1
|
Dunzhu D, Han G, Shanshan Q, Li S, Yang J, He J, Gou S, Dong G, Jiang C, Hou J. The role of Perilipin 5 in pathological myocardial remodeling. Front Pharmacol 2025; 16:1526494. [PMID: 40166465 PMCID: PMC11955653 DOI: 10.3389/fphar.2025.1526494] [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: 11/11/2024] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Pathological cardiac remodeling (REM), caused by various pathological factors and characterized by changes in cardiac structure and geometry, is strongly associated with heart failure (HF). It damages cardiac tissue, alters energy metabolism, increases oxidative stress, and cause matrix metalloproteinase activation, cardiomyocyte hypertrophy, and interstitial fibrosis, leading to HF. REM determines the outcome of cardiovascular disease. Current treatments have limitations. REM is associated with cardiac energetic remodeling, and modulation of metabolic substrates may slow down the disease. Perilipin 5 (Plin5), positioned as a structural protein located on the surface of lipid droplets (LDs), is abundant in tissues and cells that rely on mitochondrial β-oxidation for energy production. It is the most recently identified member of the perilipin protein (PAT) family, with a notable enrichment in the cardiac muscle. Emerging evidence highlights the critical role of intracellular LD in the regulation of energy metabolism, with metabolic disruptions of LD being directly correlated with the incidence of metabolic disease. As a key barrier to LD, Plin5 is instrumental in controlling the catabolism of LD and regulating the metabolism and transport of fatty acids (FAs). As a protectant against excessive β-oxidation of free fatty acids (FFAs), Plin5 acts to isolate and neutralize overly oxidized fatty acids, thereby shielding the heart from myocardial remodeling instigated by a variety of etiological factors. This protective mechanism helps to ameliorate the progression of persistent and detrimental myocardial remodeling, which can otherwise lead to the development of severe heart failure. This systematic review attempts to delineate the metabolic disorders associated with pathological cardiac remodeling, focusing on the properties and regulatory mechanisms of Plin5. By synthesising current literature, it investigates the pivotal role of Plin5 in modulating the distinctive attributes, initiating factors, and molecular signaling networks underpinning pathological cardiac remodeling.
Collapse
Affiliation(s)
| | - Gao Han
- School of Stomatology, Qilu Medical University, Zibo, China
| | - Qin Shanshan
- School of Medicine, Tibet University, Lhasa, China
| | - Shangshi Li
- The Department of High Mountain Sickness, The General Hospital of Xizang Military Area Command, Xizang, China
| | - Jiali Yang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Jian He
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Siyu Gou
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Gang Dong
- School of Stomatology, Qilu Medical University, Zibo, China
| | - Chunrong Jiang
- School of Stomatology, Qilu Medical University, Zibo, China
| | - Jun Hou
- The Third People’s Hospital of Chengdu, Chengdu, China
| |
Collapse
|
2
|
Mołek-Dziadosz P, Natorska J, Matusik PT, Wojciechowska W, Rajzer M, Rajtar-Salwa R, Bartuś S, Surdacki A, Malinowski KP, Lenart-Migdalska A, Olszowska M, Ząbczyk M. Left ventricular remodeling in atrial fibrillation is associated with elevated NAP-2 and 3-nitrotyrosine. Biomark Med 2025; 19:157-164. [PMID: 39935376 PMCID: PMC11916376 DOI: 10.1080/17520363.2025.2463882] [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: 03/30/2024] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Atrial fibrillation (AF) coexists with left ventricular (LV) dysfunction and remodeling. Neutrophil infiltration of cardiac muscle tissue and oxidative stress are associated with cardiac remodeling. MATERIALS AND METHODS In 221 AF patients aged 70 (62-76) years, LV remodeling was assessed using echocardiography. Plasma concentrations of neutrophil-activating peptide 2 (NAP-2) and 3-nitrotyrosine were measured. RESULTS AF patients with concentric and eccentric hypertrophy showed higher NAP-2 levels than those with normal LV geometry (+41% and + 29%, p = 0.001 and p = 0.025, respectively). Moreover, concentric hypertrophy was associated with 85% higher 3-nitrotyrosine level (p = 0.01) compared to normal LV geometry. CONCLUSIONS Elevated NAP-2, associated with oxidative damage, may contribute to the development of LV hypertrophy.
Collapse
Affiliation(s)
- Patrycja Mołek-Dziadosz
- Department of Coronary Artery Disease and Heart Failure, Jagiellonian University Medical College, Krakow, Poland
| | - Joanna Natorska
- Krakow Centre for Medical Research and Technologies, The St. John Paul II Hospital, Krakow, Poland
- Department of Thromboembolic Disorders, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
| | - Paweł T Matusik
- Institute of Cardiology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
- Department of Electrocardiology, the St. John Paul II Hospital, Kraków, Poland
| | - Wiktoria Wojciechowska
- 1st Department of Cardiology, Interventional Electrocardiology and Arterial Hypertension, Jagiellonian University Medical College, Kraków, Poland
| | - Marek Rajzer
- 1st Department of Cardiology, Interventional Electrocardiology and Arterial Hypertension, Jagiellonian University Medical College, Kraków, Poland
| | - Renata Rajtar-Salwa
- Department of Cardiology and Cardiovascular Interventions, University Hospital, Krakow, Poland
| | - Stanisław Bartuś
- Department of Cardiology and Cardiovascular Interventions, University Hospital, Krakow, Poland
| | - Andrzej Surdacki
- Department of Cardiology and Cardiovascular Interventions, University Hospital, Krakow, Poland
| | - Krzysztof P Malinowski
- Department of Bioinformatics and Telemedicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
- Center for Digital Medicine and Robotics, Jagiellonian University Medical College, Kraków, Poland
| | - Aleksandra Lenart-Migdalska
- Department of Cardiac and Vascular Diseases, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Maria Olszowska
- Department of Cardiac and Vascular Diseases, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Michał Ząbczyk
- Krakow Centre for Medical Research and Technologies, The St. John Paul II Hospital, Krakow, Poland
- Department of Thromboembolic Disorders, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
3
|
Familusi MA, Skatulla S, Hussan JR, Aremu OO, Jermy S, Mutithu D, Gumedze FN, Ntusi NAB. Bi-ventricular elastic material parameters estimation using 3D CMR myocardial strains in rheumatic heart disease patients. J Biomech 2025; 181:112524. [PMID: 39842217 DOI: 10.1016/j.jbiomech.2025.112524] [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: 07/11/2024] [Revised: 12/04/2024] [Accepted: 01/10/2025] [Indexed: 01/24/2025]
Abstract
Understanding the elastic material behavior of myocardium during the diastolic phase is critical for evaluating cardiac function and improving treatments for diastolic abnormalities. This study introduces a novel multi-objective optimization framework that incorporates both strain and volume measurements to enhance the accuracy of myocardial property assessments in Rheumatic Heart Disease (RHD) patients and healthy controls. By employing global volume and strain measurements instead of segmented strains from the sixteen AHA regions, we achieve a robust alignment with the Klotz curve across all groups, indicating an accurate simulation of end-diastolic pressure-volume relationships (EDPVRs). Our approach uniquely integrates combinations of longitudinal, circumferential, and radial strains, resulting in an unprecedented reduction in errors between clinical and simulated strain values, with less than one percent difference for targeted parameters. The results demonstrate that the alignment between computational predictions and clinical measurements depends significantly on the choice of optimization target. The study reveals significant differences in tissue mechanics between RHD patients and healthy controls, with notable variations in ventricular stiffness and fiber orientations across optimization targets, confirmed through rigorous statistical analyses. The observed variations in fiber angles, particularly the smaller angles for longitudinal strains and steeper angles for circumferential strains, underscore the intricate relationship between myocardial fiber architecture and cardiac deformation, offering deeper insights into ventricular biomechanics. By presenting qualitative and quantitative differences in stress and strain distributions, this research advances the understanding of myocardial mechanics, highlighting the clinical relevance of fiber orientation and material properties in modeling cardiac mechanics and distinguishing diseased from healthy myocardial behavior.
Collapse
Affiliation(s)
- Mary A Familusi
- Computational Continuum Mechanics Research Group, Department of Civil Engineering, University of Cape Town, South Africa; South African DST-NRF Centre of Excellence in Epidemiological Modelling and Analysis, Stellenbosch University, South Africa.
| | - Sebastian Skatulla
- Computational Continuum Mechanics Research Group, Department of Civil Engineering, University of Cape Town, South Africa.
| | - Jagir R Hussan
- Auckland Bioengineering Institute, University of Auckland, New Zealand.
| | - Olukayode O Aremu
- Division of Cardiology, Department of Medicine, University of Cape Town, Cape Town, South Africa; Cape Universities Body Imaging Centre, Faculty of Health Sciences, University of Cape Town, South Africa.
| | - Stephen Jermy
- Cape Universities Body Imaging Centre, Faculty of Health Sciences, University of Cape Town, South Africa; Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Cape Town, South Africa.
| | - Daniel Mutithu
- Division of Cardiology, Department of Medicine, University of Cape Town, Cape Town, South Africa; Cape Universities Body Imaging Centre, Faculty of Health Sciences, University of Cape Town, South Africa.
| | - Freedom N Gumedze
- Department of Statistical Sciences, University of Cape Town, South Africa.
| | - Ntobeko A B Ntusi
- Division of Cardiology, Department of Medicine, University of Cape Town, Cape Town, South Africa; Cape Universities Body Imaging Centre, Faculty of Health Sciences, University of Cape Town, South Africa; South African Medical Research Council Extramural Unit on Intersection of Noncommunicable Diseases and Infectious Diseases.
| |
Collapse
|
4
|
Barnet IR, Schulz NE, Ghelani SJ, Hoganson DM, Feins EN, Hammer PE, Emani SM, Sleeper LA, Beroukhim RS. Wide variation in shape of hypoplastic left ventricles undergoing recruitment and biventricular repair: A statistical shape modeling study. J Cardiovasc Magn Reson 2024; 27:101131. [PMID: 39647766 PMCID: PMC11780089 DOI: 10.1016/j.jocmr.2024.101131] [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: 04/11/2024] [Revised: 11/17/2024] [Accepted: 12/02/2024] [Indexed: 12/10/2024] Open
Abstract
BACKGROUND Patients with hypoplastic left ventricles (LV) who undergo volume-loading procedures (recruitment, biventricular [BIV] repair) are at risk for adverse outcomes, including heart failure and death. We investigated pre-BIV LV shape as a predictor of outcome after BIV repair in patients with hypoplastic LVs. METHODS Baseline and post-recruitment cardiac magnetic resonance imaging and computed tomography data were analyzed in patients with hypoplastic LV (<50 mL/m2). Statistical shape modeling (SSM) was utilized to generate a model of the shape and variability of LVs. Traditional measures of LV sphericity and eccentricity were also measured. Major adverse cardiovascular events (MACE) included heart failure, transplant, and death. RESULTS Of 95 patients with baseline mean LV volume 29 ± 13 mL/m2, 45/95 (47%) had a right dominant atrioventricular canal defect, 31/95 (33%) had a variant of hypoplastic left heart syndrome, and 18/95 (19%) had endocardial fibroelastosis (EFE). A wide variation in LV shape was found by SSM, and shape modes were associated with right ventricle (RV) and LV size, and diagnosis. BIV repair was achieved in 74/95 (78%) patients; 13/74 (18%) of BIV patients had MACE. Predictors of MACE following BIV repair included EFE, higher RV mass index, and higher RV end-diastolic volume index. No baseline or post-recruitment LV shape parameter was associated with the outcome after BIV repair. CONCLUSION The shape model of hypoplastic LVs demonstrated a wide array of LV shapes. LVs gained sphericity and size and lost eccentricity with recruitment. Though the ventricles changed shape with recruitment, no specific LV shape characteristic at the baseline or post-recruitment stage was predictive of decision to proceed with BIV repair or outcome. Higher RV mass and volume may represent new biomarkers that predict outcomes following BIV repair in patients with hypoplastic LV. Further investigation could determine the reproducibility of these findings.
Collapse
Affiliation(s)
| | - Noah E Schulz
- Harvard Medical School, Boston, Massachusetts, USA; Department of Cardiovascular Surgery, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Sunil J Ghelani
- Harvard Medical School, Boston, Massachusetts, USA; Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - David M Hoganson
- Harvard Medical School, Boston, Massachusetts, USA; Department of Cardiovascular Surgery, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Eric N Feins
- Harvard Medical School, Boston, Massachusetts, USA; Department of Cardiovascular Surgery, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Peter E Hammer
- Harvard Medical School, Boston, Massachusetts, USA; Department of Cardiovascular Surgery, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Sitaram M Emani
- Harvard Medical School, Boston, Massachusetts, USA; Department of Cardiovascular Surgery, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Lynn A Sleeper
- Harvard Medical School, Boston, Massachusetts, USA; Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Rebecca S Beroukhim
- Harvard Medical School, Boston, Massachusetts, USA; Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA.
| |
Collapse
|
5
|
Tsuda T, Robinson BW. Beneficial Effects of Exercise on Hypertension-Induced Cardiac Hypertrophy in Adolescents and Young Adults. Curr Hypertens Rep 2024; 26:451-462. [PMID: 38888690 DOI: 10.1007/s11906-024-01313-4] [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] [Accepted: 05/30/2024] [Indexed: 06/20/2024]
Abstract
PURPOSE OF REVIEW Hypertension-induced cardiac hypertrophy is widely known as a major risk factor for increased cardiovascular morbidity and mortality. Although exercise is proven to exert overall beneficial effects on hypertension and hypertension-induced cardiac hypertrophy, there are some concerns among providers about potential adverse effects induced by intense exercise, especially in hypertensive athletes. We will overview the underlying mechanisms of physiological and pathological hypertrophy and delineate the beneficial effects of exercise in young people with hypertension and consequent hypertrophy. RECENT FINDINGS Multiple studies have demonstrated that exercise training, both endurance and resistance types, reduces blood pressure and ameliorates hypertrophy in hypertensives, but certain precautions are required for hypertensive athletes when allowing competitive sports: Elevated blood pressure should be controlled before allowing them to participate in high-intensity exercise. Non-vigorous and recreational exercise are always recommended to promote cardiovascular health. Exercise-induced cardiac adaptation is a benign and favorable response that reverses or attenuates pathological cardiovascular remodeling induced by persistent hypertension. Exercise is the most effective nonpharmacological treatment for hypertensive individuals. Distinction between recreational-level exercise and competitive sports should be recognized by medical providers when allowing sports participation for adolescents and young adults.
Collapse
Affiliation(s)
- Takeshi Tsuda
- Nemours Cardiac Center, Nemours Children's Health, 1600 Rockland Rd, Wilmington, DE, 19803, USA.
