methods We selected single nucleotide polymorphisms (SNPs) associated with chronic heart failure and five psychiatric disorders (Attention-Deficit Hyperactivity Disorder (ADHD), Autism Spectrum Disorder (ASD), Major Depression, Bipolar Disorder and Schizophrenia (SCZ)). Univariable (UVMR) and multivariable two-sample Mendelian Randomization (MVMR) were employed to assess causality between these conditions. Ever smoked and alcohol consumption were controlled for mediating effects in the multivariable MR. The inverse variance weighting (IVW) and Wald ratio estimator methods served as the primary analytical methods for estimating potential causal effects. MR-Egger and weighted median analyses were also conducted to validate the results. Sensitivity analyses included the funnel plot, leave-one-out, and MR-Egger intercept tests. Additionally, potential mediators were investigated through risk factor analyses. Results Genetically predicted heart failure was significantly associated with ADHD (odds ratio (OR), 1.12; 95% CI, 1.04-1.20; p = 0.001), ASD (OR, 1.29; 95% CI, 1.07-1.56; p = 0.008), bipolar disorder (OR, 0.89; 95% CI, 0.83-0.96; p = 0.001), major depression (OR, 1.15; 95% CI, 1.03-1.29; p = 0.015), SCZ (OR, 1.04; 95% CI, 1.00-1.07; p = 0.024). Several risk factors for heart failure are implicated in the above cause-and-effect relationship, including ever smoked and alcohol consumption. Conclusion Our study demonstrated ADHD, ASD, SCZ and major depression may have a causal relationship with an increased risk of heart failure. In contrast, bipolar disorder was associated with a reduced risk of heart failure, which could potentially be mediated by ever smoked and alcohol consumption. Therefore, prevention strategies for heart failure should also incorporate mental health considerations, and vice versa.
Collapse
Affiliation(s)
- Yang Chen
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
| | - Wenke Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
| | - Min Pang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
| | - Botao Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
| | - Huixing Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
| | - Die Hu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
| | - Yonghong Luo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
| | - Shuai Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
| | - Sha Wu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
| | - Jia He
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
| | - Yang Yang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
| | - Daoquan Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
| |
Collapse
|
27
|
Luo G, Chen L, Chen M, Mao L, Zeng Q, Zou Y, Xue J, Liu P, Wu Q, Yang S, Liu M. Hirudin inhibit the formation of NLRP3 inflammasome in cardiomyocytes via suppressing oxidative stress and activating mitophagy. Heliyon 2024; 10:e23077. [PMID: 38163129 PMCID: PMC10754874 DOI: 10.1016/j.heliyon.2023.e23077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024] Open
Abstract
Context Cardiomyocyte hypertrophy due to hemodynamic overload eventually leads to heart failure. Hirudin has been widely used in the treatment of cardiovascular diseases and NLRP3 inflammasome was proven to induce cardiomyocyte pyroptosis. However, the mechanism by which it inhibits cardiomyocyte hypertrophy remains unclear. Objective To explore the mechanism of hirudin inhibiting cardiomyocyte hypertrophy based on NLRP3 inflammasome activation and mitophagy. Materials & methods 1 μM AngII was used for cardiac hypertrophy modeling in H9C2 cells, and cell viability was quantified by CCK-8 assay to screen the appropriate action concentrations of hirudin. After that, we cultured AngII induced-H9C2 cells for 24 h with 0, 0.3, 0.6, and 1.2 mM hirudin, respectively. Next, we marked H9C2 cells with phalloidine and observed them using fluorescence microscope. IL-1β, IL-18, IL-6, TNF-α, ANP, BNP, β-MHC, and mtDNA were analyzed by qRT-PCR; ROS were quantified by Flow cytometry; SOD, MDA, and GSH-Px were detected by ELISA; and proteins including NLRP3, ASC, caspase-1, pro-caspase-1, IL-1β, IL-18, PINK-1, Parkin, beclin-1, LC3-Ⅰ, LC3-Ⅱ, p62, were quantified by western blotting. Results It was discovered that hirudin reduced the superficial area of AngII-induced H9C2 cells and inhibited the AngII-induced up-regulation of ANP, BNP, and β-MHC. Besides, hirudin down-regulated the expressions of NLRP3 inflammasome-related cytokines, containing IL-1β, IL-18, IL-6, TNF-α. It also down-regulated the expression of mtDNA and ROS, decreased the expression levels of NLRP3 inflammasome activation related proteins, including NLRP3, ASC, caspase-1, pro-caspase-1, IL-1β, IL-18; and increased the expressions of PINK-1, Parkin, beclin-1, LC3-Ⅱ/LC3-Ⅰ, p62 in AngII-induced H9C2 cells. Discussion Hirudin promoted the process of mitophagy, inhibited the development of inflammation and oxidative stress, and inhibited the activation of the NLRP3 inflammasome and the PINK-1/Parkin pathway. Conclusion Hirudin has the activity to suppress cardiac hypertrophy may benefit from the inhibition of NLRP3 inflammasome and activating of PINK-1/Parkin related-mitophagy.
Collapse
Affiliation(s)
- Gang Luo
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, PR China
| | - Li Chen
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, PR China
| | - Mingtai Chen
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Tapai, Macau
- Department of Cardiovascular Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Linshen Mao
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, PR China
| | - Qihu Zeng
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, PR China
| | - Yuan Zou
- School of Integrated Traditional and Western Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Jinyi Xue
- School of Integrated Traditional and Western Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Ping Liu
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, PR China
| | - Qibiao Wu
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Tapai, Macau
| | - Sijin Yang
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, PR China
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Tapai, Macau
| | - Mengnan Liu
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, PR China
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Tapai, Macau
| |
Collapse
|
28
|
Boshra SA, Nazeam JA, Esmat A. Flaxseed oil fraction reverses cardiac remodeling at a molecular level: improves cardiac function, decreases apoptosis, and suppresses miRNA-29b and miRNA 1 gene expression. BMC Complement Med Ther 2024; 24:6. [PMID: 38167049 PMCID: PMC10759513 DOI: 10.1186/s12906-023-04319-8] [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: 08/01/2023] [Accepted: 12/17/2023] [Indexed: 01/05/2024] Open
Abstract
Flaxseed is an ancient commercial oil that historically has been used as a functional food to lower cholesterol levels. However, despite its longstanding treatment, there is currently a lack of scientific evidence to support its role in the management of cardiac remodeling. This study aimed to address this gap in knowledge by examining the molecular mechanism of standardized flaxseed oil in restoring cardiac remodeling in the heart toxicity vivo model. The oil fraction was purified, and the major components were standardized by qualitative and quantitative analysis. In vivo experimental design was conducted using isoproterenol ISO (85 mg/kg) twice subcutaneously within 24 h between each dose. The rats were treated with flaxseed oil fraction (100 mg/kg orally) and the same dose was used for omega 3 supplement as a positive control group. The GC-MS analysis revealed that α-linolenic acid (24.6%), oleic acid (10.5%), glycerol oleate (9.0%) and 2,3-dihydroxypropyl elaidate (7%) are the major components of oil fraction. Physicochemical analysis indicated that the acidity percentage, saponification, peroxide, and iodine values were 0.43, 188.57, 1.22, and 122.34 respectively. As compared with healthy control, ISO group-induced changes in functional cardiac parameters. After 28-day pretreatment with flaxseed oil, the results indicated an improvement in cardiac function, a decrease in apoptosis, and simultaneous prevention of myocardial fibrosis. The plasma levels of BNP, NT-pro-BNP, endothelin-1, Lp-PLA2, and MMP2, and cTnI and cTn were significantly diminished, while a higher plasma level of Topo 2B was observed. Additionally, miRNA - 1 and 29b were significantly downregulated. These findings provide novel insight into the mechanism of flaxseed oil in restoring cardiac remodeling and support its future application as a cardioprotective against heart diseases.