- Department of Pediatrics, Sidney Kimmel Medical College at Thomas Jefferson University, Philadephia, PA, 19107, USA.
| | - Bradley W Robinson
- Nemours Cardiac Center, Nemours Children's Health, 1600 Rockland Rd, Wilmington, DE, 19803, USA
- Department of Pediatrics, Sidney Kimmel Medical College at Thomas Jefferson University, Philadephia, PA, 19107, USA
| |
Collapse
|
6
|
Zia-Ul-Sabah, Alqahtani SAM, Alghamdi BH, Wani JI, Aziz S, Durrani HK, Patel AA, Rangraze I, Wani SJ. Association of type-D personality and left-ventricular remodelling in patients treated with primary percutaneous intervention after ST-segment elevation myocardial infarction. BMC Cardiovasc Disord 2024; 24:600. [PMID: 39468433 PMCID: PMC11520066 DOI: 10.1186/s12872-024-04254-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 10/14/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Type-D personality is an established predisposing factor for various diseases. Type-D traits have been shown to pose a 26% increased risk of coronary artery disease after controlling for other confounding factors. Significant associations have been reported between type-D personality traits and dyslipidaemia, impaired endothelial function, coronary heart disease (CAD), acute myocardial infarction, and other adverse cardiovascular events. OBJECTIVE To assess the association between type-D personality and left-ventricular adverse remodelling in patients treated with percutaneous coronary intervention following index ST-segment elevation myocardial infarction. METHODS All patients hospitalized and treated with percutaneous coronary intervention (PCI) after their index ST-segment elevation myocardial infarction (STEMI) between 1 January 2022 to 31 December 2023 were prospectively enrolled. Type-D personality traits in the study population were determined at baseline using type-D Scale-14 (DS14) instrument, whereas any positive change in left ventricular end diastolic volume (LVEDV) ≥ 20% at follow up period of 12-months from baseline was defined as left-ventricular adverse remodelling (LVAR). Univariate and multivariate analysis was done to establish the independent predictors of LVAR. The area under receiver-operating characteristic curve (AUROC) was employed to assess the sensitivity and specificity of the identified independent predictors. RESULTS A total of 124 patients were enrolled in the study. The mean age of the study population was 67 ± 10 years and the overall incidence of LVAR was found to be 25%. Multivariate regression analysis revealed that type-D personality is a significant independent predictor of LVAR [Formula: see text] apart from the already established independent predictors Killip Class[Formula: see text], baseline Global Longitudinal strain (GLS)[Formula: see text], and 3-vessel CAD[Formula: see text]. In ROC curve analysis type-D personality as an independent predictor of LVAR achieved a sensitivity of 41.4% and a specificity of 87.1%, p < 0.02. CONCLUSION Type-D personality trait is a significant independent predictor of LVAR in patients treated with PCI after their index-STEMI.
Collapse
Affiliation(s)
- Zia-Ul-Sabah
- Department of Medicine, College of Medicine, King Khalid University, Abha, Saudi Arabia.
| | | | - Bandar Hezam Alghamdi
- Prince Faisal bin Khalid Cardiac Center, King Faisal Medical City, Abha, Saudi Arabia
| | - Javed Iqbal Wani
- Department of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Shahid Aziz
- Department of Medicine, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | | | - Ayyub Ali Patel
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Imran Rangraze
- Department of Internal Medicine, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, UAE
| | | |
Collapse
|
7
|
Czapska AH, Kosińska-Kaczyńska K. The Significance of the Myocardial Performance Index and Fetal Doppler Abnormalities in Growth-Restricted Fetuses: A Systematic Review of the Literature. J Clin Med 2024; 13:6469. [PMID: 39518608 PMCID: PMC11546427 DOI: 10.3390/jcm13216469] [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: 09/14/2024] [Revised: 10/09/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction: This review aims to investigate the clinical implications of using the myocardial performance index (MPI), obtained through tissue Doppler imaging (TDI) and spectral Doppler, in assessing fetal cardiac function in growth-restricted fetuses. It explores the MPI's potential in predicting adverse perinatal outcomes and its utility when combined with conventional pulsed-wave Doppler assessments for enhanced fetal well-being evaluations. Material and Methods: A systematic search of PubMed and Google Scholar databases spanning from 2004 to 2023 was conducted to identify pertinent articles on the MPI's clinical application in managing growth-restricted fetuses. Inclusion criteria followed the Fetal Medicine Barcelona definition of fetal growth restriction (FGR) to mitigate study group heterogeneity. The research sources were PubMed and Google Scholar databases, and the review was conducted without any specific clinical or laboratory setting. Only articles meeting the inclusion criteria for FGR, as per the Fetal Medicine Barcelona definition, were considered. Six studies meeting these criteria were included in the review. The review analyzed the correlation between MPI values and conventional Doppler parameters, investigating the progression of myocardial function impairment and its association with the risk of fetal demise. The primary outcome measures included the relationship between MPI values, fetal well-being, and the potential for prenatal cardiac dysfunction in growth-restricted fetuses. Results: The findings indicate that as conventional Doppler parameters deteriorate, MPI values increase, suggesting progressive myocardial dysfunction. The MPI may cross the 95th percentile before abnormal flow in the ductus venosus and aortic isthmus, highlighting the potential for diastolic dysfunction preceding hypoxia in growth-restricted fetuses. Elevated MPI levels were observed in both growth-restricted and small-for-gestational-age (SGA) fetuses, indicating prenatal cardiac impairment. The strong association between an abnormal MPI and perinatal mortality has been shown for early FGR. Conclusions: MPI alterations appear to precede abnormal Doppler parameters in early- and late- onset FGR, potentially indicating diastolic dysfunction preceding hypoxia. Additionally, the MPI correlates with the risk of fetal demise. However, larger studies are needed to establish its sensitivity and specificity. Furthermore, the significance of prenatal cardiac impairment in some SGA fetuses raises questions about its potential impact on perinatal outcomes and cardiovascular programming.
Collapse
Affiliation(s)
| | - Katarzyna Kosińska-Kaczyńska
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, Cegłowska St. 80, 01-809 Warsaw, Poland;
| |
Collapse
|
8
|
Cheng Y, Lin G, Xie Y, Xuan B, He S, Shang Z, Yan M, Lin J, Wei L, Peng J, Shen A. Baicalin ameliorates angiotensin II-induced cardiac hypertrophy and mitogen-activated protein kinase signaling pathway activation: A target-based network pharmacology approach. Eur J Pharmacol 2024; 981:176876. [PMID: 39127302 DOI: 10.1016/j.ejphar.2024.176876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/20/2024] [Accepted: 08/08/2024] [Indexed: 08/12/2024]
Abstract
Baicalin, a flavonoid glycoside from Scutellaria baicalensis Georgi., exerts anti-hypertensive effects. The present study aimed to assess the cardioprotective role of baicalin and explore its potential mechanisms. Network pharmacology analysis pointed out a total of 477 potential targets of baicalin were obtained from the PharmMapper and SwissTargetPrediction databases, while 11,280 targets were identified associating with hypertensive heart disease from GeneCards database. Based on the above 382 common targets, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses revealed enrichment in the regulation of cardiac hypertrophy, cardiac contraction, cardiac relaxation, as well as the mitogen-activated protein kinase (MAPK) and other signaling pathways. Moreover, baicalin treatment exhibited the amelioration of increased cardiac index and pathological alterations in angiotensin II (Ang II)-infused C57BL/6 mice. Furthermore, baicalin treatment demonstrated a reduction in cell surface area and a down-regulation of hypertrophy markers (including atrial natriuretic peptide and brain natriuretic peptide) in vivo and in vitro. In addition, baicalin treatment led to a decrease in the expression of phosphorylated c-Jun N-terminal kinase (p-JNK)/JNK, phosphorylated p38 (p-p38)/p38, and phosphorylated extracellular signal-regulated kinase (p-ERK)/ERK in the cardiac tissues of Ang II-infused mice and Ang II-stimulated H9c2 cells. These findings highlight the cardioprotective effects of baicalin, as it alleviates hypertensive cardiac injury, cardiac hypertrophy, and the activation of the MAPK pathway.
Collapse
Affiliation(s)
- Ying Cheng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Guosheng Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Yi Xie
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Bihan Xuan
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Shuyu He
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Zucheng Shang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Mengchao Yan
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Jing Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Lihui Wei
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China; Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China.
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China.
| | - Aling Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China; Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China.
| |
Collapse
|
9
|
Lee YS, Choi JR, Kim JB. Gene Therapy for Cardiovascular Disease: Clinical Perspectives. Yonsei Med J 2024; 65:557-571. [PMID: 39313446 PMCID: PMC11427124 DOI: 10.3349/ymj.2024.0127] [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: 05/22/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 09/25/2024] Open
Abstract
Cardiovascular disease (CVD) stands as one of the leading causes of death in the United States, with its prevalence steadily on the rise. Traditional therapeutic approaches, such as pharmacological treatment, cardiovascular intervention, and surgery, have inherent limitations. In response to these challenges, cardiac gene therapy has emerged as a promising alternative for treating CVD patients. However, several obstacles persist, including the low efficiency of gene transduction, immune reactions to vectors or transduced cells, and the occurrence of off-target effects. While preclinical research has demonstrated significant success in various CVD model in both small and large animals, the translation of these findings to clinical applications has, for the most part, yielded disappointing results, except for some early, albeit small, trials. This review aims to provide a comprehensive summary of recent preclinical and clinical studies on gene therapy for various CVDs. Additionally, we discuss the existing limitations and challenges that hinder the widespread clinical application of cardiac gene therapy.
Collapse
Affiliation(s)
- Young Shin Lee
- Division of Cardiology, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Jung Ran Choi
- College of Medicine, Kyung Hee University, Seoul, Korea
| | - Jin-Bae Kim
- Division of Cardiology, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul, Korea
- College of Medicine, Kyung Hee University, Seoul, Korea.
| |
Collapse
|
10
|
Pedrizzetti G, Numata R, Collia D, Pedrizzetti G, Zovatto L, Banerjee A. A scenario for heart failure during the filling phase. Sci Rep 2024; 14:22760. [PMID: 39354060 PMCID: PMC11445582 DOI: 10.1038/s41598-024-74155-4] [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: 08/17/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024] Open
Abstract
Heart failure (HF) is a life-threating cardiac disease that develops progressively for the reduced ability of the left ventricle (LV) to pump blood into the circulation during systole. HF can also develop in patients with a preserved systolic function, typically in presence of hypertrophic cardiomyopathy (HCM). This type of HF is sometimes termed as diastolic HF, but its biomechanical origin is still unclear. This study employs a physics-based analysis of both the LV and left atrium (LA) in selected HCM patients and matched healthy subjects using 3D echocardiography and demonstrates that alteration on the LV side (stiffening) reduces the elastic recovery of the LA. Moreover, the analysis of the forces exchanged between the two chambers demonstrates that they result unbalanced, keeping the LA in a sustained stretched condition that leads to dilation. This scenario clarifies the diastolic root of the dysfunction that may likely be the cause of the spiraling of events progressing toward failure of both LA emptying and LV filling. This deeply interdisciplinary study provides a physics-based basis for both physics/engineering modeling of heart function and to cardiologists for the design of clinical studies.
Collapse
Affiliation(s)
- Gianni Pedrizzetti
- Department of Engineering and Architecture, University of Trieste, Trieste, Italy.
| | - Ryusuke Numata
- Division of Cardiology, Department of Pediatrics, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - Dario Collia
- Department of Cardiovascular Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giulia Pedrizzetti
- Department of Chemical Engineering, Materials, Environment, INSTM Reference Laboratory for Engineering and Surface Treatments, Sapienza University of Rome, Rome, Italy
| | - Luigino Zovatto
- Department of Engineering and Architecture, University of Trieste, Trieste, Italy
| | - Anirban Banerjee
- Division of Cardiology, Department of Pediatrics, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
11
|
Ruud M, Frisk M, Melleby AO, Norseng PA, Mohamed BA, Li J, Aronsen JM, Setterberg IE, Jakubiczka J, van Hout I, Coffey S, Shen X, Nygård S, Lunde IG, Tønnessen T, Jones PP, Sjaastad I, Gullestad L, Toischer K, Dahl CP, Christensen G, Louch WE. Regulation of cardiomyocyte t-tubule structure by preload and afterload: Roles in cardiac compensation and decompensation. J Physiol 2024; 602:4487-4510. [PMID: 38686538 DOI: 10.1113/jp284566] [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: 02/22/2023] [Accepted: 04/02/2024] [Indexed: 05/02/2024] Open
Abstract
Mechanical load is a potent regulator of cardiac structure and function. Although high workload during heart failure is associated with disruption of cardiomyocyte t-tubules and Ca2+ homeostasis, it remains unclear whether changes in preload and afterload may promote adaptive t-tubule remodelling. We examined this issue by first investigating isolated effects of stepwise increases in load in cultured rat papillary muscles. Both preload and afterload increases produced a biphasic response, with the highest t-tubule densities observed at moderate loads, whereas excessively low and high loads resulted in low t-tubule levels. To determine the baseline position of the heart on this bell-shaped curve, mice were subjected to mildly elevated preload or afterload (1 week of aortic shunt or banding). Both interventions resulted in compensated cardiac function linked to increased t-tubule density, consistent with ascension up the rising limb of the curve. Similar t-tubule proliferation was observed in human patients with moderately increased preload or afterload (mitral valve regurgitation, aortic stenosis). T-tubule growth was associated with larger Ca2+ transients, linked to upregulation of L-type Ca2+ channels, Na+-Ca2+ exchanger, mechanosensors and regulators of t-tubule structure. By contrast, marked elevation of cardiac load in rodents and patients advanced the heart down the declining limb of the t-tubule-load relationship. This bell-shaped relationship was lost in the absence of electrical stimulation, indicating a key role of systolic stress in controlling t-tubule plasticity. In conclusion, modest augmentation of workload promotes compensatory increases in t-tubule density and Ca2+ cycling, whereas this adaptation is reversed in overloaded hearts during heart failure progression. KEY POINTS: Excised papillary muscle experiments demonstrated a bell-shaped relationship between cardiomyocyte t-tubule density and workload (preload or afterload), which was only present when muscles were electrically stimulated. The in vivo heart at baseline is positioned on the rising phase of this curve because moderate increases in preload (mice with brief aortic shunt surgery, patients with mitral valve regurgitation) resulted in t-tubule growth. Moderate increases in afterload (mice and patients with mild aortic banding/stenosis) similarly increased t-tubule density. T-tubule proliferation was associated with larger Ca2+ transients, with upregulation of the L-type Ca2+ channel, Na+-Ca2+ exchanger, mechanosensors and regulators of t-tubule structure. By contrast, marked elevation of cardiac load in rodents and patients placed the heart on the declining phase of the t-tubule-load relationship, promoting heart failure progression. The dependence of t-tubule structure on preload and afterload thus enables both compensatory and maladaptive remodelling, in rodents and humans.