Collapse
Affiliation(s)
- Sylvia A Boshra
- Biochemistry Department, Faculty of Pharmacy, October 6 University, 6 of October City, Giza, 12585, Egypt.
| | - Jilan A Nazeam
- Pharmacognosy Department, Faculty of Pharmacy, October 6 University, 6 of October City, Giza, 12585, Egypt.
| | - Ahmed Esmat
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| |
Collapse
|
29
|
Kumar R, Chhillar N, Gupta DS, Kaur G, Singhal S, Chauhan T. Cholesterol Homeostasis, Mechanisms of Molecular Pathways, and Cardiac Health: A Current Outlook. Curr Probl Cardiol 2024; 49:102081. [PMID: 37716543 DOI: 10.1016/j.cpcardiol.2023.102081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Abstract
The metabolism of lipoproteins, which regulate the transit of the lipid to and from tissues, is crucial to maintaining cholesterol homeostasis. Cardiac remodeling is referred to as a set of molecular, cellular, and interstitial changes that, following injury, affect the size, shape, function, mass, and geometry of the heart. Acetyl coenzyme A (acetyl CoA), which can be made from glucose, amino acids, or fatty acids, is the precursor for the synthesis of cholesterol. In this article, the authors explain concepts behind cardiac remodeling, its clinical ramifications, and the pathophysiological roles played by numerous various components, such as cell death, neurohormonal activation, oxidative stress, contractile proteins, energy metabolism, collagen, calcium transport, inflammation, and geometry. The levels of cholesterol are traditionally regulated by 2 biological mechanisms at the transcriptional stage. First, the SREBP transcription factor family regulates the transcription of crucial rate-limiting cholesterogenic and lipogenic proteins, which in turn limits cholesterol production. Immune cells become activated, differentiated, and divided, during an immune response with the objective of eradicating the danger signal. In addition to creating ATP, which is used as energy, this process relies on metabolic reprogramming of both catabolic and anabolic pathways to create metabolites that play a crucial role in regulating the response. Because of changes in signal transduction, malfunction of the sarcoplasmic reticulum and sarcolemma, impairment of calcium handling, increases in cardiac fibrosis, and progressive loss of cardiomyocytes, oxidative stress appears to be the primary mechanism that causes the transition from cardiac hypertrophy to heart failure. De novo cholesterol production, intestinal cholesterol absorption, and biliary cholesterol output are consequently crucial processes in cholesterol homeostasis. In the article's final section, the pharmacological management of cardiac remodeling is explored. The route of treatment is explained in different steps: including, promising, and potential strategies. This chapter offers a brief overview of the history of the study of cholesterol absorption as well as the different potential therapeutic targets.
Collapse
Affiliation(s)
| | - Neelam Chhillar
- Deparetment of Biochemistry, School of Medicine, DY Patil University, Navi Mumbai, India
| | - Dhruv Sanjay Gupta
- Department of Pharmacology, SPP School of Pharmacy & Technology Management, SVKM's NMIMS, Mumbai, India
| | - Ginpreet Kaur
- Department of Pharmacology, SPP School of Pharmacy & Technology Management, SVKM's NMIMS, Mumbai, India
| | - Shailey Singhal
- Cluster of Applied Sciences, University of Petroleum and Energy Studies, Dehradun, India
| | - Tanya Chauhan
- Division of Forensic Biology, National Forensic Sciences University, Delhi Campus (LNJN NICFS) Delhi, India
| |
Collapse
|
30
|
Momeni Z, Danesh S, Ahmadpour M, Eshraghi R, Farkhondeh T, Pourhanifeh MH, Samarghandian S. Protective Roles and Therapeutic Effects of Gallic Acid in the Treatment of Cardiovascular Diseases: Current Trends and Future Directions. Curr Med Chem 2024; 31:3733-3751. [PMID: 37815180 DOI: 10.2174/0109298673259299230921150030] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/19/2023] [Accepted: 08/18/2023] [Indexed: 10/11/2023]
Abstract
Cardiovascular diseases (CVDs) are serious life-threatening illnesses and significant problematic issues for public health having a heavy economic burden on all society worldwide. The high incidence of these diseases as well as high mortality rates make them the leading causes of death and disability. Therefore, finding novel and more effective therapeutic methods is urgently required. Gallic acid, an herbal medicine with numerous biological properties, has been utilized in the treatment of various diseases for thousands of years. It has been demonstrated that gallic acid possesses pharmacological potential in regulating several molecular and cellular processes such as apoptosis and autophagy. Moreover, gallic acid has been investigated in the treatment of CVDs both in vivo and in vitro. Herein, we aimed to review the available evidence on the therapeutic application of gallic acid for CVDs including myocardial ischemia-reperfusion injury and infarction, drug-induced cardiotoxicity, hypertension, cardiac fibrosis, and heart failure, with a focus on underlying mechanisms.
Collapse
Affiliation(s)
- Zahra Momeni
- Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Sepideh Danesh
- Research Hub Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Ahmadpour
- Research Hub Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Eshraghi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Tahereh Farkhondeh
- Department of Toxicology and Pharmacology, School of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Hub Institute, Tehran University of Medical Sciences, Tehran, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Saeed Samarghandian
- University of Neyshabur Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
31
|
Zhang P, Liu X, Yu X, Zhuo Y, Li D, Yang L, Lu Y. Protective Effects of Liriodendrin on Myocardial Infarction-Induced Fibrosis in Rats via the PI3K/Akt Autophagy Pathway: A Network Pharmacology Study. Comb Chem High Throughput Screen 2024; 27:1566-1575. [PMID: 37461344 DOI: 10.2174/1386207326666230717155641] [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: 08/01/2022] [Revised: 05/31/2023] [Accepted: 05/31/2023] [Indexed: 07/04/2024]
Abstract
BACKGROUND Liriodendrin (LIR) has been reported to improve cardiac function in rats following myocardial infarction. However, its role and mechanism in reparative myocardial fibrosis remain unclear. METHODS In this study, a rat model of myocardial fibrosis was established via left anterior descending artery ligation and randomly divided into three groups (n = 6 per group): sham-operated, myocardial infarction, and LIR intervention (100 mg/kg/day) groups. The pharmacological effects of LIR were assessed using echocardiography, hematoxylin, and eosin (H&E) staining, and Masson staining. Network pharmacology and bioinformatics were utilized to identify potential mechanisms of LIR, which were further validated via western blot analysis. RESULTS Our findings demonstrated that LIR improved cardiac function, histology scores, and col lagen volume fraction. Moreover, LIR downregulated the expression of Beclin-1, LC3-II/LC3-I while upregulating the expression of p62, indicating LIR-inhibited autophagy in the heart after myocardial infarction. Further analysis revealed that the PI3K/Akt signaling pathway was significantly enriched and validated by western blot. This analysis suggested that the ratios of p- PI3K/PI3K, p-Akt/Akt, and p-mTOR/mTOR were significantly increased. CONCLUSION LIR may attenuate myocardial infarction-induced fibrosis in rats by inhibiting excessive myocardial autophagy, with the potential mechanism involving the activation of the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Cardiology, Tianjin Nankai Hospital, Tianjin, 300100, China
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Xuanming Liu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xin Yu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuzhen Zhuo
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Dihua Li
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Lei Yang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Yanmin Lu
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, 300100, China
| |
Collapse
|
32
|
Shi J, Yang C, Zhang J, Zhao K, Li P, Kong C, Wu X, Sun H, Zheng R, Sun W, Chen L, Kong X. NAT10 Is Involved in Cardiac Remodeling Through ac4C-Mediated Transcriptomic Regulation. Circ Res 2023; 133:989-1002. [PMID: 37955115 DOI: 10.1161/circresaha.122.322244] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 10/30/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND Heart failure, characterized by cardiac remodeling, is associated with abnormal epigenetic processes and aberrant gene expression. Here, we aimed to elucidate the effects and mechanisms of NAT10 (N-acetyltransferase 10)-mediated N4-acetylcytidine (ac4C) acetylation during cardiac remodeling. METHODS NAT10 and ac4C expression were detected in both human and mouse subjects with cardiac remodeling through multiple assays. Subsequently, acetylated RNA immunoprecipitation and sequencing, thiol-linked alkylation for the metabolic sequencing of RNA (SLAM-seq), and ribosome sequencing (Ribo-seq) were employed to elucidate the role of ac4C-modified posttranscriptional regulation in cardiac remodeling. Additionally, functional experiments involving the overexpression or knockdown of NAT10 were conducted in mice models challenged with Ang II (angiotensin II) and transverse aortic constriction. RESULTS NAT10 expression and RNA ac4C levels were increased in in vitro and in vivo cardiac remodeling models, as well as in patients with cardiac hypertrophy. Silencing and inhibiting NAT10 attenuated Ang II-induced cardiomyocyte hypertrophy and cardiofibroblast activation. Next-generation sequencing revealed ac4C changes in both mice and humans with cardiac hypertrophy were associated with changes in global mRNA abundance, stability, and translation efficiency. Mechanistically, NAT10 could enhance the stability and translation efficiency of CD47 and ROCK2 transcripts by upregulating their mRNA ac4C modification, thereby resulting in an increase in their protein expression during cardiac remodeling. Furthermore, the administration of Remodelin, a NAT10 inhibitor, has been shown to prevent cardiac functional impairments in mice subjected to transverse aortic constriction by suppressing cardiac fibrosis, hypertrophy, and inflammatory responses, while also regulating the expression levels of CD47 and ROCK2 (Rho associated coiled-coil containing protein kinase 2). CONCLUSIONS Therefore, our data suggest that modulating epitranscriptomic processes, such as ac4C acetylation through NAT10, may be a promising therapeutic target against cardiac remodeling.
Collapse
Affiliation(s)
- Jing Shi
- Department of Cardiology (J.S., K.Z., J.Z., P.L., X.W., W.S., X.K.), The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
| | - Chuanxi Yang
- Department of Cardiology, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China (C.Y.)
| | - Jing Zhang
- Department of Cardiology (J.S., K.Z., J.Z., P.L., X.W., W.S., X.K.), The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
| | - Kun Zhao
- Department of Cardiology (J.S., K.Z., J.Z., P.L., X.W., W.S., X.K.), The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
| | - Peng Li
- Department of Cardiology (J.S., K.Z., J.Z., P.L., X.W., W.S., X.K.), The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
| | - Chuiyu Kong
- Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Jiangsu, China (C.K.)
| | - Xiaoguang Wu
- Department of Cardiology (J.S., K.Z., J.Z., P.L., X.W., W.S., X.K.), The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
| | - Haoliang Sun
- Department of Cardiovascular Surgery (H.S., R.Z.), The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
| | - Rui Zheng
- Department of Cardiovascular Surgery (H.S., R.Z.), The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
| | - Wei Sun
- Department of Cardiology (J.S., K.Z., J.Z., P.L., X.W., W.S., X.K.), The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
| | - Lianmin Chen
- Changzhou Medical Center of the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University and Department of Cardiology of the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China (L.C.)
| | - Xiangqing Kong
- Department of Cardiology (J.S., K.Z., J.Z., P.L., X.W., W.S., X.K.), The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
- Cardiovascular Research Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China (X.K.)
| |
Collapse
|
33
|
Grujić-Milanović J, Rajković J, Milanović S, Jaćević V, Miloradović Z, Nežić L, Novaković R. Natural Substances vs. Approved Drugs in the Treatment of Main Cardiovascular Disorders-Is There a Breakthrough? Antioxidants (Basel) 2023; 12:2088. [PMID: 38136208 PMCID: PMC10740850 DOI: 10.3390/antiox12122088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/29/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
Cardiovascular diseases (CVDs) are a group of diseases with a very high rate of morbidity and mortality. The clinical presentation of CVDs can vary from asymptomatic to classic symptoms such as chest pain in patients with myocardial infarction. Current therapeutics for CVDs mainly target disease symptoms. The most common CVDs are coronary artery disease, acute myocardial infarction, atrial fibrillation, chronic heart failure, arterial hypertension, and valvular heart disease. In their treatment, conventional therapies and pharmacological therapies are used. However, the use of herbal medicines in the therapy of these diseases has also been reported in the literature, resulting in a need for critical evaluation of advances related to their use. Therefore, we carried out a narrative review of pharmacological and herbal therapeutic effects reported for these diseases. Data for this comprehensive review were obtained from electronic databases such as MedLine, PubMed, Web of Science, Scopus, and Google Scholar. Conventional therapy requires an individual approach to the patients, as when patients do not respond well, this often causes allergic effects or various other unwanted effects. Nowadays, medicinal plants as therapeutics are frequently used in different parts of the world. Preclinical/clinical pharmacology studies have confirmed that some bioactive compounds may have beneficial therapeutic effects in some common CVDs. The natural products analyzed in this review are promising phytochemicals for adjuvant and complementary drug candidates in CVDs pharmacotherapy, and some of them have already been approved by the FDA. There are insufficient clinical studies to compare the effectiveness of natural products compared to approved therapeutics for the treatment of CVDs. Further long-term studies are needed to accelerate the potential of using natural products for these diseases. Despite this undoubted beneficence on CVDs, there are no strong breakthroughs supporting the implementation of natural products in clinical practice. Nevertheless, they are promising agents in the supplementation and co-therapy of CVDs.
Collapse
Affiliation(s)
- Jelica Grujić-Milanović
- Institute for Medical Research, National Institute of the Republic of Serbia, Department of Cardiovascular Research, University of Belgrade, 11 000 Belgrade, Serbia;
| | - Jovana Rajković
- Institute for Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11 000 Belgrade, Serbia
| | - Sladjan Milanović
- Institute for Medical Research, National Institute of the Republic of Serbia, Department for Biomechanics, Biomedical Engineering and Physics of Complex Systems, University of Belgrade, 11 000 Belgrade, Serbia;
| | - Vesna Jaćević
- Department for Experimental Toxicology and Pharmacology, National Poison Control Centre, Military Medical Academy, 11 000 Belgrade, Serbia;
- Medical Faculty of the Military Medical Academy, University of Defense, 11 000 Belgrade, Serbia
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, 500 002 Hradec Kralove, Czech Republic
| | - Zoran Miloradović
- Institute for Medical Research, National Institute of the Republic of Serbia, Department of Cardiovascular Research, University of Belgrade, 11 000 Belgrade, Serbia;
| | - Lana Nežić
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, Save Mrkalja 14, 78000 Banja Luka, Bosnia and Herzegovina;
| | - Radmila Novaković
- Institute of Molecular Genetics and Genetic Engineering, Center for Genome Sequencing and Bioinformatics, University of Belgrade, 11 000 Belgrade, Serbia;
| |
Collapse
|
34
|
Gebauer AM, Pfaller MR, Braeu FA, Cyron CJ, Wall WA. A homogenized constrained mixture model of cardiac growth and remodeling: analyzing mechanobiological stability and reversal. Biomech Model Mechanobiol 2023; 22:1983-2002. [PMID: 37482576 PMCID: PMC10613155 DOI: 10.1007/s10237-023-01747-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 07/06/2023] [Indexed: 07/25/2023]
Abstract
Cardiac growth and remodeling (G&R) patterns change ventricular size, shape, and function both globally and locally. Biomechanical, neurohormonal, and genetic stimuli drive these patterns through changes in myocyte dimension and fibrosis. We propose a novel microstructure-motivated model that predicts organ-scale G&R in the heart based on the homogenized constrained mixture theory. Previous models, based on the kinematic growth theory, reproduced consequences of G&R in bulk myocardial tissue by prescribing the direction and extent of growth but neglected underlying cellular mechanisms. In our model, the direction and extent of G&R emerge naturally from intra- and extracellular turnover processes in myocardial tissue constituents and their preferred homeostatic stretch state. We additionally propose a method to obtain a mechanobiologically equilibrated reference configuration. We test our model on an idealized 3D left ventricular geometry and demonstrate that our model aims to maintain tensional homeostasis in hypertension conditions. In a stability map, we identify regions of stable and unstable G&R from an identical parameter set with varying systolic pressures and growth factors. Furthermore, we show the extent of G&R reversal after returning the systolic pressure to baseline following stage 1 and 2 hypertension. A realistic model of organ-scale cardiac G&R has the potential to identify patients at risk of heart failure, enable personalized cardiac therapies, and facilitate the optimal design of medical devices.
Collapse
Affiliation(s)
- Amadeus M Gebauer
- Institute for Computational Mechanics, Technical University of Munich, 85748, Garching, Germany.