Collapse
Affiliation(s)
- Marianne Ruud
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Michael Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Arne Olav Melleby
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Per Andreas Norseng
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Belal A Mohamed
- Department of Cardiology and Pneumology, Georg-August-University, Göttingen, Germany
| | - Jia Li
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Jan Magnus Aronsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Ingunn E Setterberg
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Joanna Jakubiczka
- Department of Cardiology and Pneumology, Georg-August-University, Göttingen, Germany
| | - Isabelle van Hout
- Department of Physiology, School of Biomedical Sciences and HeartOtago, University of Otago, Dunedin, New Zealand
| | - Sean Coffey
- Department of Medicine and HeartOtago, Dunedin School of Medicine, Dunedin Hospital, Dunedin, New Zealand
| | - Xin Shen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Ståle Nygård
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Ida G Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Theis Tønnessen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | - Peter P Jones
- Department of Physiology, School of Biomedical Sciences and HeartOtago, University of Otago, Dunedin, New Zealand
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Lars Gullestad
- Department of Cardiology, Oslo University Hospital, Oslo, Norway
| | - Karl Toischer
- Department of Cardiology and Pneumology, Georg-August-University, Göttingen, Germany
| | - Cristen P Dahl
- Department of Cardiology, Oslo University Hospital, Oslo, Norway
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| |
Collapse
|
12
|
Cai Y, Zhang J, Zhang H, Qi J, Shi C, Xu Y. The Kv4 potassium channel modulator NS5806 attenuates cardiac hypertrophy in vivo and in vitro. Sci Rep 2024; 14:19839. [PMID: 39191928 PMCID: PMC11349892 DOI: 10.1038/s41598-024-70962-x] [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: 01/19/2024] [Accepted: 08/22/2024] [Indexed: 08/29/2024] Open
Abstract
The compound NS5806 is a Kv4 channel modulator. This study investigated the chronic effects of NS5806 on cardiac hypertrophy induced by transverse aortic constriction (TAC) in mice in vivo and on neonatal rat ventricular cardiomyocyte hypertrophy induced by endothelin-1 (ET-1) in vitro. Four weeks after TAC, NS5806 was administered by gavage for 4 weeks. Echocardiograms revealed pronounced left ventricular (LV) hypertrophy in TAC-treated mice compared with sham mice. NS5806 attenuated LV hypertrophy, as manifested by the restoration of LV wall thickness and weight and the reversal of contractile dysfunction in TAC-treated mice. NS5806 also blunted the TAC-induced increases in the expression of cardiac hypertrophic and fibrotic genes, including ANP, BNP and TGF-β. Electrophysiological recordings revealed a significant prolongation of action potential duration and QT intervals, accompanied by an increase in susceptibility to ventricular arrhythmias in mice with cardiac hypertrophy. However, NS5806 restored these alterations in electrical parameters and thus reduced the incidence of mouse sudden death. Furthermore, NS5806 abrogated the downregulation of the Kv4 protein in the hypertrophic myocardium but did not influence the reduction in Kv4 mRNA expression. In addition, NS5806 suppressed in vitro cardiomyocyte hypertrophy. The results provide novel insight for further ion channel modulator development as a potential treatment option for cardiac hypertrophy.
Collapse
Affiliation(s)
- Yue Cai
- Department of Pharmacology, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, 050017, Hebei, China
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, Shijiazhuang, 050017, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
- Department of Pharmacy, Hebei General Hospital, Shijiazhuang, 050051, China
- Hebei Key Laboratory of Clinical Pharmacy, Shijiazhuang, 050051, China
| | - Jiali Zhang
- Department of Pharmacology, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, 050017, Hebei, China
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, Shijiazhuang, 050017, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Hongxue Zhang
- Department of Pharmacology, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, 050017, Hebei, China
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, Shijiazhuang, 050017, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jinlong Qi
- Department of Pharmacology, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, 050017, Hebei, China
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, Shijiazhuang, 050017, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Chenxia Shi
- Department of Pharmacology, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, 050017, Hebei, China
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, Shijiazhuang, 050017, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yanfang Xu
- Department of Pharmacology, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, 050017, Hebei, China.
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, Shijiazhuang, 050017, China.
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China.
| |
Collapse
|
13
|
Zhou L, Nishimura A, Umezawa K, Kato Y, Mi X, Ito T, Urano Y, Akaike T, Nishida M. Supersulfide catabolism participates in maladaptive remodeling of cardiac cells. J Pharmacol Sci 2024; 155:121-130. [PMID: 38880546 DOI: 10.1016/j.jphs.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/29/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
The atrophic myocardium resulting from mechanical unloading and nutritional deprivation is considered crucial as maladaptive remodeling directly associated with heart failure, as well as interstitial fibrosis. Conversely, myocardial hypertrophy resulting from hemodynamic loading is perceived as compensatory stress adaptation. We previously reported the abundant presence of highly redox-active polysulfide molecules, termed supersulfide, with two or more sulfur atoms catenated in normal hearts, and the supersulfide catabolism in pathologic hearts after myocardial infarction correlated with worsened prognosis of heart failure. However, the impact of supersulfide on myocardial remodeling remains unclear. Here, we investigated the involvement of supersulfide metabolism in cardiomyocyte remodeling, using a model of adenosine 5'-triphosphate (ATP) receptor-stimulated atrophy and endothelin-1 receptor-stimulated hypertrophy in neonatal rat cardiomyocytes. Results revealed contrasting changes in intracellular supersulfide and its catabolite, hydrogen sulfide (H2S), between cardiomyocyte atrophy and hypertrophy. Stimulation of cardiomyocytes with ATP decreased supersulfide activity, while H2S accumulation itself did not affect cardiomyocyte atrophy. This supersulfide catabolism was also involved in myofibroblast formation of neonatal rat cardiac fibroblasts. Thus, unraveling supersulfide metabolism during myocardial remodeling may lead to the development of novel therapeutic strategies to improve heart failure.
Collapse
Affiliation(s)
- Liuchenzi Zhou
- National Institute for Physiological Sciences, National Institutes of Natural Sciences (NINS), Okazaki, 444-8787, Japan; Exploratory Research Center on Life and Living Systems, NINS, Okazaki, 444-8787, Japan; SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8787, Japan
| | - Akiyuki Nishimura
- National Institute for Physiological Sciences, National Institutes of Natural Sciences (NINS), Okazaki, 444-8787, Japan; Exploratory Research Center on Life and Living Systems, NINS, Okazaki, 444-8787, Japan; SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8787, Japan
| | - Keitaro Umezawa
- Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| | - Yuri Kato
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Xinya Mi
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Tomoya Ito
- National Institute for Physiological Sciences, National Institutes of Natural Sciences (NINS), Okazaki, 444-8787, Japan; Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Yasuteru Urano
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan; Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Takaaki Akaike
- Graduate School of Medicine, Tohoku University, Sendai, 980-8575, Japan
| | - Motohiro Nishida
- National Institute for Physiological Sciences, National Institutes of Natural Sciences (NINS), Okazaki, 444-8787, Japan; Exploratory Research Center on Life and Living Systems, NINS, Okazaki, 444-8787, Japan; SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8787, Japan; Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| |
Collapse
|
14
|
Kuprytė M, Lesauskaitė V, Siratavičiūtė V, Utkienė L, Jusienė L, Pangonytė D. Expression of Osteopontin and Gremlin 1 Proteins in Cardiomyocytes in Ischemic Heart Failure. Int J Mol Sci 2024; 25:8240. [PMID: 39125809 PMCID: PMC11311846 DOI: 10.3390/ijms25158240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/12/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
A relevant role of osteopontin (OPN) and gremlin 1 (Grem1) in regulating cardiac tissue remodeling and formation of heart failure (HF) are documented, with the changes of OPN and Grem1 levels in blood plasma due to acute ischemia, ischemic heart disease-induced advanced HF or dilatative cardiomyopathy being the primary focus in most of these studies. However, knowledge on the early OPN and Grem1 proteins expression changes within cardiomyocytes during remodeling due to chronic ischemia remains insufficient. The aim of this study was to determine the OPN and Grem1 proteins expression changes in human cardiomyocytes at different stages of ischemic HF. A semi-quantitative immunohistochemical analysis was performed in 105 myocardial tissue samples obtained from the left cardiac ventricles. Increased OPN immunostaining intensity was already detected in the stage A HF group, compared to the control group (p < 0.001), and continued to increase in the stage B HF (p < 0.001), achieving the peak of immunostaining in the stages C/D HF group (p < 0.001). Similar data of Grem1 immunostaining intensity changes in cardiomyocytes were documented. Significantly positive correlations were detected between OPN, Grem1 expression in cardiomyocytes and their diameter as well as the length, in addition to positive correlation between OPN and Grem1 expression changes within cardiomyocytes. These novel findings suggest that OPN and Grem1 contribute significantly to reorganization of cellular geometry from the earliest stage of cardiomyocyte remodeling, providing new insights into the ischemic HF pathogenesis.
Collapse
Affiliation(s)
- Milda Kuprytė
- Laboratory of Cardiac Pathology, Institute of Cardiology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (M.K.); (V.S.); (L.U.); (L.J.)
| | - Vaiva Lesauskaitė
- Laboratory of Molecular Cardiology, Institute of Cardiology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania;
| | - Vitalija Siratavičiūtė
- Laboratory of Cardiac Pathology, Institute of Cardiology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (M.K.); (V.S.); (L.U.); (L.J.)
| | - Lina Utkienė
- Laboratory of Cardiac Pathology, Institute of Cardiology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (M.K.); (V.S.); (L.U.); (L.J.)
| | - Lina Jusienė
- Laboratory of Cardiac Pathology, Institute of Cardiology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (M.K.); (V.S.); (L.U.); (L.J.)
| | - Dalia Pangonytė
- Laboratory of Cardiac Pathology, Institute of Cardiology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (M.K.); (V.S.); (L.U.); (L.J.)
| |
Collapse
|
15
|
Sillanmäki S, Hartikainen S, Ylä-Herttuala E. Review of Myocardial Ischemia, Scar, and Viability Estimation with Molecular Magnetic Resonance Imaging. Biomedicines 2024; 12:1681. [PMID: 39200146 PMCID: PMC11351116 DOI: 10.3390/biomedicines12081681] [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: 06/12/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Cardiovascular diseases, particularly myocardial ischemia from coronary artery obstruction, remain a leading cause of global morbidity. This review explores cardiac molecular magnetic resonance imaging (mMRI) and other molecular imaging techniques for the evaluation of myocardial ischemia, scarring, and viability. RESULTS AND FINDINGS mMRI imaging methods provide detailed information on myocardial ischemia, edema, and scar tissue using techniques like cine imaging, T1 and T2 mapping, and gadolinium-based contrast agents. These methods enable the precise assessment of the myocardial tissue properties, crucial in diagnosing and treating cardiovascular diseases. Advanced techniques, such as the T1ρ and RAFFn methods, might provide enhanced contrast and sensitivity for the detection of myocardial scarring without contrast agents. Molecular probes, including gadolinium-based and protein-targeted contrast agents, improve the detection of molecular changes, facilitating early diagnosis and personalized treatment. Integrating MRI with positron emission tomography (PET) combines the high spatial and temporal resolution with molecular and functional imaging. CONCLUSION Recent advancements in mMRI and molecular imaging have changed the evaluation of myocardial ischemia, scarring, and viability. Despite significant progress, extensive research is needed to validate these techniques clinically and further develop imaging methods for better diagnostic and prognostic outcomes.
Collapse
Affiliation(s)
- Saara Sillanmäki
- Institute of Clinical Medicine, University of Eastern Finland, 70029 Kuopio, Finland
- Diagnostic Imaging Center, Kuopio University Hospital, 70200 Kuopio, Finland
| | - Suvi Hartikainen
- Institute of Clinical Medicine, University of Eastern Finland, 70029 Kuopio, Finland
- Diagnostic Imaging Center, Kuopio University Hospital, 70200 Kuopio, Finland
| | - Elias Ylä-Herttuala
- Diagnostic Imaging Center, Kuopio University Hospital, 70200 Kuopio, Finland
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| |
Collapse
|
16
|
Dhalla NS, Mota KO, Elimban V, Shah AK, de Vasconcelos CML, Bhullar SK. Role of Vasoactive Hormone-Induced Signal Transduction in Cardiac Hypertrophy and Heart Failure. Cells 2024; 13:856. [PMID: 38786079 PMCID: PMC11119949 DOI: 10.3390/cells13100856] [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: 03/25/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
Heart failure is the common concluding pathway for a majority of cardiovascular diseases and is associated with cardiac dysfunction. Since heart failure is invariably preceded by adaptive or maladaptive cardiac hypertrophy, several biochemical mechanisms have been proposed to explain the development of cardiac hypertrophy and progression to heart failure. One of these includes the activation of different neuroendocrine systems for elevating the circulating levels of different vasoactive hormones such as catecholamines, angiotensin II, vasopressin, serotonin and endothelins. All these hormones are released in the circulation and stimulate different signal transduction systems by acting on their respective receptors on the cell membrane to promote protein synthesis in cardiomyocytes and induce cardiac hypertrophy. The elevated levels of these vasoactive hormones induce hemodynamic overload, increase ventricular wall tension, increase protein synthesis and the occurrence of cardiac remodeling. In addition, there occurs an increase in proinflammatory cytokines and collagen synthesis for the induction of myocardial fibrosis and the transition of adaptive to maladaptive hypertrophy. The prolonged exposure of the hypertrophied heart to these vasoactive hormones has been reported to result in the oxidation of catecholamines and serotonin via monoamine oxidase as well as the activation of NADPH oxidase via angiotensin II and endothelins to promote oxidative stress. The development of oxidative stress produces subcellular defects, Ca2+-handling abnormalities, mitochondrial Ca2+-overload and cardiac dysfunction by activating different proteases and depressing cardiac gene expression, in addition to destabilizing the extracellular matrix upon activating some metalloproteinases. These observations support the view that elevated levels of various vasoactive hormones, by producing hemodynamic overload and activating their respective receptor-mediated signal transduction mechanisms, induce cardiac hypertrophy. Furthermore, the occurrence of oxidative stress due to the prolonged exposure of the hypertrophied heart to these hormones plays a critical role in the progression of heart failure.