| | - Martin R Pfaller
- Pediatric Cardiology, Stanford Maternal & Child Health Research Institute, and Institute for Computational and Mathematical Engineering, Stanford University, Stanford, USA
| | - Fabian A Braeu
- Ophthalmic Engineering & Innovation Laboratory, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Christian J Cyron
- Institute of Continuum and Material Mechanics, Hamburg University of Technology, 21073, Hamburg, Germany
- Institute of Material Systems Modeling, Helmholtz-Zentrum Hereon, 21502, Geesthacht, Germany
| | - Wolfgang A Wall
- Institute for Computational Mechanics, Technical University of Munich, 85748, Garching, Germany
| |
Collapse
|
35
|
Wu S, Zhang J, Peng C, Ma Y, Tian X. SIRT6 mediated histone H3K9ac deacetylation involves myocardial remodelling through regulating myocardial energy metabolism in TAC mice. J Cell Mol Med 2023; 27:3451-3464. [PMID: 37603612 PMCID: PMC10660608 DOI: 10.1111/jcmm.17915] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 08/06/2023] [Accepted: 08/09/2023] [Indexed: 08/23/2023] Open
Abstract
Pathological myocardial remodelling is the initial factor of chronic heart failure (CHF) and is induced by multiple factors. We previously demonstrated that histone acetylation is involved in CHF in transverse aortic constriction (TAC) mice, a model for pressure overload-induced heart failure. In this study, we investigated whether the histone deacetylase Sirtuin 6 (SIRT6), which mediates deacetylation of histone 3 acetylated at lysine 9 (H3K9ac), is involved pathological myocardial remodelling by regulating myocardial energy metabolism and explored the underlying mechanisms. We generated a TAC mouse model by partial thoracic aortic banding. TAC mice were injected with the SIRT6 agonist MDL-800 at a dose of 65 mg/kg for 8 weeks. At 4, 8 and 12 weeks after TAC, the level of H3K9ac increased gradually, while the expression of SIRT6 and vascular endothelial growth factor A (VEGFA) decreased gradually. MDL-800 reversed the effects of SIRT6 on H3K9ac in TAC mice and promoted the expression of VEGFA in the hearts of TAC mice. MDL-800 also attenuated mitochondria damage and improved mitochondrial respiratory function through upregulating SIRT6 in the hearts of TAC mice. These results revealed a novel mechanism in which SIRT6-mediated H3K9ac level is involved pathological myocardial remodelling in TAC mice through regulating myocardial energy metabolism. These findings may assist in the development of novel methods for preventing and treating pathological myocardial remodelling.
Collapse
Affiliation(s)
- Shuqi Wu
- Department of Pediatrics, Guizhou Children's HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Jiaojiao Zhang
- Department of Pediatrics, Guizhou Children's HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Chang Peng
- Department of Pediatrics, Guizhou Children's HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Yixiang Ma
- Department of Pediatrics, Guizhou Children's HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Xiaochun Tian
- Department of Pediatrics, Guizhou Children's HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| |
Collapse
|
36
|
Yao W, Pei Z, Zhang X. NAD +: A key metabolic regulator with great therapeutic potential for myocardial infarction via Sirtuins family. Heliyon 2023; 9:e21890. [PMID: 38027748 PMCID: PMC10663897 DOI: 10.1016/j.heliyon.2023.e21890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/19/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Myocardial infarction (MI) is one of the complex phenotypes of coronary artery disease, which results from the interaction of multiple genetic and environmental factors. Nicotinamide Adenine Dinucleotide (NAD+) is an important cofactor regulating metabolic homeostasis and a rate-limiting substrate for sirtuin (SIRT) deacetylase. Numerous NAD+ studies have shown that it can be used as an anti-MI treatment. However, there have been few systematic reviews of the overall role of NAD+ in treating MI. MI, which has long been a global health problem, still lacks effective treatment till now, and the discovery of NAD+ provides a new perspective on its adjuvant treatment. This review summarizes the role of NAD+ signaling in SIRTs in alleviating MI.
Collapse
Affiliation(s)
- Wei Yao
- Department of Internal Medicine, Affiliated Zhong Shan Hospital of Dalian University, Dalian, 116001, China
| | - Zuowei Pei
- Department of Cardiology, Central Hospital of Dalian University of Technology, Dalian, 116089, China
- Department of Central Laboratory, Central Hospital of Dalian University of Technology, Dalian, 116033, China
- Faculty of Medicine, Dalian University of Technology, Dalian, 116024, China
| | - Xiaoqing Zhang
- Department of Infection, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China
| |
Collapse
|
37
|
Yuan X, Liu K, Dong P, Han H. Protective effect and mechanism of different proportions of " Danggui-Kushen" herb pair on ischemic heart disease. Heliyon 2023; 9:e22150. [PMID: 38034717 PMCID: PMC10685368 DOI: 10.1016/j.heliyon.2023.e22150] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 11/03/2023] [Accepted: 11/05/2023] [Indexed: 12/02/2023] Open
Abstract
This study aims to investigate the protective effect and mechanism of "Danggui-Kushen" herb pair (DKHP) on ischemic heart disease (IHD). The rat model of myocardial reperfusion injury (MIRI) was established by ligation of the left anterior descending coronary artery. Rats were randomly divided into seven groups and administered orally for 7 days: control group, IHD group, DKHP1:1 group, DKHP1:2 group, DKHP2:1 group, DKHP1:3 group, DKHP3:1 group, the dosage was 2.7 g/kg. Measure electrocardiogram (ECG), myocardial infarction and injury assessment, Hematoxylin and eosin (HE) staining to evaluate myocardial injury and the protective effect of DKHP. Lactate dehydrogenase (LDH), Reactive oxygen species (ROS), IL-1β and IL-6 kit detection, immunohistochemical analysis, establishment of H9c2 cardiomyocyte hypoxia (Hypoxia) model, DKHP pretreatment for 3 h, MTT method to detect cell survival rate, cell immunofluorescence to observe NF- The expression of TLR-4, NF-κB, p-NF-κB, IKβα, p-IKβα, HIF-1α, VEGF and other genes and proteins were detected by κB nuclear translocation, mitochondrial membrane potential measurement, Western blot and Polymerase Chain Reaction (PCR). Compared with the model group, DKHP can reduce the size of myocardial infarction, reduce the levels of factors such as LDH, ROS, IL-1β and IL-6, and improve the cell survival rate; Compared with the model group, DKHP can inhibit the nuclear transfer of NF-κB and reduce mitochondrial damage; the results of immunohistochemical analysis, PCR and Western blot showed that compared with the model group, DKHP can reduce TLR-4, p-NF-κB, Expression levels of p-IKβα, HIF-1α, VEGF and other proteins. Reveal that DKHP may play a protective role in ischemic heart disease by reducing inflammation and oxidative stress damage. DKHP may have protective effect on ischemic heart disease, and its mechanism may be through reducing inflammatory response and oxidative stress damage to achieve this protective effect.
Collapse
Affiliation(s)
- Xu Yuan
- College of Medicine, Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, China
| | - Kemeng Liu
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Peiliang Dong
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Hua Han
- College of Medicine, Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, China
| |
Collapse
|
38
|
Wang Y, Li Q, Zhao J, Chen J, Wu D, Zheng Y, Wu J, Liu J, Lu J, Zhang J, Wu Z. Mechanically induced pyroptosis enhances cardiosphere oxidative stress resistance and metabolism for myocardial infarction therapy. Nat Commun 2023; 14:6148. [PMID: 37783697 PMCID: PMC10545739 DOI: 10.1038/s41467-023-41700-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 09/14/2023] [Indexed: 10/04/2023] Open
Abstract
Current approaches in myocardial infarction treatment are limited by low cellular oxidative stress resistance, reducing the long-term survival of therapeutic cells. Here we develop a liquid-crystal substrate with unique surface properties and mechanical responsiveness to produce size-controllable cardiospheres that undergo pyroptosis to improve cellular bioactivities and resistance to oxidative stress. We perform RNA sequencing and study cell metabolism to reveal increased metabolic levels and improved mitochondrial function in the preconditioned cardiospheres. We test therapeutic outcomes in a rat model of myocardial infarction to show that cardiospheres improve long-term cardiac function, promote angiogenesis and reduce cardiac remodeling during the 3-month observation. Overall, this study presents a promising and effective system for preparing a large quantity of functional cardiospheres, showcasing potential for clinical application.
Collapse
Affiliation(s)
- Yingwei Wang
- Key Laboratory for Regenerative Medicine, Ministry of Education, Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Qi Li
- Key Laboratory for Regenerative Medicine, Ministry of Education, Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Jupeng Zhao
- Key Laboratory for Regenerative Medicine, Ministry of Education, Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Jiamin Chen
- Key Laboratory for Regenerative Medicine, Ministry of Education, Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Dongxue Wu
- Department of Cardiology, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Youling Zheng
- Key Laboratory for Regenerative Medicine, Ministry of Education, Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Jiaxin Wu
- Key Laboratory for Regenerative Medicine, Ministry of Education, Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Jie Liu
- Key Laboratory for Regenerative Medicine, Ministry of Education, Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Jianlong Lu
- Key Laboratory for Regenerative Medicine, Ministry of Education, Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Jianhua Zhang
- Department of Cardiology, First Affiliated Hospital of Jinan University, Guangzhou, China.
| | - Zheng Wu
- Key Laboratory for Regenerative Medicine, Ministry of Education, Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China.
| |
Collapse
|
39
|
Pang X, Guan Q, Lin X, Chang N. Knockdown of HDAC6 alleviates ventricular remodeling in experimental dilated cardiomyopathy via inhibition of NLRP3 inflammasome activation and promotion of cardiomyocyte autophagy. Cell Biol Toxicol 2023; 39:2365-2379. [PMID: 35764897 DOI: 10.1007/s10565-022-09727-z] [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: 12/13/2021] [Accepted: 05/10/2022] [Indexed: 11/02/2022]
Abstract
Histone deacetylases (HDACs) has been implicated in cardiac diseases, while the role of HDAC6 in dilated cardiomyopathy (DCM) remains obscure. The in silico analyses predicted potential association of HDAC6 with autophagy-related genes and DCM. Thus, we evaluated the functional relevance of HDAC6 in DCM in vivo and in vitro. We developed a rat model in vivo and a cell model in vitro by doxorubicin (DOX) induction to simulate DCM. HDAC6 expression was determined in myocardial tissues of DCM rats. DCM rats exhibited elevated HDAC6 mRNA and protein expression as compared to sham-operated rats. We knocked HDAC6 down and/or overexpressed NLRP3 in vivo and in vitro to characterize their roles in cardiomyocyte autophagy. It was established that shRNA-mediated HDAC6 silencing augmented cardiomyocyte autophagy and suppressed NLRP3 inflammasome activation, thus ameliorating cardiac injury in myocardial tissues of DCM rats. Besides, in DOX-injured cardiomyocytes, HDAC6 silencing also diminished NLRP3 inflammasome activation and cell apoptosis but enhanced cell autophagy, whereas ectopic NLRP3 expression negated the effects of HDAC6 silencing. Since HDAC6 knockdown correlates with enhanced cardiomyocyte autophagy and suppressed NLRP3 inflammasome activation through an interplay with NLRP3, it is expected to be a potential biomarker and therapeutic target for DCM. 1. HDAC6 was up-regulated in DCM rats. 2. HDAC6 knockdown promoted cardiomyocyte autophagy to relieve cardiac dysfunction. 3. HDAC6 knockdown inhibited NLRP3 inflammasome and promoted cardiomyocyte autophagy. 4. Silencing HDAC6 promoted autophagy and repressed apoptosis in cardiomyocytes. 5. This study provides novel therapeutic targets for DCM.
Collapse
Affiliation(s)
- Xuefeng Pang
- Department of Cardiovascular Medicine, the First Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Qigang Guan
- Department of Cardiovascular Medicine, the First Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Xue Lin
- Department of Cardiovascular Medicine, Peking Union Medical College Hospital, Beijing, 100730, People's Republic of China
| | - Ning Chang
- Department of Digestive Diseases, the First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China.
| |
Collapse
|
40
|
Jiang H, Fang T, Cheng Z. Mechanism of heart failure after myocardial infarction. J Int Med Res 2023; 51:3000605231202573. [PMID: 37818767 PMCID: PMC10566288 DOI: 10.1177/03000605231202573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 08/14/2023] [Indexed: 10/13/2023] Open
Abstract
Despite the widespread use of early revascularization and drugs to regulate the neuroendocrine system, the impact of such measures on alleviating the development of heart failure (HF) after myocardial infarction (MI) remains limited. Therefore, it is important to discuss the development of new therapeutic strategies to prevent or reverse HF after MI. This requires a better understanding of the potential mechanisms involved. HF after MI is the result of complex pathophysiological processes, with adverse ventricular remodeling playing a major role. Adverse ventricular remodeling refers to the heart's adaptation in terms of changes in ventricular size, shape, and function under the influence of various regulatory factors, including the mechanical, neurohormonal, and cardiac inflammatory immune environments; ischemia/reperfusion injury; energy metabolism; and genetic correlation factors. Additionally, unique right ventricular dysfunction can occur secondary to ischemic shock in the surviving myocardium. HF after MI may also be influenced by other factors. This review summarizes the main pathophysiological mechanisms of HF after MI and highlights sex-related differences in the prognosis of patients with acute MI. These findings provide new insights for guiding the development of targeted treatments to delay the progression of HF after MI and offering incremental benefits to existing therapies.
Collapse
Affiliation(s)
- Huaiyu Jiang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Fang
- Department of Cardiology, The Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Zeyi Cheng
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
41
|
Ye Q, Chen W, Fu H, Ding Y, Jing Y, Shen J, Yuan Z, Zha K. Targeting Autophagy in Atrial Fibrillation. Rev Cardiovasc Med 2023; 24:288. [PMID: 39077569 PMCID: PMC11273128 DOI: 10.31083/j.rcm2410288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 05/04/2023] [Accepted: 05/19/2023] [Indexed: 07/31/2024] Open
Abstract
Atrial fibrillation (AF) is the most common type of arrhythmia in clinical practice, and its incidence is positively correlated with risk factors that include advanced age, hypertension, diabetes, and heart failure. Although our understanding of the mechanisms that govern the occurrence and persistence of AF has been increasing rapidly, the exact mechanism of AF is still not fully understood. Autophagy is an evolutionarily highly conserved and specific physiological process in cells that has been suggested as a potential therapeutic target for several cardiovascular diseases including the pathophysiology of AF. The present article provides an updated review of the fast-progressing field of research surrounding autophagy in AF, and how regulating autophagy might be a therapeutic target to reduce the incidence of AF.
Collapse
Affiliation(s)
- Qiang Ye
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Wen Chen
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Hengsong Fu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Yanling Ding
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Yuling Jing
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Jingsong Shen
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Ziyang Yuan
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Kelan Zha
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| |
Collapse
|
42
|
Magodoro IM, Guerrero-Chalela CE, Claggett B, Jermy S, Samuels P, Zar H, Myer L, Danaei G, Jao J, Ntusi NAB, Siedner MJ, Ntsekhe M. Left ventricular remodeling and its correlates among adolescents with perinatally acquired HIV in South Africa. Int J Cardiol 2023; 387:131121. [PMID: 37336247 PMCID: PMC10529415 DOI: 10.1016/j.ijcard.2023.131121] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 06/21/2023]
Abstract
BACKGROUND Left ventricular (LV) remodeling and its transitions from compensatory adaptations to LV dysfunction have not been examined in adolescents with perinatally acquired HIV infection (PHIV). We used cardiovascular magnetic resonance (CMR) in a cross-sectional study to characterize PHIV-related progressive LV remodeling in adolescents in South Africa. METHODS Adolescents with PHIV on antiretroviral treatment and their HIV uninfected peers completed 3 T CMR examination. We defined LV remodeling by LV mass/volume (M/V) ratio, modelling progressive LV remodeling as increasing M/V ratio. Linear regression models were applied to estimate the correlates of progressive LV remodeling. RESULTS Overall, 71 adolescents with PHIV [mean age: 15.2 years; 54% male] and 36 HIV uninfected [15.1 years; 42% male] peers were enrolled. Adolescents with PHIV had lower mean LV M/V ratio (0.68 vs. 0.75 g/mL; p = 0.004) than HIV uninfected peers, without LV hypertrophy in either group. Among adolescents with PHIV, increasing M/V ratio was accompanied by increasing interstitial volume [adjusted mean change (AMC) per 0.1 g/mL M/V ratio: 1.75 mL, p < 0.001] with no change in global circumferential strain (GCS) [AMC per 0.1 g/mL M/V ratio: -0.21%, p = 0.48]. However, in HIV uninfected individuals, increasing M/V ratio was accompanied by increasing peak GCS [AMC per 0.1 g/mL M/V ratio: -1.25%, p = 0.039] with no change in interstitial volume (AMC per 0.1 g/mL M/V ratio: 1.16 mL, p = 0.32]. CONCLUSIONS Successfully treated PHIV is associated with less severe LV remodeling in adolescence when compared to HIV uninfected controls. LV remodeling in PHIV is associated with disproportionate expansion of the non-contractile interstitium not accompanied by improved GCS.