Collapse
Affiliation(s)
- Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R2H 2A6, Canada; (V.E.); (S.K.B.)
| | - Karina O. Mota
- Department of Physiology, Center of Biological and Health Sciences, Federal University of Sergipe, Sao Cristóvao 49100-000, Brazil; (K.O.M.); (C.M.L.d.V.)
| | - Vijayan Elimban
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R2H 2A6, Canada; (V.E.); (S.K.B.)
| | - Anureet K. Shah
- Department of Nutrition and Food Science, California State University, Los Angeles, CA 90032-8162, USA;
| | - Carla M. L. de Vasconcelos
- Department of Physiology, Center of Biological and Health Sciences, Federal University of Sergipe, Sao Cristóvao 49100-000, Brazil; (K.O.M.); (C.M.L.d.V.)
| | - Sukhwinder K. Bhullar
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R2H 2A6, Canada; (V.E.); (S.K.B.)
| |
Collapse
|
17
|
Lorusso R, Matteucci M, Lerakis S, Ronco D, Menicanti L, Sharma SK, Moreno PR. Postmyocardial Infarction Ventricular Aneurysm: JACC Focus Seminar 5/5. J Am Coll Cardiol 2024; 83:1917-1935. [PMID: 38719371 DOI: 10.1016/j.jacc.2024.02.044] [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: 11/28/2023] [Revised: 01/18/2024] [Accepted: 02/09/2024] [Indexed: 07/16/2024]
Abstract
Ventricular aneurysm represents a rare complication of transmural acute myocardial infarction, although other cardiac, congenital, or metabolic diseases may also predispose to such condition. Ventricular expansion includes all the cardiac layers, usually with a large segment involved. Adverse events include recurrent angina, reduced ventricular stroke volume with congestive heart failure, mitral regurgitation, thromboembolism, and ventricular arrhythmias. Multimodality imaging is paramount to provide comprehensive assessment, allowing for appropriate therapeutic decision-making. When indicated, surgical intervention remains the gold standard, although additional therapy (heart failure, anticoagulation, and advanced antiarrhythmic treatment) might be required. However, the STICH (Surgical Treatment for Ischemic Heart Failure) trial did not show any advantage from adding surgical ventricular reconstruction to coronary artery bypass surgery in terms of survival, rehospitalization or symptoms, compared with revascularization alone. Finally, implantable cardiac defibrillator may reduce the risk of fatal arrhythmias.
Collapse
Affiliation(s)
- Roberto Lorusso
- Cardio-Thoracic Surgery Department, Maastricht University Medical Centre (MUMC), Maastricht, the Netherlands; Cardiovascular Research Institute Maastricht (CARIM), Maastricht, the Netherlands
| | - Matteo Matteucci
- Cardio-Thoracic Surgery Department, Maastricht University Medical Centre (MUMC), Maastricht, the Netherlands; Cardiovascular Research Institute Maastricht (CARIM), Maastricht, the Netherlands; Cardiac Surgery Unit, ASSTSette Laghi, Varese, Italy
| | - Stamatios Lerakis
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Daniele Ronco
- Cardio-Thoracic Surgery Department, Maastricht University Medical Centre (MUMC), Maastricht, the Netherlands; Cardiovascular Research Institute Maastricht (CARIM), Maastricht, the Netherlands; Cardiac Surgery Unit, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | | | - Samin K Sharma
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Pedro R Moreno
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Igor Palacios Fellows Foundation, Boston Massachusetts, USA.
| |
Collapse
|
18
|
Horitani K, Shiojima I. Wnt signaling in cardiac development and heart diseases. In Vitro Cell Dev Biol Anim 2024; 60:482-488. [PMID: 38709417 PMCID: PMC11126472 DOI: 10.1007/s11626-024-00917-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: 01/03/2024] [Accepted: 04/22/2024] [Indexed: 05/07/2024]
Abstract
The Wnt signaling pathway is a fundamental cellular communication system with extensive implications in various organs including the heart. In cardiac homeostasis, it governs essential processes like cellular proliferation, differentiation, and apoptosis, ensuring the heart's structural and functional integrity from embryonic stages and throughout life. Both canonical and non-canonical Wnt signaling pathways play a critical role during embryonic heart development in a region- and stage-specific manner. Canonical Wnt signaling also plays a significant role in heart diseases such as myocardial infarction and heart failure. However, the role of non-canonical Wnt signaling in heart diseases has not been fully elucidated. Wnt5a is a major ligand that activates non-canonical Wnt pathway, and recent studies start to clarify the role of the Wnt5a signaling axis in cardiac health and disease. In this review, we will briefly summarize the previous findings on the role of Wnt signaling pathways in heart development and diseases, and then focus on the role of Wnt5a signaling in heart failure progression. The multifaceted roles of the Wnt signaling pathway highlight its therapeutic potential for various types of heart diseases.
Collapse
Affiliation(s)
- Keita Horitani
- Department of Medicine II, Kansai Medical University, 2-5-1, Shin-Machi, Hirakata, Osaka, 573-1010, Japan
| | - Ichiro Shiojima
- Department of Medicine II, Kansai Medical University, 2-5-1, Shin-Machi, Hirakata, Osaka, 573-1010, Japan.
| |
Collapse
|
19
|
Shpigelman J, Blaine C, Nugent CA, Kiernan L, Cahir C, Curtain BM, Bachari A, Irfan W, O'Boyle P, O'Neill J, Daly M. Electrocardiographic predictors of response to sacubitril/valsartan therapy in heart failure with reduced ejection fraction. J Electrocardiol 2024; 84:104-108. [PMID: 38615617 DOI: 10.1016/j.jelectrocard.2024.03.008] [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: 09/18/2023] [Revised: 12/06/2023] [Accepted: 03/19/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND Sacubitril/valsartan (SV) is currently recommended as a first-line therapy in patients with heart failure and reduced ejection fraction (HFrEF) due to its significant clinical and prognostic benefit; however, not all patients respond to therapy and predictors of clinical response to SV remain under-studied. AIMS To identify electrocardiographic (ECG) predictors of response to SV therapy in HFrEF patients. METHODS A retrospective analysis of a hospital heart failure registry was undertaken. Consecutive HFrEF patients (New York Heart Association class II-III) on maximal-dose SV were studied. Response to SV was defined as ≥10% relative improvement in left ventricular ejection fraction (LVEF) at 3-months post-maximal-dose therapy. Pre-therapy ECGs were retrospectively analyzed for axes and standard wave and interval durations. Logistic regression was used to estimate odds ratios and 95% confidence intervals for associations between predictors and therapeutic response. Backward stepwise regression was employed to develop a parsimonious model. RESULTS P-wave duration (PWD) 100-120 ms, PWD >120 ms, and QTc >460 ms were associated with response to SV on univariate analysis: OR 18.00 (4.45-122.90), 5.00 (1.47-20.42), and 3.10 (1.18-9.22), respectively. The preferred model that included the former two predictors in combination with pre-therapy creatinine, mineralocorticoid receptor antagonist use, and LVEF was highly selective (area under the ROC curve = 0.868). CONCLUSIONS Prolongation of both PWD and QTc interval on baseline ECG in HFrEF patients is predictive of therapeutic response to maximal-dose SV therapy and may indicate early cardiac remodeling that is highly amenable to reversal.
Collapse
Affiliation(s)
| | - Ciara Blaine
- Department of Cardiology, Connolly Hospital, Blanchardstown, Dublin, Ireland
| | - Carol-Ann Nugent
- Department of Cardiology, Connolly Hospital, Blanchardstown, Dublin, Ireland
| | - Louise Kiernan
- Department of Cardiology, Connolly Hospital, Blanchardstown, Dublin, Ireland
| | - Caitriona Cahir
- Data Science Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | - Amir Bachari
- School of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Wadeed Irfan
- School of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Patrick O'Boyle
- School of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Department of Cardiology, Connolly Hospital, Blanchardstown, Dublin, Ireland
| | - James O'Neill
- School of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Department of Cardiology, Connolly Hospital, Blanchardstown, Dublin, Ireland
| | - Michael Daly
- School of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Department of Cardiology, Connolly Hospital, Blanchardstown, Dublin, Ireland.
| |
Collapse
|
20
|
Luo ZR, Meng WT, Li H, Wang Y, Wang YC, Zhao Y, Lu PP, Yuan Y, Huang W, Guo HD. Transplantation of induced pluripotent stem cells-derived cardiomyocytes combined with modified Taohong Siwu decoction improved heart repair after myocardial infarction. Heliyon 2024; 10:e26700. [PMID: 38434034 PMCID: PMC10906439 DOI: 10.1016/j.heliyon.2024.e26700] [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: 10/29/2022] [Revised: 02/02/2024] [Accepted: 02/19/2024] [Indexed: 03/05/2024] Open
Abstract
Objective This study aimed to study whether modified Taohong Siwu decoction (MTHSWD) combined with human induced pluripotent stem cells-derived cardiomyocytes (iPS-CMs) transplantation can promote cardiac function in myocardial infarction (MI) nude mouse model and explore its possible mechanism. Methods The MI mouse model was established by the ligation of left anterior descending coronary artery. After 4 weeks of gavage of MTHSWD combined with iPS-CMs transplantation, the changes in heart function of mice were examined by echocardiography. The histological changes were observed by Masson's trichrome staining. The survival and differentiation of transplanted cells were detected by double immunofluorescence staining of human nuclear antigen (HNA) and cardiac troponin T (cTnT). The number of c-kit-positive cells in the infarct area were evaluated by immunofluorescent staining. The levels of stromal cell-derived factor 1 (SDF-1), stem cell factor (SCF), vascular endothelial growth factor (VEGF) and basic fibroblast growth factor in infarcted myocardium tissues were detected by ELISA. Results MTHSWD combined with iPS-CMs transplantation can improve the heart function of MI mice, reduce the infarct size and collagen deposition in infarct area. By immunofluorescence double-label detection of HNA and cTnT, it was found that MTHSWD combined with iPS-CMs transplantation can improve the survival and maturation of iPS-CMs. In addition, MTHSWD combined with iPS-CMs transplantation can activate more endogenous c-kit positive cardiac mesenchymal cells, and significantly increase the content of SDF-1, SCF and VEGF in myocardial tissues. Conclusions The combination of MTHSWD with iPS-CMs transplantation promoted cardiac function of nude mice with MI by improving the survival and maturation of iPS-CMs in the infarct area, activating the endogenous c-kit positive cardiac mesenchymal cells, and increasing paracrine.
Collapse
Affiliation(s)
- Zhi-rong Luo
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wan-ting Meng
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Han Li
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yu Wang
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ya-chao Wang
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yue Zhao
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ping-ping Lu
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yuan Yuan
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wei Huang
- Department of Chinese Internal Medicine, Dahua Hospital, Xuhui District, Shanghai, China
| | - Hai-dong Guo
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
21
|
Choi HM, Hwang IC, Choi HJ, Yoon YE, Lee HJ, Park JB, Lee SP, Kim HK, Kim YJ, Cho GY. Irreversible myocardial injury attenuates the benefits of sacubitril/valsartan in heart failure patients. Int J Cardiol 2024; 397:131611. [PMID: 38030041 DOI: 10.1016/j.ijcard.2023.131611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 11/08/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Despite the established benefits of angiotensin receptor-neprilysin inhibitor (ARNI) in heart failure with reduced ejection fraction (HFrEF) across various etiologies, there are controversies regarding the effects of ARNI in patients with irreversible myocardial injury. The aim of this study is to investigate the impact of irreversible myocardial injury on the benefits of ARNI treatment in patients with HFrEF, consisted of both ischemic and non-ischemic etiologies. METHODS AND RESULTS We conducted a retrospective single-center study including 409 consecutive patients with HFrEF treated with ARNI between March 2017 and May 2020. Irreversible myocardial injury was defined as nonviable myocardium without contractile reserve, which suggests a limited potential for recovery of left ventricular function and geometry. At baseline, irreversible myocardial injury was observed in 129 (31.5%) patients. Composite outcome was cardiovascular death or hospitalization for heart failure, which occurred in 56 (43.4%) and 61 (21.8%) patients with and without irreversible myocardial injury, respectively. On multivariable analysis, irreversible injury presence, but not ischemic etiology, was an independent predictor of composite outcome (hazard ratio 2.16, 95% confidence interval 1.33-3.49). Mediation analysis revealed that the increased risk of the composite outcome due to irreversible myocardial injury was mediated by attenuated LV reverse remodeling (Z value = 2.02, P = 0.043). CONCLUSIONS The presence of irreversible myocardial injury was significantly associated with the response to ARNI treatment in patients with HFrEF, regardless of etiology.
Collapse
Affiliation(s)
- Hong-Mi Choi
- Department of Cardiology, Cardiovascular Center, Seoul National University Bundang Hospital, 82 Gumi-ro-173-gil, Bundang, Seongnam, Gyeonggi 13620, Republic of Korea; Department of Internal Medicine, Seoul National University College of Medicine, 103 Daehak-ro Jongno-gu, Seoul 03080, Republic of Korea
| | - In-Chang Hwang
- Department of Cardiology, Cardiovascular Center, Seoul National University Bundang Hospital, 82 Gumi-ro-173-gil, Bundang, Seongnam, Gyeonggi 13620, Republic of Korea; Department of Internal Medicine, Seoul National University College of Medicine, 103 Daehak-ro Jongno-gu, Seoul 03080, Republic of Korea.
| | - Hye Jung Choi
- Department of Cardiology, Cardiovascular Center, Seoul National University Bundang Hospital, 82 Gumi-ro-173-gil, Bundang, Seongnam, Gyeonggi 13620, Republic of Korea; Department of Internal Medicine, Seoul National University College of Medicine, 103 Daehak-ro Jongno-gu, Seoul 03080, Republic of Korea
| | - Yeonyee E Yoon
- Department of Cardiology, Cardiovascular Center, Seoul National University Bundang Hospital, 82 Gumi-ro-173-gil, Bundang, Seongnam, Gyeonggi 13620, Republic of Korea; Department of Internal Medicine, Seoul National University College of Medicine, 103 Daehak-ro Jongno-gu, Seoul 03080, Republic of Korea
| | - Hyun-Jung Lee
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jun-Bean Park
- Department of Internal Medicine, Seoul National University College of Medicine, 103 Daehak-ro Jongno-gu, Seoul 03080, Republic of Korea; Cardiovascular Center and Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro Jongno-gu, Seoul 03080, Republic of Korea
| | - Seung-Pyo Lee
- Department of Internal Medicine, Seoul National University College of Medicine, 103 Daehak-ro Jongno-gu, Seoul 03080, Republic of Korea; Cardiovascular Center and Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro Jongno-gu, Seoul 03080, Republic of Korea
| | - Hyung-Kwan Kim
- Department of Internal Medicine, Seoul National University College of Medicine, 103 Daehak-ro Jongno-gu, Seoul 03080, Republic of Korea; Cardiovascular Center and Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro Jongno-gu, Seoul 03080, Republic of Korea
| | - Yong-Jin Kim
- Department of Internal Medicine, Seoul National University College of Medicine, 103 Daehak-ro Jongno-gu, Seoul 03080, Republic of Korea; Cardiovascular Center and Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro Jongno-gu, Seoul 03080, Republic of Korea
| | - Goo-Yeong Cho
- Department of Cardiology, Cardiovascular Center, Seoul National University Bundang Hospital, 82 Gumi-ro-173-gil, Bundang, Seongnam, Gyeonggi 13620, Republic of Korea; Department of Internal Medicine, Seoul National University College of Medicine, 103 Daehak-ro Jongno-gu, Seoul 03080, Republic of Korea
| |
Collapse
|
22
|
Labbé P, Martel C, Shi YF, Montezano A, He Y, Gillis MA, Higgins MÈ, Villeneuve L, Touyz R, Tardif JC, Thorin-Trescases N, Thorin E. Knockdown of ANGPTL2 promotes left ventricular systolic dysfunction by upregulation of NOX4 in mice. Front Physiol 2024; 15:1320065. [PMID: 38426206 PMCID: PMC10902461 DOI: 10.3389/fphys.2024.1320065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
Background: Angiopoietin-like 2 (ANGPTL2) is a pro-inflammatory and pro-oxidant circulating protein that predicts and promotes chronic inflammatory diseases such as atherosclerosis in humans. Transgenic murine models demonstrated the deleterious role of ANGPTL2 in vascular diseases, while deletion of ANGPTL2 was protective. The nature of its role in cardiac tissues is, however, less clear. Indeed, in adult mice knocked down (KD) for ANGPTL2, we recently reported a mild left ventricular (LV) dysfunction originating from a congenital aortic valve stenosis, demonstrating that ANGPTL2 is essential to cardiac development and function. Hypothesis: Because we originally demonstrated that the KD of ANGPTL2 protected vascular endothelial function via an upregulation of arterial NOX4, promoting the beneficial production of dilatory H2O2, we tested the hypothesis that increased cardiac NOX4 could negatively affect cardiac redox and remodeling and contribute to LV dysfunction observed in adult Angptl2-KD mice. Methods and results: Cardiac expression and activity of NOX4 were higher in KD mice, promoting higher levels of cardiac H2O2 when compared to wild-type (WT) mice. Immunofluorescence showed that ANGPTL2 and NOX4 were co-expressed in cardiac cells from WT mice and both proteins co-immunoprecipitated in HEK293 cells, suggesting that ANGPTL2 and NOX4 physically interact. Pressure overload induced by transverse aortic constriction surgery (TAC) promoted LV systolic dysfunction in WT mice but did not further exacerbate the dysfunction in KD mice. Importantly, the severity of LV systolic dysfunction in KD mice (TAC and control SHAM) correlated with cardiac Nox4 expression. Injection of an adeno-associated virus (AAV9) delivering shRNA targeting cardiac Nox4 expression fully reversed LV systolic dysfunction in KD-SHAM mice, demonstrating the causal role of NOX4 in cardiac dysfunction in KD mice. Targeting cardiac Nox4 expression in KD mice also induced an antioxidant response characterized by increased expression of NRF2/KEAP1 and catalase. Conclusion: Together, these data reveal that the absence of ANGPTL2 induces an upregulation of cardiac NOX4 that contributes to oxidative stress and LV dysfunction. By interacting and repressing cardiac NOX4, ANGPTL2 could play a new beneficial role in the maintenance of cardiac redox homeostasis and function.
Collapse
Affiliation(s)
- Pauline Labbé
- Montreal Heart Institute, Research Center, Montreal, QC, Canada
- Department of Pharmacology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Cécile Martel
- Montreal Heart Institute, Research Center, Montreal, QC, Canada
- Department of Pharmacology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Yan-Fen Shi
- Montreal Heart Institute, Research Center, Montreal, QC, Canada
| | - Augusto Montezano
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Ying He
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | | | | | | | - Rhian Touyz
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Jean-Claude Tardif
- Montreal Heart Institute, Research Center, Montreal, QC, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | | | - Eric Thorin
- Montreal Heart Institute, Research Center, Montreal, QC, Canada
- Department of Pharmacology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Surgery, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
23
|
Han W, Wang W, Wang Q, Maduray K, Hao L, Zhong J. A review on regulation of DNA methylation during post-myocardial infarction. Front Pharmacol 2024; 15:1267585. [PMID: 38414735 PMCID: PMC10896928 DOI: 10.3389/fphar.2024.1267585] [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: 07/26/2023] [Accepted: 01/25/2024] [Indexed: 02/29/2024] Open
Abstract
Myocardial infarction (MI) imposes a huge medical and economic burden on society, and cardiac repair after MI involves a complex series of processes. Understanding the key mechanisms (such as apoptosis, autophagy, inflammation, and fibrosis) will facilitate further drug development and patient treatment. Presently, a substantial body of evidence suggests that the regulation of epigenetic processes contributes to cardiac repair following MI, with DNA methylation being among the notable epigenetic factors involved. This article will review the research on the mechanism of DNA methylation regulation after MI to provide some insights for future research and development of related drugs.
Collapse
Affiliation(s)
- Wenqiang Han
- 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 Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wenxin 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 Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Qinhong 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 Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Kellina Maduray
- 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 Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Li Hao
- Department of Gerontology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Jingquan 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 Cardiology, Qilu Hospital of Shandong University, Jinan, China
- Department of Cardiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| |
Collapse
|
24
|
Stanley A, Athanasuleas C. Timing of Surgery for Asymptomatic Primary Mitral Regurgitation: Possible Value of Early, Serial Measurements of Left Ventricular Sphericity. Curr Cardiol Rev 2024; 20:93-101. [PMID: 38351687 PMCID: PMC11107465 DOI: 10.2174/011573403x277223240206062319] [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: 11/10/2023] [Revised: 01/11/2024] [Accepted: 01/24/2024] [Indexed: 04/30/2024] Open
Abstract
Asymptomatic primary mitral regurgitation due to myxomatous degeneration of the mitral valve leaflets may remain so for long periods, even as left ventricular function progresses to a decompensated stage. During the early compensated stage, the ventricle's initial response to the volume overload is an asymmetric increase in the diastolic short axis dimension, accomplished by a diastolic shift of the interventricular septum into the right ventricular cavity, creating a more spherical left ventricular diastolic shape, increasing diastolic filling and stroke volume. Early valve repair is recommended to reduce postoperative left ventricular dysfunction. Early serial measurements of left ventricular sphericity index [LV-Si]. during the compensated stage of mitral regurgitation might identify subtle changes in left ventricular shape and assist in determining the optimal earliest timing for surgical intervention.
Collapse
Affiliation(s)
- Alfred Stanley
- Cardiovascular Associates of the Southeast, Birmingham AL and Kemp-Carraway Heart Institute, Birmingham, AL, USA
| | - Constantine Athanasuleas
- Department of Surgery, North Alabama Medical Center and Kemp-Carraway Heart Institute, Birmingham, AL, USA
| |
Collapse
|
25
|
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
|
26
|
Zhao Y, Xiong W, Li C, Zhao R, Lu H, Song S, Zhou Y, Hu Y, Shi B, Ge J. Hypoxia-induced signaling in the cardiovascular system: pathogenesis and therapeutic targets. Signal Transduct Target Ther 2023; 8:431. [PMID: 37981648 PMCID: PMC10658171 DOI: 10.1038/s41392-023-01652-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/10/2023] [Accepted: 09/13/2023] [Indexed: 11/21/2023] Open
Abstract
Hypoxia, characterized by reduced oxygen concentration, is a significant stressor that affects the survival of aerobic species and plays a prominent role in cardiovascular diseases. From the research history and milestone events related to hypoxia in cardiovascular development and diseases, The "hypoxia-inducible factors (HIFs) switch" can be observed from both temporal and spatial perspectives, encompassing the occurrence and progression of hypoxia (gradual decline in oxygen concentration), the acute and chronic manifestations of hypoxia, and the geographical characteristics of hypoxia (natural selection at high altitudes). Furthermore, hypoxia signaling pathways are associated with natural rhythms, such as diurnal and hibernation processes. In addition to innate factors and natural selection, it has been found that epigenetics, as a postnatal factor, profoundly influences the hypoxic response and progression within the cardiovascular system. Within this intricate process, interactions between different tissues and organs within the cardiovascular system and other systems in the context of hypoxia signaling pathways have been established. Thus, it is the time to summarize and to construct a multi-level regulatory framework of hypoxia signaling and mechanisms in cardiovascular diseases for developing more therapeutic targets and make reasonable advancements in clinical research, including FDA-approved drugs and ongoing clinical trials, to guide future clinical practice in the field of hypoxia signaling in cardiovascular diseases.
Collapse
Affiliation(s)
- Yongchao Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Weidong Xiong
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, 200032, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, 200032, China
| | - Chaofu Li
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Ranzun Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - Hao Lu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Shuai Song
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - You Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Yiqing Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| | - Bei Shi
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
| | - Junbo Ge
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, 200032, China.
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
27
|
Arnautu DA, Gheorghiu A, Arnautu SF, Tomescu MC, Malita CD, Banciu C, Vacarescu C, Ionac I, Luca S, Cozma D, Mornos C, Gaita D, Luca CT. Subtle Changes in Myocardial Work Indices Assessed by 2D-Speckle Tracking Echocardiography Are Linked with Pathological LV Remodeling and MACEs Following an Acute Myocardial Infarction Treated by Primary Percutaneous Coronary Intervention. Diagnostics (Basel) 2023; 13:3108. [PMID: 37835851 PMCID: PMC10572832 DOI: 10.3390/diagnostics13193108] [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: 07/22/2023] [Revised: 09/12/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
The goal of this study was to assess whether subtle changes in myocardial work indices may predict left ventricular (LV) remodeling and major cardiac events (MACEs) in patients with a first ST-elevation acute myocardial infarction (STEMI) and preserved LVEF after successful myocardial revascularization with PCI. Methods. Consecutive STEMI patients in sinus rhythm and with an LV ejection fraction ≥ 50% following a successful PCI were recruited. Conventional and two-dimensional speckle tracking echocardiography (2D-STE) was conducted within 36 h of the PCI and 3 months later. Patients having an increase of more than 20% in LV diastolic volume were included in the LV remodeling group. MACEs were noted throughout a four-year period of follow-up. Results: The study comprised 246 STEMI patients with a mean age of 66; 72% of whom were men. In 24% (58) of the patients, LV remodeling developed. These patients were older, more frequently hypertensive, and had a smoking history. They also exhibited significantly lower baseline and 3-month values for the myocardial global index (GWI), global constructive work (GCW), and global myocardial efficiency (GWE). The cut-off values of 1670 mmHg% for GWI and 83% for GWE were predictive of LV remodeling (p < 0.0001). During the four-year follow-up period, 19% of STEMI patients experienced a MACE, involving 15% from non-LV remodelers and 34% from LV remodelers (p = 0.01). The cut-off values for baseline GWI of 1680 mmHg% and baseline GWE of 84% had the best accuracy in predicting MACEs. In conclusion, non-invasive myocardial work indices offered a reproducible and accurate method to predict post-MI LV remodeling and MACEs.
Collapse
Affiliation(s)
- Diana-Aurora Arnautu
- Multidisciplinary Heart Research Center, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (D.-A.A.); (A.G.); (M.-C.T.)
- Department of Internal Medicine, ”Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (S.-F.A.); (C.B.)
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania; (C.V.); (I.I.); (S.L.); (D.C.); (C.M.); (D.G.); (C.-T.L.)
| | - Alexandru Gheorghiu
- Multidisciplinary Heart Research Center, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (D.-A.A.); (A.G.); (M.-C.T.)
- Timisoara Municipal Clinical Emergency Hospital, 12 Revolution of 1989 Bd., 300040 Timisoara, Romania
| | - Sergiu-Florin Arnautu
- Department of Internal Medicine, ”Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (S.-F.A.); (C.B.)
- Timisoara Municipal Clinical Emergency Hospital, 12 Revolution of 1989 Bd., 300040 Timisoara, Romania
| | - Mirela-Cleopatra Tomescu
- Multidisciplinary Heart Research Center, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (D.-A.A.); (A.G.); (M.-C.T.)
- Department of Internal Medicine, ”Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (S.-F.A.); (C.B.)
- Timisoara Municipal Clinical Emergency Hospital, 12 Revolution of 1989 Bd., 300040 Timisoara, Romania
| | - Claudiu-Daniel Malita
- Timisoara Municipal Clinical Emergency Hospital, 12 Revolution of 1989 Bd., 300040 Timisoara, Romania
- Department of Radiology and Medical Imaging, ”Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Christian Banciu
- Department of Internal Medicine, ”Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (S.-F.A.); (C.B.)
| | - Cristina Vacarescu
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania; (C.V.); (I.I.); (S.L.); (D.C.); (C.M.); (D.G.); (C.-T.L.)
- Department of Cardiology, ”Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Ioana Ionac
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania; (C.V.); (I.I.); (S.L.); (D.C.); (C.M.); (D.G.); (C.-T.L.)
- Department of Cardiology, ”Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Silvia Luca
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania; (C.V.); (I.I.); (S.L.); (D.C.); (C.M.); (D.G.); (C.-T.L.)
- Department of Cardiology, ”Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Dragos Cozma
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania; (C.V.); (I.I.); (S.L.); (D.C.); (C.M.); (D.G.); (C.-T.L.)
- Department of Cardiology, ”Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Cristian Mornos
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania; (C.V.); (I.I.); (S.L.); (D.C.); (C.M.); (D.G.); (C.-T.L.)
- Department of Cardiology, ”Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Dan Gaita
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania; (C.V.); (I.I.); (S.L.); (D.C.); (C.M.); (D.G.); (C.-T.L.)
- Department of Cardiology, ”Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Constantin-Tudor Luca
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania; (C.V.); (I.I.); (S.L.); (D.C.); (C.M.); (D.G.); (C.-T.L.)
- Department of Cardiology, ”Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| |
Collapse
|
28
|
Ribeiro Vitorino T, Ferraz do Prado A, Bruno de Assis Cau S, Rizzi E. MMP-2 and its implications on cardiac function and structure: Interplay with inflammation in hypertension. Biochem Pharmacol 2023; 215:115684. [PMID: 37459959 DOI: 10.1016/j.bcp.2023.115684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 08/01/2023]
Abstract
Hypertension is one of the leading risk factors for the development of heart failure. Despite being a multifactorial disease, in recent years, preclinical and clinical studies suggest strong evidence of the pivotal role of inflammatory cells and cytokines in the remodeling process and cardiac dysfunction. During the heart remodeling, activation of extracellular matrix metalloproteinases (MMPs) occurs, with MMP-2 being one of the main proteases secreted by cardiomyocytes, fibroblasts, endothelial and inflammatory cells in cardiac tissue. In this review, we will address the process of cardiac remodeling and injury induced by the increase in MMP-2 and the main signaling pathways involving cytokines and inflammatory cells in the process of transcriptional, secretion and activation of MMP-2. In addition, an interaction and coordinated action between MMP-2 and inflammation are explored and significant in maintaining the cardiac cycle. These observations suggest that new therapeutic opportunities targeting MMP-2 could be used to reduce inflammatory biomarkers and reduce cardiac damage in hypertension.