Collapse
Affiliation(s)
- Itai M Magodoro
- Division of Cardiology, University of Cape Town, Cape Town, South Africa; Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
| | | | - Brian Claggett
- Cardiology Division, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Stephen Jermy
- Cape Universities Body Imaging Centre, University of Cape Town, Cape Town, South Africa
| | - Petronella Samuels
- Cape Universities Body Imaging Centre, University of Cape Town, Cape Town, South Africa
| | - Heather Zar
- Department of Pediatrics and Child Health, and SA-MRC Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa; Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Landon Myer
- School of Public Health and Family Medicine, University of Cape Town, South Africa
| | - Goodarz Danaei
- Department of Global Health and Population, Harvard School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
| | - Jennifer Jao
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Department of Internal Medicine, Division of Adult Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ntobeko A B Ntusi
- Cape Universities Body Imaging Centre, University of Cape Town, Cape Town, South Africa; Department of Medicine, University of Cape Town, Cape Town, South Africa; Cape Heart Institute, University of Cape Town, Cape Town, South Africa; South African Medical Research Council Extramural Unit on Noncommunicable and Infectious Diseases, Cape Town, South Africa
| | - Mark J Siedner
- Harvard Medical School, Boston, MA, USA; Massachusetts General Hospital, Boston, MA, USA; Africa Health Research Institute, KwaZulu-Natal, South Africa
| | - Mpiko Ntsekhe
- Division of Cardiology, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
43
|
Narayan SI, Terre GV, Amin R, Shanghavi KV, Chandrashekar G, Ghouse F, Ahmad BA, S GN, Satram C, Majid HA, Bayoro DK. The Pathophysiology and New Advancements in the Pharmacologic and Exercise-Based Management of Heart Failure With Reduced Ejection Fraction: A Narrative Review. Cureus 2023; 15:e45719. [PMID: 37868488 PMCID: PMC10590213 DOI: 10.7759/cureus.45719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2023] [Indexed: 10/24/2023] Open
Abstract
Heart failure with reduced ejection fraction (HFrEF) is a clinical syndrome whose management has significantly evolved based on the pathophysiology and disease process. It is widely prevalent, has a relatively high mortality rate, and is comparatively more common in men than women. In HFrEF, the series of maladaptive processes that occur lead to an inability of the muscle of the left ventricle to pump blood efficiently and effectively, causing cardiac dysfunction. The neurohormonal and hemodynamic adaptations play a significant role in the advancement of the disease and are critical to guiding the treatment and management of HFrEF. The first-line therapy, which includes loop diuretics, β-blockers, angiotensin-converting enzyme inhibitors/angiotensin II receptor blockers, hydralazine/isosorbide-dinitrate, and mineralocorticoid receptor antagonists (MRAs), has been proven to provide symptomatic relief and decrease mortality and complications. The newly recommended drugs for guideline-based therapy, angiotensin receptor/neprilysin inhibitor (ARNI), sodium-glucose cotransporter 2 inhibitors, soluble guanylate cyclase, and myosin activators and modulators have also been shown to improve cardiac function, reverse cardiac remodeling, and reduce mortality rates. Recent studies have demonstrated that exercise-based therapy has resulted in an improved quality of life, exercise capacity, cardiac function, and decreased hospital readmission rates, but it has not had a considerable reduction in mortality rates. Combining multiple therapies alongside holistic advances such as exercise therapy may provide synergistic benefits, ultimately leading to improved outcomes for patients with HFrEF. Although first-line treatment, novel pharmacologic management, and exercise-based therapy have been shown to improve prognosis, the existing literature suggests a need for further studies evaluating the long-term effects of MRA and ARNI.
Collapse
Affiliation(s)
| | - Giselle V Terre
- Department of Medicine, Universidad Iberoamericana (UNIBE), Santo Domingo, DOM
| | - Rutvi Amin
- Department of Medicine, Surat Municipal Institute of Medical Education and Research, Surat, IND
| | - Keshvi V Shanghavi
- Department of Medicine, Lokmanya Tilak Municipal Medical College and Sion Hospital, Mumbai, IND
| | | | - Farhana Ghouse
- Department of Medicine, Saint James School of Medicine, St. Vincent, VCT
| | - Binish A Ahmad
- Department of Medicine, King Edward Medical University, Lahore, PAK
| | - Gowri N S
- Department of Medicine, Taras Shevchenko National University of Kyiv, Kyiv, UKR
| | - Christena Satram
- Department of Medicine, Lincoln American University, Georgetown, GUY
| | - Hamna A Majid
- Department of Medicine, Dow University of Health Sciences, Dow International Medical College, Karachi, PAK
| | - Danielle K Bayoro
- Department of Medicine, Medical University of the Americas, Nevis, KNA
| |
Collapse
|
44
|
Hou H, Chen Y, Feng X, Xu G, Yan M. Tripartite motif‑containing 14 may aggravate cardiac hypertrophy via the AKT signalling pathway in neonatal rat cardiomyocytes and transgenic mice. Mol Med Rep 2023; 28:173. [PMID: 37503784 PMCID: PMC10433706 DOI: 10.3892/mmr.2023.13060] [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: 12/15/2022] [Accepted: 05/25/2023] [Indexed: 07/29/2023] Open
Abstract
Tripartite motif‑containing 14 (TRIM14) is an E3 ubiquitin ligase that primarily participates in the natural immune response and in tumour development via ubiquitination. However, the role of TRIM14 in cardiac hypertrophy is not currently clear. The present study examined the role of TRIM14 in cardiac hypertrophy and its potential molecular mechanism. TRIM14 was overexpressed in neonatal rat cardiomyocytes using adenovirus and cardiomyocyte hypertrophy was induced using phenylephrine (PE). Cardiomyocyte hypertrophy was assessed by measuring cardiomyocyte surface area and markers of hypertrophy. In addition, TRIM14‑transgenic (TRIM14‑TG) mice were created and cardiac hypertrophy was induced using transverse aortic constriction (TAC). Cardiac function, heart weight‑to‑body weight ratio (HW/BW), cardiomyocyte cross‑sectional area, cardiac fibrosis and hypertrophic markers were further examined. The expression of AKT signalling pathway‑related proteins was detected. TRIM14 overexpression in cardiomyocytes promoted PE‑induced increases in cardiomyocyte surface area and hypertrophic markers. TRIM14‑TG mice developed worse cardiac function, greater HW/BW, cross‑sectional area and cardiac fibrosis, and higher levels of hypertrophic markers in response to TAC. TRIM14 overexpression also increased the phosphorylation levels of AKT, GSK‑3β, mTOR and p70S6K in vivo and in vitro. To the best our knowledge, the present study was the first to reveal that overexpression of TRIM14 aggravated cardiac hypertrophy in vivo and in vitro, which may be related to activation of the AKT signalling pathway.
Collapse
Affiliation(s)
- Hongwei Hou
- Department of Cardiology, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, P.R. China
- Department of Cardiology, Ezhou Central Hospital, Ezhou, Hubei 436000, P.R. China
| | - Yan Chen
- Department of Cardiology, Ezhou Central Hospital, Ezhou, Hubei 436000, P.R. China
| | - Xiuyuan Feng
- Department of Cardiology, Ezhou Central Hospital, Ezhou, Hubei 436000, P.R. China
| | - Guang Xu
- Department of Cardiology, Ezhou Central Hospital, Ezhou, Hubei 436000, P.R. China
| | - Min Yan
- Department of General Practice, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, P.R. China
| |
Collapse
|
45
|
Dhalla NS, Bhullar SK, Adameova A, Mota KO, de Vasconcelos CML. Status of β 1-Adrenoceptor Signal Transduction System in Cardiac Hypertrophy and Heart Failure. Rev Cardiovasc Med 2023; 24:264. [PMID: 39076390 PMCID: PMC11270071 DOI: 10.31083/j.rcm2409264] [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: 03/16/2023] [Revised: 05/30/2023] [Accepted: 06/15/2023] [Indexed: 07/31/2024] Open
Abstract
Although β 1-adrenoceptor ( β 1-AR) signal transduction, which maintains cardiac function, is downregulated in failing hearts, the mechanisms for such a defect in heart failure are not fully understood. Since cardiac hypertrophy is invariably associated with heart failure, it is possible that the loss of β 1-AR mechanisms in failing heart occurs due to hypertrophic process. In this regard, we have reviewed the information from a rat model of adaptive cardiac hypertrophy and maladaptive hypertrophy at 4 and 24 weeks after inducing pressure overload as well as adaptive cardiac hypertrophy and heart failure at 4 and 24 weeks after inducing volume overload, respectively. Varying degrees of alterations in β 1-AR density as well as isoproterenol-induced increases in cardiac function, intracellular Ca 2 + -concentration in cardiomyocytes and adenylyl cyclase activity in crude membranes have been reported under these hypertrophic conditions. Adaptive hypertrophy at 4 weeks of pressure or volume overload showed unaltered or augmented increases in the activities of different components of β 1-AR signaling. On the other hand, maladaptive hypertrophy due to pressure overload and heart failure due to volume overload at 24 weeks revealed depressions in the activities of β 1-AR signal transduction pathway. These observations provide evidence that β 1-AR signal system is either unaltered or upregulated in adaptive cardiac hypertrophy and downregulated in maladaptive cardiac hypertrophy or heart failure. Furthermore, the information presented in this article supports the concept that downregulation of β 1-AR mechanisms in heart failure or maladaptive cardiac hypertrophy is not due to hypertrophic process per se. It is suggested that a complex mechanism involving the autonomic imbalance may be of a critical importance in determining differential alterations in non-failing and failing hearts.