Collapse
Affiliation(s)
- Thaís Ribeiro Vitorino
- Unit of Biotechnology, University of Ribeirao Preto, UNAERP, Brazil; Department of Pharmacology, Faculty of Medical Sciences, University of Campinas, UNICAMP, Brazil
| | - Alejandro Ferraz do Prado
- Cardiovascular System Pharmacology and Toxicology, Institute of Biological Sciences, Federal University of Para, UFPA, Brazil
| | - Stefany Bruno de Assis Cau
- Department of Pharmacology, Institute of Biological Science, Federal University of Minas Gerais, UFMG, Brazil.
| | - Elen Rizzi
- Unit of Biotechnology, University of Ribeirao Preto, UNAERP, Brazil.
| |
Collapse
|
29
|
Saha S, Singh P, Dutta A, Vaidya H, Negi PC, Sengupta S, Seth S, Basak T. A Comprehensive Insight and Mechanistic Understanding of the Lipidomic Alterations Associated With DCM. JACC. ASIA 2023; 3:539-555. [PMID: 37614533 PMCID: PMC10442885 DOI: 10.1016/j.jacasi.2023.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/17/2023] [Accepted: 06/03/2023] [Indexed: 08/25/2023]
Abstract
Dilated cardiomyopathy (DCM) is one of the major causes of heart failure characterized by the enlargement of the left ventricular cavity and contractile dysfunction of the myocardium. Lipids are the major sources of energy for the myocardium. Impairment of lipid homeostasis has a potential role in the pathogenesis of DCM. In this review, we have summarized the role of different lipids in the progression of DCM that can be considered as potential biomarkers. Further, we have also explained the mechanistic pathways followed by the lipid molecules in disease progression along with the cardioprotective role of certain lipids. As the global epidemiological status of DCM is alarming, it is high time to define some disease-specific biomarkers with greater prognostic value. We are proposing an adaptation of a system lipidomics-based approach to profile DCM patients in order to achieve a better diagnosis and prognosis of the disease.
Collapse
Affiliation(s)
- Shubham Saha
- School of Biosciences and Bioengineering. IIT-Mandi, Mandi, India
- BioX Center, Indian Institute of Technology-Mandi, Mandi, India
| | - Praveen Singh
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Abhi Dutta
- School of Biosciences and Bioengineering. IIT-Mandi, Mandi, India
- BioX Center, Indian Institute of Technology-Mandi, Mandi, India
| | - Hiteshi Vaidya
- Department of Cardiology, Indira Gandhi Medical College & Hospital, Shimla, India
| | - Prakash Chand Negi
- Department of Cardiology, Indira Gandhi Medical College & Hospital, Shimla, India
| | - Shantanu Sengupta
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Sandeep Seth
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | - Trayambak Basak
- School of Biosciences and Bioengineering. IIT-Mandi, Mandi, India
- BioX Center, Indian Institute of Technology-Mandi, Mandi, India
| |
Collapse
|
30
|
Kishimoto H, Iwasaki M, Wada K, Horitani K, Tsukamoto O, Kamikubo K, Nomura S, Matsumoto S, Harada T, Motooka D, Okuzaki D, Takashima S, Komuro I, Kikuchi A, Shiojima I. Wnt5a-YAP signaling axis mediates mechanotransduction in cardiac myocytes and contributes to contractile dysfunction induced by pressure overload. iScience 2023; 26:107146. [PMID: 37456848 PMCID: PMC10338234 DOI: 10.1016/j.isci.2023.107146] [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: 09/07/2022] [Revised: 03/31/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Non-canonical Wnt signaling activated by Wnt5a/Wnt11 is required for the second heart field development in mice. However, the pathophysiological role of non-canonical Wnt signaling in the adult heart has not been fully elucidated. Here we show that cardiomyocyte-specific Wnt5a knockout mice exhibit improved systolic function and reduced expression of mechanosensitive genes including Nppb when subjected to pressure overload. In cultured cardiomyocytes, Wnt5a knockdown reduced Nppb upregulation induced by cyclic cell stretch. Upstream analysis revealed that TEAD1, a transcription factor that acts with Hippo pathway co-activator YAP, was downregulated both in vitro and in vivo by Wnt5a knockdown/knockout. YAP nuclear translocation was induced by cell stretch and attenuated by Wnt5a knockdown. Wnt5a knockdown-induced Nppb downregulation during cell stretch was rescued by Hippo inhibition, and the rescue effect was canceled by knockdown of YAP. These results collectively suggest that Wnt5a-YAP signaling axis mediates mechanotransduction in cardiomyocytes and contributes to heart failure progression.
Collapse
Affiliation(s)
- Hiroshi Kishimoto
- Department of Medicine II, Kansai Medical University, Osaka 573-1010, Japan
| | - Masayoshi Iwasaki
- Department of Medicine II, Kansai Medical University, Osaka 573-1010, Japan
| | - Kensaku Wada
- Department of Medicine II, Kansai Medical University, Osaka 573-1010, Japan
| | - Keita Horitani
- Department of Medicine II, Kansai Medical University, Osaka 573-1010, Japan
| | - Osamu Tsukamoto
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Kenta Kamikubo
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Seitaro Nomura
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Shinji Matsumoto
- Department of Molecular Biology and Biochemistry, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Takeshi Harada
- Department of Molecular Biology and Biochemistry, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Daisuke Motooka
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Akira Kikuchi
- Department of Molecular Biology and Biochemistry, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka 565-0871, Japan
| | - Ichiro Shiojima
- Department of Medicine II, Kansai Medical University, Osaka 573-1010, Japan
| |
Collapse
|
31
|
Grillo TG, Silveira CFDSMP, Quaglio AEV, Dutra RDM, Baima JP, Bazan SGZ, Sassaki LY. Acute heart failure as an adverse event of tumor necrosis factor inhibitor therapy in inflammatory bowel disease: A review of the literature. World J Cardiol 2023; 15:217-228. [PMID: 37274378 PMCID: PMC10237008 DOI: 10.4330/wjc.v15.i5.217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/09/2023] [Accepted: 04/12/2023] [Indexed: 05/19/2023] Open
Abstract
Tumor necrosis factor inhibitors (anti-TNFs) are widely used therapies for the treatment of inflammatory bowel diseases (IBD); however, their administration is not risk-free. Heart failure (HF), although rare, is a potential adverse event related to administration of these medications. However, the exact mechanism of development of HF remains obscure. TNFα is found in both healthy and damaged hearts. Its effects are concentration- and receptor-dependent, promoting either cardio-protection or cardiomyocyte apoptosis. Experimental rat models with TNFα receptor knockout showed increased survival rates, less reactive oxygen species formation, and improved diastolic left ventricle pressure. However, clinical trials employing anti-TNF therapy to treat HF had disappointing results, suggesting abolishment of the cardioprotective properties of TNFα, making cardiomyocytes susceptible to apoptosis and oxidation. Thus, patients with IBD who have risk factors should be screened for HF before initiating anti-TNF therapy. This review aims to discuss adverse events associated with the administration of anti-TNF therapy, with a focus on HF, and propose some approaches to avoid cardiac adverse events in patients with IBD.
Collapse
Affiliation(s)
- Thais Gagno Grillo
- Department of Internal Medicine, Medical School, São Paulo State University, Botucatu, Botucatu 18618686, Brazil
| | | | - Ana Elisa Valencise Quaglio
- Department of Biophysics and Pharmacology, Institute of Biosciences, São Paulo State University, Botucatu, Botucatu 18618689, Brazil
| | - Renata de Medeiros Dutra
- Department of Internal Medicine, Medical School, São Paulo State University, Botucatu, Botucatu 18618686, Brazil
| | - Julio Pinheiro Baima
- Department of Internal Medicine, Medical School, São Paulo State University, Botucatu, Botucatu 18618686, Brazil
| | - Silmeia Garcia Zanati Bazan
- Department of Internal Medicine, Medical School, São Paulo State University, Botucatu, Botucatu 18618686, Brazil
| | - Ligia Yukie Sassaki
- Department of Internal Medicine, Medical School, São Paulo State University, Botucatu, Botucatu 18618686, Brazil.
| |
Collapse
|
32
|
Yao Y, Lin L, Tang W, Shen Y, Chen F, Li N. Geniposide alleviates pressure overload in cardiac fibrosis with suppressed TGF-β1 pathway. Acta Histochem 2023; 125:152044. [PMID: 37196380 DOI: 10.1016/j.acthis.2023.152044] [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: 01/31/2023] [Revised: 04/26/2023] [Accepted: 05/08/2023] [Indexed: 05/19/2023]
Abstract
BACKGROUND Cardiac fibrosis is one of the main contributors to the pathogenesis of heart failure. Geniposide (GE), a major iridoid in gardenia fruit extract, has recently been reported to improve skeletal muscle fibrosis through the modulation of inflammation response. This investigation aimed to illuminate the cardio-protective effect and the potential mechanism of GE in cardiac fibrosis. MATERIAL AND METHODS A transverse aortic contraction (TAC) induction mice model was established and GE (0 mg/kg; 10 mg/kg; 20 mg/kg; 40 mg/kg) was administered by oral gavage daily for 4 weeks. Hemodynamic parameters, Masson's trichrome stain, and hematoxylin-eosin (HE) staining were estimated and cardiomyocyte fibrosis, interstitial collagen levels, and hypertrophic markers were analyzed using qPCR and western blot. In vitro, H9C2 cells were exposed to the Ang II (1 μM) pretreated with GE (0.1 μM, 1 μM, and 10 μM). Cardiomyocyte apoptosis was detected. Moreover, the transforming growth factor β1 (TGF-β1)/Smad2 pathway was assessed in vivo and in vitro. RESULTS GE significantly ameliorated TAC-induced cardiac hypertrophy, ventricular remodeling, myocardial fibrosis, and improved cardiac function in vivo, and it inhibited Ang II-induced cardiomyocyte apoptosis in vitro. We further observed that the inflammatory channel TGF-β1/Smad2 pathway was suppressed by GE both in vivo and in vitro. CONCLUSION These results indicate that GE inhibited myocardial fibrosis and improved hypertrophic cardiomyocytes with attenuated the TGF-β1/Smad2 pathway and proposed to be an important therapeutic of cardiac fibrosis reduced by TAC.
Collapse
Affiliation(s)
- Yanmei Yao
- Department of General Medicine, The Affiliated Hospital, Hangzhou Normal University, Hangzhou, Zhejiang 310015, People's Republic of China
| | - Leqing Lin
- Department of Critical Care Medicine, The Affiliated Hospital, Hangzhou Normal University, Hangzhou, Zhejiang 310015, People's Republic of China
| | - Wenxue Tang
- Department of Critical Care Medicine, The Affiliated Hospital, Hangzhou Normal University, Hangzhou, Zhejiang 310015, People's Republic of China
| | - Yueliang Shen
- Department of Pathophysiology, Zhejiang University Medical College, Hangzhou, Zhejiang 310000, People's Republic of China
| | - Fayu Chen
- Department of General Medicine, The Affiliated Hospital, Hangzhou Normal University, Hangzhou, Zhejiang 310015, People's Republic of China
| | - Ning Li
- Department of Hematology and Oncology, The Affiliated Hospital, Hangzhou Normal University, Hangzhou, Zhejiang 310015, People's Republic of China.
| |
Collapse
|
33
|
Castelvecchio S, Frigelli M, Sturla F, Milani V, Pappalardo OA, Citarella M, Menicanti L, Votta E. Elucidating the mechanisms underlying left ventricular function recovery in patients with ischemic heart failure undergoing surgical remodeling: A 3-dimensional ultrasound analysis. J Thorac Cardiovasc Surg 2023; 165:1418-1429.e4. [PMID: 33781593 DOI: 10.1016/j.jtcvs.2021.02.067] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 02/02/2021] [Accepted: 02/10/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The study objective was to elucidate the mechanisms of left ventricle functional recovery in terms of endocardial contractility and synchronicity after surgical ventricular reconstruction. METHODS Real-time 3-dimensional transthoracic echocardiography was performed on 20 patients with anterior left ventricle remodeling and ischemic heart failure before surgical ventricular reconstruction and at 6-month follow-up, and on 15 healthy controls matched by age and body surface area. Real-time 3-dimensional transthoracic echocardiography datasets were analyzed through TomTec software (4D LV-Analysis; TomTec Imaging Systems GmbH, Unterschleissheim, Germany): Left ventricle volumes, ejection fraction, and global longitudinal strain were computed; the time-dependent endocardial surface yielded by 3-dimensional speckle-tracking echocardiography was postprocessed through in-house software to quantify local systolic minimum principal strain as a measure of fiber shortening and mechanical dispersion as a measure of fiber synchronicity. RESULTS Compared with controls, patients with heart failure before surgical ventricular reconstruction showed lower ejection fraction (P < .0001) and significantly impaired mechanical dispersion (P < .0001) and minimum principal strain (P < .0001); the latter worsened progressively from left ventricle base to apex. After surgical ventricular reconstruction, global longitudinal strain improved from -6.7% to -11.3% (P < .0001); mechanical dispersion decreased in every left ventricle region (P ≤ .017) and mostly in the basal region, where computed mechanical dispersion values were comparable to physiologic values (P ≥ .046); minimum principal strain improved mostly in the basal region, changing from -16.6% to -22.3% (P = .0027). CONCLUSIONS At 6-month follow-up, surgical ventricular reconstruction was associated with significant recovery in global left ventricle function, improved mechanical dispersion indicating a more synchronous left ventricle contraction, and improved left ventricle fiber shortening mostly in the basal region, suggesting the major role of the remote myocardium in enhancing left ventricle functional recovery.