Collapse
Affiliation(s)
- Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, and Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Sukhwinder K. Bhullar
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, and Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Adriana Adameova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University and Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 811 03 Bratislava, Slovakia
| | - Karina Oliveira Mota
- Heart Biophysics Laboratory, Department of Physiology, Center for Biological and Health Sciences, Federal University of Sergipe, 73330 Sergipe, Brazil
| | - Carla Maria Lins de Vasconcelos
- Heart Biophysics Laboratory, Department of Physiology, Center for Biological and Health Sciences, Federal University of Sergipe, 73330 Sergipe, Brazil
| |
Collapse
|
46
|
Xanthopoulos A, Katsiadas N, Skoularigkis S, Magouliotis DE, Skopeliti N, Patsilinakos S, Briasoulis A, Triposkiadis F, Skoularigis J. Association between Dapagliflozin, Cardiac Biomarkers and Cardiac Remodeling in Patients with Diabetes Mellitus and Heart Failure. Life (Basel) 2023; 13:1778. [PMID: 37629635 PMCID: PMC10455594 DOI: 10.3390/life13081778] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/17/2023] [Accepted: 08/19/2023] [Indexed: 08/27/2023] Open
Abstract
Sodium-glucose cotransporter-2 inhibitors (SGLT2is) are a relatively new class of antidiabetic drugs that have shown favorable effects in heart failure (HF) patients, irrespective of the left ventricular ejection fraction (LVEF). Recent studies have demonstrated the beneficial effects of empagliflozin on cardiac function and structure; however, less is known about dapagliflozin. The purpose of the current work was to investigate the association between the use of dapagliflozin and cardiac biomarkers as well as the cardiac structure in a cohort of patients with HF and diabetes mellitus (DM). The present work was an observational study that included 118 patients (dapagliflozin group n = 60; control group n = 58) with HF and DM. The inclusion criteria included: age > 18 years, a history of DM and HF, regardless of LVEF, and hospitalization for HF exacerbation within the previous 6 months. The exclusion criteria were previous treatment with SGLT2i or glucagon-like peptide-1 receptor agonists, a GFR< 30 and life expectancy < 1 year. The evaluation of patients (at baseline, 6 and 12 months) included a clinical assessment, laboratory blood tests and echocardiography. The Mann-Whitney test was used for the comparison of continuous variables between the two groups, while Friedman's analysis of variance for repeated measures was used for the comparison of continuous variables. Troponin (p < 0.001) and brain natriuretic peptide (BNP) (p < 0.001) decreased significantly throughout the follow-up period in the dapagliflozin group, but not in the control group (p > 0.05 for both). The LV end-diastolic volume index (p < 0.001 for both groups) and LV end-systolic volume index (p < 0.001 for both groups) decreased significantly in the dapagliflozin and the control group, respectively. The LVEF increased significantly (p < 0.001) only in the dapagliflozin group, whereas the global longitudinal strain (GLS) improved in the dapagliflozin group (p < 0.001) and was impaired in the control group (p = 0.021). The left atrial volume index decreased in the dapagliflozin group (p < 0.001) but remained unchanged in the control group (p = 0.114). Lastly, the left ventricular mass index increased significantly both in the dapagliflozin (p = 0.003) and control group (p = 0.001). Dapagliflozin, an SGLT2i, was associated with a reduction in cardiac biomarkers and with reverse cardiac remodeling in patients with HF and DM.
Collapse
Affiliation(s)
- Andrew Xanthopoulos
- Department of Cardiology, University Hospital of Larissa, 41110 Larissa, Greece; (S.S.); (N.S.); (F.T.)
| | - Nikolaos Katsiadas
- Department of Cardiology, Konstantopoulio General Hospital, 14233 Nea Ionia, Greece
| | - Spyridon Skoularigkis
- Department of Cardiology, University Hospital of Larissa, 41110 Larissa, Greece; (S.S.); (N.S.); (F.T.)
| | - Dimitrios E. Magouliotis
- Unit of Quality Improvement, Department of Cardiothoracic Surgery, University of Thessaly, 41110 Larissa, Greece;
| | - Niki Skopeliti
- Department of Cardiology, University Hospital of Larissa, 41110 Larissa, Greece; (S.S.); (N.S.); (F.T.)
| | | | - Alexandros Briasoulis
- Department of Therapeutics, Heart Failure and Cardio-Oncology Clinic, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Filippos Triposkiadis
- Department of Cardiology, University Hospital of Larissa, 41110 Larissa, Greece; (S.S.); (N.S.); (F.T.)
| | - John Skoularigis
- Department of Cardiology, University Hospital of Larissa, 41110 Larissa, Greece; (S.S.); (N.S.); (F.T.)
| |
Collapse
|
47
|
Glavaški M, Velicki L, Vučinić N. Hypertrophic Cardiomyopathy: Genetic Foundations, Outcomes, Interconnections, and Their Modifiers. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1424. [PMID: 37629714 PMCID: PMC10456451 DOI: 10.3390/medicina59081424] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/30/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is the most prevalent heritable cardiomyopathy. HCM is considered to be caused by mutations in cardiac sarcomeric protein genes. Recent research suggests that the genetic foundation of HCM is much more complex than originally postulated. The clinical presentations of HCM are very variable. Some mutation carriers remain asymptomatic, while others develop severe HCM, terminal heart failure, or sudden cardiac death. Heterogeneity regarding both genetic mutations and the clinical course of HCM hinders the establishment of universal genotype-phenotype correlations. However, some trends have been identified. The presence of a mutation in some genes encoding sarcomeric proteins is associated with earlier HCM onset, more severe left ventricular hypertrophy, and worse clinical outcomes. There is a diversity in the mechanisms implicated in the pathogenesis of HCM. They may be classified into groups, but they are interrelated. The lack of known supplementary elements that control the progression of HCM indicates that molecular mechanisms that exist between genotype and clinical presentations may be crucial. Secondary molecular changes in pathways implicated in HCM pathogenesis, post-translational protein modifications, and epigenetic factors affect HCM phenotypes. Cardiac loading conditions, exercise, hypertension, diet, alcohol consumption, microbial infection, obstructive sleep apnea, obesity, and environmental factors are non-molecular aspects that change the HCM phenotype. Many mechanisms are implicated in the course of HCM. They are mostly interconnected and contribute to some extent to final outcomes.
Collapse
Affiliation(s)
- Mila Glavaški
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia; (L.V.)
| | - Lazar Velicki
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia; (L.V.)
- Institute of Cardiovascular Diseases Vojvodina, Put Doktora Goldmana 4, 21204 Sremska Kamenica, Serbia
| | - Nataša Vučinić
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia; (L.V.)
| |
Collapse
|
48
|
Shang Y, Liu R, Gan J, Yang Y, Sun L. Construction of cardiac fibrosis for biomedical research. SMART MEDICINE 2023; 2:e20230020. [PMID: 39188350 PMCID: PMC11235890 DOI: 10.1002/smmd.20230020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 07/22/2023] [Indexed: 08/28/2024]
Abstract
Cardiac remodeling is critical for effective tissue recuperation, nevertheless, excessive formation and deposition of extracellular matrix components can result in the onset of cardiac fibrosis. Despite the emergence of novel therapies, there are still no lifelong therapeutic solutions for this issue. Understanding the detrimental cardiac remodeling may aid in the development of innovative treatment strategies to prevent or reverse fibrotic alterations in the heart. Further combining the latest understanding of disease pathogenesis with cardiac tissue engineering has provided the conversion of basic laboratory studies into the therapy of cardiac fibrosis patients as an increasingly viable prospect. This review presents the current main mechanisms and the potential tissue engineering of cardiac fibrosis. Approaches using biomedical materials-based cardiac constructions are reviewed to consider key issues for simulating in vitro cardiac fibrosis, outlining a future perspective for preclinical applications.
Collapse
Affiliation(s)
- Yixuan Shang
- Department of Medical Supplies SupportNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Rui Liu
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Jingjing Gan
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Yuzhi Yang
- Department of Medical Supplies SupportNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Lingyun Sun
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| |
Collapse
|
49
|
da Silva VL, Mota GAF, de Souza SLB, de Campos DHS, Melo AB, Vileigas DF, Coelho PM, Sant’Ana PG, Padovani C, Lima-Leopoldo AP, Bazan SGZ, Leopoldo AS, Cicogna AC. Aerobic Exercise Training Improves Calcium Handling and Cardiac Function in Rats with Heart Failure Resulting from Aortic Stenosis. Int J Mol Sci 2023; 24:12306. [PMID: 37569680 PMCID: PMC10418739 DOI: 10.3390/ijms241512306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/20/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Aerobic exercise training (AET) has been used to manage heart disease. AET may totally or partially restore the activity and/or expression of proteins that regulate calcium (Ca2+) handling, optimize intracellular Ca2+ flow, and attenuate cardiac functional impairment in failing hearts. However, the literature presents conflicting data regarding the effects of AET on Ca2+ transit and cardiac function in rats with heart failure resulting from aortic stenosis (AoS). This study aimed to evaluate the impact of AET on Ca2+ handling and cardiac function in rats with heart failure due to AoS. Wistar rats were distributed into two groups: control (Sham; n = 61) and aortic stenosis (AoS; n = 44). After 18 weeks, the groups were redistributed into: non-exposed to exercise training (Sham, n = 28 and AoS, n = 22) and trained (Sham-ET, n = 33 and AoS-ET, n = 22) for 10 weeks. Treadmill exercise training was performed with a velocity equivalent to the lactate threshold. The cardiac function was analyzed by echocardiogram, isolated papillary muscles, and isolated cardiomyocytes. During assays of isolated papillary muscles and isolated cardiomyocytes, the Ca2+ concentrations were evaluated. The expression of regulatory proteins for diastolic Ca2+ was assessed via Western Blot. AET attenuated the diastolic dysfunction and improved the systolic function. AoS-ET animals presented an enhanced response to post-rest contraction and SERCA2a and L-type Ca2+ channel blockage compared to the AoS. Furthermore, AET was able to improve aspects of the mechanical function and the responsiveness of the myofilaments to the Ca2+ of the AoS-ET animals. AoS animals presented an alteration in the protein expression of SERCA2a and NCX, and AET restored SERCA2a and NCX levels near normal values. Therefore, AET increased SERCA2a activity and myofilament responsiveness to Ca2+ and improved the cellular Ca2+ influx mechanism, attenuating cardiac dysfunction at cellular, tissue, and chamber levels in animals with AoS and heart failure.
Collapse
Affiliation(s)
- Vítor Loureiro da Silva
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, Brazil; (G.A.F.M.); (S.L.B.d.S.); (D.H.S.d.C.); (D.F.V.); (P.G.S.); (S.G.Z.B.); (A.C.C.)
| | - Gustavo Augusto Ferreira Mota
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, Brazil; (G.A.F.M.); (S.L.B.d.S.); (D.H.S.d.C.); (D.F.V.); (P.G.S.); (S.G.Z.B.); (A.C.C.)
| | - Sérgio Luiz Borges de Souza
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, Brazil; (G.A.F.M.); (S.L.B.d.S.); (D.H.S.d.C.); (D.F.V.); (P.G.S.); (S.G.Z.B.); (A.C.C.)
| | - Dijon Henrique Salomé de Campos
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, Brazil; (G.A.F.M.); (S.L.B.d.S.); (D.H.S.d.C.); (D.F.V.); (P.G.S.); (S.G.Z.B.); (A.C.C.)
| | - Alexandre Barroso Melo
- Department of Sports, Federal University of Espirito Santo, Vitória 29075-910, Brazil; alexandre-- (A.B.M.); (P.M.C.); (A.P.L.-L.); (A.S.L.)
| | - Danielle Fernandes Vileigas
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, Brazil; (G.A.F.M.); (S.L.B.d.S.); (D.H.S.d.C.); (D.F.V.); (P.G.S.); (S.G.Z.B.); (A.C.C.)
| | - Priscila Murucci Coelho
- Department of Sports, Federal University of Espirito Santo, Vitória 29075-910, Brazil; alexandre-- (A.B.M.); (P.M.C.); (A.P.L.-L.); (A.S.L.)
| | - Paula Grippa Sant’Ana
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, Brazil; (G.A.F.M.); (S.L.B.d.S.); (D.H.S.d.C.); (D.F.V.); (P.G.S.); (S.G.Z.B.); (A.C.C.)
| | - Carlos Padovani
- Department of Biostatistics, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-689, Brazil;
| | - Ana Paula Lima-Leopoldo
- Department of Sports, Federal University of Espirito Santo, Vitória 29075-910, Brazil; alexandre-- (A.B.M.); (P.M.C.); (A.P.L.-L.); (A.S.L.)
| | - Silméia Garcia Zanati Bazan
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, Brazil; (G.A.F.M.); (S.L.B.d.S.); (D.H.S.d.C.); (D.F.V.); (P.G.S.); (S.G.Z.B.); (A.C.C.)
| | - André Soares Leopoldo
- Department of Sports, Federal University of Espirito Santo, Vitória 29075-910, Brazil; alexandre-- (A.B.M.); (P.M.C.); (A.P.L.-L.); (A.S.L.)
| | - Antonio Carlos Cicogna
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, Brazil; (G.A.F.M.); (S.L.B.d.S.); (D.H.S.d.C.); (D.F.V.); (P.G.S.); (S.G.Z.B.); (A.C.C.)
| |
Collapse
|
50
|
Guo Y, You Y, Shang FF, Wang X, Huang B, Zhao B, Lv D, Yang S, Xie M, Kong L, Du D, Luo S, Tian X, Xia Y. iNOS aggravates pressure overload-induced cardiac dysfunction via activation of the cytosolic-mtDNA-mediated cGAS-STING pathway. Theranostics 2023; 13:4229-4246. [PMID: 37554263 PMCID: PMC10405855 DOI: 10.7150/thno.84049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 07/14/2023] [Indexed: 08/10/2023] Open
Abstract
Background: Sterile inflammation contributes to the pathogenesis of cardiac dysfunction caused by various conditions including pressure overload in hypertension. Mitochondrial DNA (mtDNA) released from damaged mitochondria has been implicated in cardiac inflammation. However, the upstream mechanisms governing mtDNA release and how mtDNA activates sterile inflammation in pressure-overloaded hearts remain largely unknown. Here, we investigated the role of inducible NO synthase (iNOS) on pressure overload-induced cytosolic accumulation of mtDNA and whether mtDNA activated inflammation through the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway. Methods: To investigate whether the cGAS-STING cascade was involved in sterile inflammation and cardiac dysfunction upon pressure overload, cardiomyocyte-specific STING depletion mice and mice injected with adeno-associated virus-9 (AAV-9) to suppress the cGAS-STING cascade in the heart were subjected to transverse aortic constriction (TAC). iNOS null mice were used to determine the role of iNOS in cGAS-STING pathway activation in pressure-stressed hearts. Results: iNOS knockout abrogated mtDNA release and alleviated cardiac sterile inflammation resulting in improved cardiac function. Conversely, activating the cGAS-STING pathway blunted the protective effects of iNOS knockout. Moreover, iNOS activated the cGAS-STING pathway in isolated myocytes and this was prevented by depleting cytosolic mtDNA. In addition, disruption of the cGAS-STING pathway suppressed inflammatory cytokine transcription and modulated M1/M2 macrophage polarization, and thus mitigated cardiac remodeling and improved heart function. Finally, increased iNOS expression along with cytosolic mtDNA accumulation and cGAS-STING activation were also seen in human hypertensive hearts. Conclusion: Our findings demonstrate that mtDNA is released into the cytosol and triggers sterile inflammation through the cGAS-STING pathway leading to cardiac dysfunction after pressure overload. iNOS controls mtDNA release and subsequent cGAS activation in pressure-stressed hearts.
Collapse
Affiliation(s)
- Yongzheng Guo
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yuehua You
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Fei-Fei Shang
- Institute of Life Science, Chongqing Medical University, Chongqing 400016, China
| | - Xiaowen Wang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Bi Huang
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Boying Zhao
- Department of Cardiothoracic Surgery, Chongqing University Central Hospital, Chongqing 400014, China
| | - Dingyi Lv
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Shenglan Yang
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Ming Xie
- Department of Cardiothoracic Surgery, Chongqing University Central Hospital, Chongqing 400014, China
| | - Lingwen Kong
- Department of Cardiothoracic Surgery, Chongqing University Central Hospital, Chongqing 400014, China
| | - Dingyuan Du
- Department of Cardiothoracic Surgery, Chongqing University Central Hospital, Chongqing 400014, China
| | - Suxin Luo
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xin Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Yong Xia
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Institute of Life Science, Chongqing Medical University, Chongqing 400016, China
- Davis Heart & Lung Research Institute, Division of Cardiovascular Medicine, The Ohio State University College of Medicine, OH 43210, USA
| |
Collapse
|