Collapse
Affiliation(s)
| | - Matteo Frigelli
- 3D and Computer Simulation Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Italy; Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Francesco Sturla
- 3D and Computer Simulation Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Italy; Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy.
| | - Valentina Milani
- Scientific Directorate, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Omar A Pappalardo
- 3D and Computer Simulation Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Michele Citarella
- Cardiac Surgery Department, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Lorenzo Menicanti
- Cardiac Surgery Department, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Emiliano Votta
- 3D and Computer Simulation Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Italy; Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| |
Collapse
|
34
|
Vasciaveo L, Zanzarelli E, D'Antonio F. Fetal cardiac function evaluation: A review. JOURNAL OF CLINICAL ULTRASOUND : JCU 2023; 51:215-224. [PMID: 36785505 DOI: 10.1002/jcu.23421] [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: 10/28/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 06/18/2023]
Abstract
The aim of this review is to provide an up to date on the current use of fetal echocardiography in assessing the fetal cardiac function and its potential research and clinical applications. Despite classically is been used for prenatal diagnosis of fetal heart defects, assessment of fetal cardiac function has been recently proposed as a fundamental tool to assess pregnancies complicated by several disorders with long-term impact on post-natal cardiovascular health, such as placental insufficiency and fetal growth restriction. In this review we present anatomical and functional fetal cardiac development mechanisms and an overview of the currently available techniques for evaluating fetal heart function.
Collapse
Affiliation(s)
- Lorenzo Vasciaveo
- Maternal Fetal Medicine Unit, Department of Obstetrics and Gynaecology, University of Foggia, Foggia, Italy
| | - Erika Zanzarelli
- Maternal Fetal Medicine Unit, Department of Obstetrics and Gynaecology, University of Foggia, Foggia, Italy
| | - Francesco D'Antonio
- Centre for High-Risk Pregnancy and Fetal Care, Department of Obstetrics and Gynecology, University of Chieti, Chieti, Italy
| |
Collapse
|
35
|
Dalal S, Shook PL, Singh M, Singh K. Post-ischemic cardioprotective potential of exogenous ubiquitin in myocardial remodeling late after ischemia/reperfusion injury. Life Sci 2023; 312:121216. [PMID: 36435225 PMCID: PMC9784153 DOI: 10.1016/j.lfs.2022.121216] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/10/2022] [Accepted: 11/18/2022] [Indexed: 11/27/2022]
Abstract
AIMS Pretreatment with ubiquitin (UB) associates with preservation of heart function 3 days post-ischemia/reperfusion (I/R) injury. This study investigated the cardioprotective potential of exogenous UB late after myocardial I/R injury. To enhance the clinical relevance, UB treatment was started at the time of reperfusion and continued for 28 days post-I/R. MAIN METHODS Mice underwent ligation of the left anterior descending coronary artery for 45 min. At the time of reperfusion, mice were treated with UB or saline which was continued until 28 days post-I/R. Heart function was measured at 3, 7, 14 and 28 days post-I/R using echocardiography. Biochemical parameters of the heart and serum cytokines/chemokines levels were measured 28 days post-I/R. KEY FINDINGS I/R decreased heart function and induced LV dilation at all time points post-I/R. However, I/R + UB exhibited improved heart function throughout the observation period, while LV dilation was lower in I/R + UB group at 3, 14 and 28 days post-I/R. I/R-mediated increase in myocardial fibrosis, hypertrophy and apoptosis were significantly lower in I/R + UB vs. I/R. Collagen-1α1 and MMP-2 expression was lower, while MMP-9 and TIMP-2 expression was higher in I/R + UB vs. I/R. MYH-7B (hypertrophy marker) expression was lower in I/R + UB vs. I/R. GSK3β activation was lower (vs. Sham), while activation of ERK1/2 (vs. I/R) and AKT (vs. Sham) was higher in I/R + UB. Serum levels of IL-6, G-CSF and IL-2 were lower in I/R + UB vs. I/R. SIGNIFICANCE Post-ischemic UB treatment improves heart function, and associates with decreased myocardial fibrosis, apoptosis, hypertrophy and serum cytokine/chemokine levels.
Collapse
Affiliation(s)
- Suman Dalal
- Department of Biomedical Sciences, James H Quillen College of Medicine, USA; Department of Health Sciences, USA; Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, TN, USA
| | - Paige L Shook
- Department of Biomedical Sciences, James H Quillen College of Medicine, USA
| | - Mahipal Singh
- Department of Biomedical Sciences, James H Quillen College of Medicine, USA
| | - Krishna Singh
- Department of Biomedical Sciences, James H Quillen College of Medicine, USA; Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, TN, USA; James H Quillen Veterans Affairs Medical Center, Mountain Home, TN, USA.
| |
Collapse
|
36
|
Angelovski M, Hadzi-Petrushev N, Mitrokhin V, Kamkin A, Mladenov M. Myocardial infarction and oxidative damage in animal models: objective and expectations from the application of cysteine derivatives. Toxicol Mech Methods 2023; 33:1-17. [PMID: 35450505 DOI: 10.1080/15376516.2022.2069530] [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] [Indexed: 01/05/2023]
Abstract
Reactive oxygen species (ROS) and associated oxidative stress are the main contributors to pathophysiological changes following myocardial infarction (MI), which is the principal cause of death from cardiovascular disease. The glutathione (GSH)/glutathione peroxidase (GPx) system appears to be the main and most active cardiac antioxidant mechanism. Hence, enhancement of the myocardial GSH system might have protective effects in the setting of MI. It follows that by increasing antioxidant capacity, the heart will be able to reduce the damage associated with MI and even prevent/weaken the occurrence of oxidative stress, which is highly ranked among the factors responsible for the occurrence of acute MI. For these reasons, the primary goal of future investigations should be to address the effects of different antioxidative compounds and especially cysteine derivatives like N-acetyl cysteine (NAC) and L-2-oxothiazolidine-4-carboxylic acid (OTC) as precursors responsible for the enhancement of the GSH-related antioxidant system's capacity. It is assumed that this will lay down the basis for elucidation of the mechanisms throughout which applicable doses of OTC will manifest a potentially positive impact in the reduction of adverse effects of acute MI. The inclusion of OTC in the models for prediction of the distribution of oxygen in infarcted animal hearts can help to upgrade existing computational models. Such a model would be based on computational geometries of the heart, but the inclusion of biochemical redox features in addition to angiogenic therapy, despite improvement of the post-infarcted oxygenated outcome could enhance the accuracy of the predictive values of oxygenation.
Collapse
Affiliation(s)
- Marija Angelovski
- Institute of Biology, Faculty of Natural Science and Mathematics, Ss Cyril and Methodius University, Skopje, North Macedonia
| | - Nikola Hadzi-Petrushev
- Institute of Biology, Faculty of Natural Science and Mathematics, Ss Cyril and Methodius University, Skopje, North Macedonia
| | - Vadim Mitrokhin
- Department of Fundamental and Applied Physiology, Russian National Research Medical University, Moscow, Russia
| | - Andre Kamkin
- Department of Fundamental and Applied Physiology, Russian National Research Medical University, Moscow, Russia
| | - Mitko Mladenov
- Institute of Biology, Faculty of Natural Science and Mathematics, Ss Cyril and Methodius University, Skopje, North Macedonia.,Department of Fundamental and Applied Physiology, Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
37
|
Xu L, He D, Wu Y, Shen L, Wang Y, Xu Y. Tanshinone IIA inhibits cardiomyocyte apoptosis and rescues cardiac function during doxorubicin-induced cardiotoxicity by activating the DAXX/MEK/ERK1/2 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 107:154471. [PMID: 36182795 DOI: 10.1016/j.phymed.2022.154471] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 09/05/2022] [Accepted: 09/20/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Heart failure (HF) is a common cardiovascular syndrome. Tanshinone IIA (Tan IIA) is a pharmacologically active monomer that exerts a significant cardioprotective effect in the clinic; however, the specific mechanisms are not fully understood. PURPOSE We mainly investigated the protective effects of Tan IIA on doxorubicin (DOX)-induced HF. METHODS In an in vitro study, H9C2 and HL-1 cells were cultured and treated with DOX and Tan IIA for 24 h, we investigated the mechanism underlying Tan IIA-mediated protection. In an in vivo study, a model of DOX-induced HF was established in C57BL/6 mice that were divided into the six groups randomly: a control group, a DOX group, DOX groups treated with Tan IIA (DOX+Tan IIA) at dosages of 2.5, 5 and 10 mg/kg/day and DOX groups treated with N-acetylcysteine (NAC) at dosages of 200 mg/kg/day. RESULT The results demonstrated that Tan IIA significantly increased cell viability and protected against DOX-induced apoptosis. RNA-sequencing showed that the genes expression associated with the apoptotic signaling pathway was altered by Tan IIA. Among the differentially expressed genes, death-domain associated protein (DAXX), which plays an critical role in apoptotic signaling, exhibited increased expression under Tan IIA treatment. In addition, RNA interference was used to silence the expression of DAXX, which abolished Tan IIA-mediated protection against DOX-induced apoptosis; this effect was associated with extracellular signal-regulated protein kinase 1/2 (ERK1/2) and mitogen-activated protein kinase (MEK) expression. In the in vivo study, the echocardiography results revealed that heart function was rescued by Tan IIA, and the histomorphology results showed that Tan IIA prevented myocardial structural alteration and myofibril disruption. Furthermore, Tan IIA induced the expressions of DAXX, p-ERK1/2 and p-MEK. Tan IIA also inhibited apoptosis by suppressing the expression of cleaved caspase-8, p-P38 and cleaved caspase-3. CONCLUSION Our results provide novel interpretations into the important role of DAXX in DOX-induced cardiotoxicity and show that Tan IIA may be a novel agent strategy for HF treatment via activating the DAXX/MEK/ERK1/2 pathway.
Collapse
Affiliation(s)
- Linhao Xu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, #261 Huansha Road, Shangcheng District, Hangzhou, Zhejiang 310006, China; Translational Medicine Research Center, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Daqiang He
- Department of Laboratory Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Yirong Wu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, #261 Huansha Road, Shangcheng District, Hangzhou, Zhejiang 310006, China
| | - Lishui Shen
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, #261 Huansha Road, Shangcheng District, Hangzhou, Zhejiang 310006, China
| | - Yongmei Wang
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, #261 Huansha Road, Shangcheng District, Hangzhou, Zhejiang 310006, China
| | - Yizhou Xu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, #261 Huansha Road, Shangcheng District, Hangzhou, Zhejiang 310006, China.
| |
Collapse
|
38
|
Zhao X, Zhao X, Jin F, Wang L, Zhang L. Prognostic Value of
Cardiac‐MRI
Scar Heterogeneity Combined With Left Ventricular Strain in Patients With Myocardial Infarction. J Magn Reson Imaging 2022. [DOI: 10.1002/jmri.28478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/29/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Affiliation(s)
- Xiaoying Zhao
- Department of Radiology The Second Affiliated Hospital of Kunming Medical University Kunming China
| | - Xinxiang Zhao
- Department of Radiology The Second Affiliated Hospital of Kunming Medical University Kunming China
| | - Fuwei Jin
- Department of Radiology The Second Affiliated Hospital of Kunming Medical University Kunming China
| | - Lujing Wang
- Department of Radiology The Second Affiliated Hospital of Kunming Medical University Kunming China
| | - Li Zhang
- Department of Radiology The Second Affiliated Hospital of Kunming Medical University Kunming China
| |
Collapse
|
39
|
Gu YH, Ren KW, Wang Y, Wang SH, Yu XH, Xu LW, Li HH, Bi HL. Administration of USP7 inhibitor P22077 inhibited cardiac hypertrophy and remodeling in Ang II-induced hypertensive mice. Front Pharmacol 2022; 13:1021361. [PMID: 36386139 PMCID: PMC9640964 DOI: 10.3389/fphar.2022.1021361] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/10/2022] [Indexed: 09/12/2023] Open
Abstract
Hypertension is one of the common causes of pathological cardiac hypertrophy and a major risk for morbidity and mortality of cardiovascular diseases worldwide. Ubiquitin-Specific Protease 7 (USP7), the first identified deubiquitinating enzymes, participated in a variety of biological processes, such as cell proliferation, DNA damage response, tumourigenesis, and apoptosis. However, its role and mechanism in cardiac remodeling remain unclear. Here, our data indicated that USP7 expression was increased during Ang II-induced cardiac hypertrophy and remodeling in mice and humans with heart failure, while the administration of its inhibitor p22077 attenuated cardiac hypertrophy, cardiac fibrosis, inflammation, and oxidase stress. Mechanistically, the administration of p22077 inhibited the multiple signaling pathways, including AKT/ERK, TGF-β/SMAD2/Collagen I/Collagen III, NF-κB/NLRP3, and NAPDH oxidases (NOX2 and NOX4). Taken together, these findings demonstrate that USP7 may be a new therapeutic target for hypertrophic remodeling and HF.
Collapse
Affiliation(s)
- Yu-Hui Gu
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Kai-Wen Ren
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yu Wang
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shi-Hao Wang
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiao-Hong Yu
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Li-Wen Xu
- Department of Obstetrics, Dalian Maternal and Child Health Institute, Dalian, China
| | - Hui-Hua Li
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Hai-Lian Bi
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
40
|
Left Ventricular Remodeling and Heart Failure Predictors in Acute Myocardial Infarction Patients with Preserved Left Ventricular Ejection Fraction after Successful Percutaneous Intervention in Western Romania. Life (Basel) 2022; 12:life12101636. [PMID: 36295071 PMCID: PMC9604641 DOI: 10.3390/life12101636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/08/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022] Open
Abstract
(1) Acute myocardial infarction (AMI) patients are at risk of left ventricular (LV) remodeling and heart failure (HF), even after successful revascularization by percutaneous coronary intervention (PCI). We wanted to assess the independent predictors of these outcomes in AMI patients. (2) Methods: The study enrolled patients with a LVEF ≥50% after a successful PCI for their first AMI. After 24 months, patients were separated into two groups based on whether their LVEF remained ≥50% (group I), or decreased to <50% (group II). (3) Outcomes: 26% of the patients experienced a decrease in LVEF below 50%, 41% showed LV remodeling, and 8% had experienced HF hospitalizations. HF hospitalizations were significantly more frequent in group II patients (p < 0.0001). The Killip class at admission >2, infarct-related longitudinal strain ≤−12.5%, and the presence of LV remodeling were identified as independent predictors of HF hospitalizations. (4) Conclusions: About 26% of AMI patients with normal LV function after a successful PCI developed HF. More sensitive techniques are required that allow for a more efficient risk-stratification and preventive therapy to reduce LV remodeling and HF in AMI patients with LVEF ≥50% after a successful PCI. The detection of abnormal ventricular deformation patterns after PCI by speckle-tracking echocardiography might be a valuable method in this approach.
Collapse
|
41
|
Tomimatsu M, Matsumoto K, Ashizuka M, Kumagai S, Tanaka S, Nakae T, Yokota K, Kominami S, Kajiura R, Okuzaki D, Motooka D, Shiraishi A, Abe T, Matsuda H, Okada Y, Maeda M, Seno S, Obana M, Fujio Y. Myeloid cell-specific ablation of Runx2 gene exacerbates post-infarct cardiac remodeling. Sci Rep 2022; 12:16656. [PMID: 36198906 PMCID: PMC9534857 DOI: 10.1038/s41598-022-21202-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
Runt-related transcription factor 2 (Runx2), a regulator of osteoblast differentiation, is pathologically involved in vascular calcification; however, the significance of Runx2 in cardiac homeostasis remains unclear. Here, we investigated the roles of Runx2 in cardiac remodeling after myocardial infarction (MI). The expression of Runx2 mRNA and protein was upregulated in murine hearts after MI. Runx2 was expressed in heart-infiltrating myeloid cells, especially in macrophages, at the border zone of post-infarct myocardium. To analyze the biological functions of Runx2 in cardiac remodeling, myeloid cell-specific Runx2 deficient (CKO) mice were exposed to MI. After MI, ventricular weight/tibia length ratio was increased in CKO mice, concomitant with severe cardiac dysfunction. Cardiac fibrosis was exacerbated in CKO mice, consistent with the upregulation of collagen 1a1 expression. Mechanistically, immunohistochemical analysis using anti-CD31 antibody showed that capillary density was decreased in CKO mice. Additionally, conditioned culture media of myeloid cells from Runx2 deficient mice exposed to MI induced the tube formation of vascular endothelial cells to a lesser extent than those from control mice. RNA-sequence showed that the expression of pro-angiogenic or anti-angiogenic factors was altered in macrophages from Runx2-deficient mice. Collectively, Runx2+ myeloid cells infiltrate into post-infarct myocardium and prevent adverse cardiac remodeling, at least partially, by regulating endothelial cell function.
Collapse
Affiliation(s)
- Masashi Tomimatsu
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Kotaro Matsumoto
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Moe Ashizuka
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Shohei Kumagai
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Shota Tanaka
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Takafumi Nakae
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Kosei Yokota
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Shunsuke Kominami
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Ryota Kajiura
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Daisuke Motooka
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Aki Shiraishi
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Takaya Abe
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Hideo Matsuda
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, Suita, Osaka, Japan
| | - Yoshiaki Okada
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Makiko Maeda
- Laboratory of Clinical Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan.,Medical Center for Translational Research, Department of Medical Innovation, Osaka University Hospital, Suita, Osaka, Japan
| | - Shigeto Seno
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, Suita, Osaka, Japan
| | - Masanori Obana
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan. .,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan. .,Global Center for Medical Engineering and Informatics (MEI), Osaka University, Suita, Osaka, Japan. .,Radioisotope Research Center, Institute for Radiation Science, Osaka University, Suita, Osaka, Japan.
| | - Yasushi Fujio
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan. .,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan.
| |
Collapse
|
42
|
Bredin F. Long-term follow-up of survival after passive containment surgery in dilated cardiomyopathy. Ann Med Surg (Lond) 2022; 80:104241. [PMID: 35992206 PMCID: PMC9382418 DOI: 10.1016/j.amsu.2022.104241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/14/2022] [Accepted: 07/20/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Fredrik Bredin
- Karolinska Institutet, Department of Molecular Medicine and Surgery, Karolinska University Hospital, 171 76, Stockholm, Sweden.
| |
Collapse
|
43
|
Leancă SA, Crișu D, Petriș AO, Afrăsânie I, Genes A, Costache AD, Tesloianu DN, Costache II. Left Ventricular Remodeling after Myocardial Infarction: From Physiopathology to Treatment. Life (Basel) 2022; 12:1111. [PMID: 35892913 PMCID: PMC9332014 DOI: 10.3390/life12081111] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 12/11/2022] Open
Abstract
Myocardial infarction (MI) is the leading cause of death and morbidity worldwide, with an incidence relatively high in developed countries and rapidly growing in developing countries. The most common cause of MI is the rupture of an atherosclerotic plaque with subsequent thrombotic occlusion in the coronary circulation. This causes cardiomyocyte death and myocardial necrosis, with subsequent inflammation and fibrosis. Current therapies aim to restore coronary flow by thrombus dissolution with pharmaceutical treatment and/or intravascular stent implantation and to counteract neurohormonal activation. Despite these therapies, the injury caused by myocardial ischemia leads to left ventricular remodeling; this process involves changes in cardiac geometry, dimension and function and eventually progression to heart failure (HF). This review describes the pathophysiological mechanism that leads to cardiac remodeling and the therapeutic strategies with a role in slowing the progression of remodeling and improving cardiac structure and function.
Collapse
Affiliation(s)
- Sabina Andreea Leancă
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Daniela Crișu
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Antoniu Octavian Petriș
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Str. University nr. 16, 700083 Iasi, Romania;
| | - Irina Afrăsânie
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Antonia Genes
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Alexandru Dan Costache
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Str. University nr. 16, 700083 Iasi, Romania;
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Dan Nicolae Tesloianu
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Irina Iuliana Costache
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Str. University nr. 16, 700083 Iasi, Romania;
| |
Collapse
|
44
|
Luo M, Mou Q, Liu L, Tian J, Liu L. Treg/Th17 Ratio Regulation May Play an Important Role in Epigallocatechin-3-Gallate-Mediated Attenuation of Increased Afterload-Induced Cardiac Hypertrophy. J Cardiovasc Pharmacol 2022; 79:711-718. [PMID: 35058409 PMCID: PMC9067088 DOI: 10.1097/fjc.0000000000001220] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/23/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT The aim of this study was to investigate whether Treg/Th17 ratio regulation plays an important role in epigallocatechin-3-gallate (EGCG) in attenuating increased afterload-induced cardiac hypertrophy. Three-month-old male C57BL/6 mice were divided into sham + vehicle, abdominal aortic constriction (AAC) + vehicle, and AAC + EGCG groups. Intraperitoneal EGCG (50 mg/kg/d) administration was conducted. Cardiac structure and function were examined by ultrasonography. Pathology was examined by hematoxylin and eosin staining, wheat germ agglutinin staining, and Masson's trichome staining. T-lymphocyte subtypes were analyzed using immunofluorescence and flow cytometry assays. Ultrasonography showed that the ventricular wall in the AAC + vehicle group was thicker than that in the sham + vehicle group (P < 0.05). Hematoxylin and eosin staining revealed cardiomyocyte hypertrophy accompanied by a small amount of inflammatory cell infiltration in the AAC + vehicle group. The results of wheat germ agglutinin staining demonstrated the presence of hypertrophic cardiomyocytes in the AAC + vehicle group (P < 0.01). Masson's trichome staining showed cardiac fibrosis in the AAC + vehicle group, and the immunofluorescence assay revealed infiltration of CD4+ cells in both AAC + vehicle and AAC + EGCG groups. Splenic flow cytometry showed a significant increase in the proportion of Treg cells in the AAC + EGCG group (P < 0.05). The proportion of Th17 cells in the AAC + vehicle group was significantly higher than that in the sham + vehicle group (P < 0.05). In conclusion, changes in the Treg/Th17 ratio are associated with the occurrence of myocardial hypertrophy caused by increased afterload. Moreover, regulation of the Treg/Th17 ratio by EGCG may play an important role in the attenuation of myocardial hypertrophy.
Collapse
Affiliation(s)
- Min Luo
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Department of Cardiology, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China; and
| | - Qiuhong Mou
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Department of Cardiology, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China; and
| | - Lingjuan Liu
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Department of Cardiology, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China; and
| | - Jie Tian
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Department of Cardiology, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China; and
| | - Lifei Liu
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Department of Cardiology, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China; and
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
| |
Collapse
|
45
|
Clavere NG, Alqallaf A, Rostron KA, Parnell A, Mitchell R, Patel K, Boateng SY. Inhibition of activin A receptor signalling attenuates age-related pathological cardiac remodelling. Dis Model Mech 2022; 15:275323. [PMID: 35380160 PMCID: PMC9118092 DOI: 10.1242/dmm.049424] [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: 12/17/2021] [Accepted: 03/16/2022] [Indexed: 11/20/2022] Open
Abstract
In the heart, ageing is associated with DNA damage, oxidative stress, fibrosis and activation of the activin signalling pathway, leading to cardiac dysfunction. The cardiac effects of activin signalling blockade in progeria are unknown. This study investigated the cardiac effects of progeria induced by attenuated levels of Ercc1, which is required for DNA excision and repair, and the impact of activin signalling blockade using a soluble activin receptor type IIB (sActRIIB). DNA damage and oxidative stress were significantly increased in Ercc1Δ/− hearts, but were reduced by sActRIIB treatment. sActRIIB treatment improved cardiac systolic function and induced cardiomyocyte hypertrophy in Ercc1Δ/− hearts. RNA-sequencing analysis showed that in Ercc1Δ/− hearts, there was an increase in pro-oxidant and a decrease in antioxidant gene expression, whereas sActRIIB treatment reversed this effect. Ercc1Δ/− hearts also expressed higher levels of anti-hypertrophic genes and decreased levels of pro-hypertrophic ones, which were also reversed by sActRIIB treatment. These results show for the first time that inhibition of activin A receptor signalling attenuates cardiac dysfunction, pathological tissue remodelling and gene expression in Ercc1-deficient mice and presents a potentially novel therapeutic target for heart diseases. Summary: Attenuated DNA repair is associated with pathological cardiac remodelling and gene expression. Much of this phenotype is attenuated by inhibition of the activin signalling pathway using soluble activin receptor treatment.
Collapse
Affiliation(s)
- Nicolas G Clavere
- Institute of Cardiovascular and Metabolic Research, School of Biological Sciences, Health and Life Sciences Building, University of Reading, Whiteknights, Reading RG6 6UB, UK
| | - Ali Alqallaf
- Institute of Cardiovascular and Metabolic Research, School of Biological Sciences, Health and Life Sciences Building, University of Reading, Whiteknights, Reading RG6 6UB, UK
| | - Kerry A Rostron
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Commonwealth Building, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - Andrew Parnell
- Institute of Cardiovascular and Metabolic Research, School of Biological Sciences, Health and Life Sciences Building, University of Reading, Whiteknights, Reading RG6 6UB, UK
| | - Robert Mitchell
- Institute of Cardiovascular and Metabolic Research, School of Biological Sciences, Health and Life Sciences Building, University of Reading, Whiteknights, Reading RG6 6UB, UK
| | - Ketan Patel
- Institute of Cardiovascular and Metabolic Research, School of Biological Sciences, Health and Life Sciences Building, University of Reading, Whiteknights, Reading RG6 6UB, UK
| | - Samuel Y Boateng
- Institute of Cardiovascular and Metabolic Research, School of Biological Sciences, Health and Life Sciences Building, University of Reading, Whiteknights, Reading RG6 6UB, UK
| |
Collapse
|
46
|
Toncelli L, Pasquini L, Masini G, Orlandi M, Paci G, Mecacci F, Pedrizzetti G, Galanti G. Difference in cardiac remodeling between female athletes and pregnant women: a case control study. Cardiovasc Ultrasound 2022; 20:10. [PMID: 35418063 PMCID: PMC9006435 DOI: 10.1186/s12947-022-00280-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 03/25/2022] [Indexed: 12/05/2022] Open
Abstract
OBJECTIVES The aim of this study was to detect possible differences in reversible cardiac remodeling occurring in sport training and twin pregnancy. BACKGROUND cardiac remodeling occurs in athletes and pregnant women due to training and fetal requirements, respectively. These changes could be apparently similar. METHODS 21 female elite athletes (23.2 ± 5.3 years), 25 women with twin pregnancies (35.4 ± 5.7 years) and 25 healthy competitive female athletes (controls), age-matched with pregnant women (34.9 ± 7.9 years), were enrolled. This latter group was included to minimize the effect of age on cardiac remodeling. All women evaluated through anamnestic collection, physical examination, 12 leads ECG, standard echocardiogram and strain analysis. Sphericity (SI) and apical conicity (ACI) indexes were also calculated. RESULTS Pregnant women showed higher LA dimension (p < 0.001) compared to both groups of athletes. LV e RV GLS were significantly different in pregnant women compared to female athletes (p = 0.02 and 0.03, respectively). RV GLS was also different between pregnant women and controls (p = 0.02). Pregnant women showed significantly higher S' wave compared to female athletes (p = 0.02) but not controls. Parameters of diastolic function were significantly higher in athletes (p = 0.08 for IVRT and p < 0.001 for E/A,). SI was lower in athletes in both diastole (p = 0.01) and systole (p < 0.001), while ACIs was lower in pregnant women (p = 0.04). CONCLUSIONS Cardiac remodeling of athletes and pregnant women could be similar at first sight but different in LV shape and in GLS, highlighting a profound difference in longitudinal deformation between athletes and pregnant women. This difference seems not to be related with age. These findings suggest that an initial maternal cardiovascular maladaptation could occur in the third trimester of twin pregnancies.
Collapse
Affiliation(s)
- Loira Toncelli
- Sport and Exercise Medicine Department of University of Florence, via delle Oblate 4, 50100, Florence, FI, Italy
| | - Lucia Pasquini
- Fetal Medicine Unit, Department for Women and Children Health, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Giulia Masini
- Fetal Medicine Unit, Department for Women and Children Health, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Melissa Orlandi
- Sport and Exercise Medicine Department of University of Florence, via delle Oblate 4, 50100, Florence, FI, Italy
| | - Gabriele Paci
- Sport and Exercise Medicine Department of University of Florence, via delle Oblate 4, 50100, Florence, FI, Italy
| | - Federico Mecacci
- Fetal Medicine Unit, Department for Women and Children Health, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Gianni Pedrizzetti
- Architectural and Engineering Department of University of Trieste, Trieste, Italy
| | - Giorgio Galanti
- Sport and Exercise Medicine Department of University of Florence, via delle Oblate 4, 50100, Florence, FI, Italy.
| |
Collapse
|
47
|
Remodeling and Fibrosis of the Cardiac Muscle in the Course of Obesity-Pathogenesis and Involvement of the Extracellular Matrix. Int J Mol Sci 2022; 23:ijms23084195. [PMID: 35457013 PMCID: PMC9032681 DOI: 10.3390/ijms23084195] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/09/2022] [Indexed: 12/16/2022] Open
Abstract
Obesity is a growing epidemiological problem, as two-thirds of the adult population are carrying excess weight. It is a risk factor for the development of cardiovascular diseases (hypertension, ischemic heart disease, myocardial infarct, and atrial fibrillation). It has also been shown that chronic obesity in people may be a cause for the development of heart failure with preserved ejection fraction (HFpEF), whose components include cellular hypertrophy, left ventricular diastolic dysfunction, and increased extracellular collagen deposition. Several animal models with induced obesity, via the administration of a high-fat diet, also developed increased heart fibrosis as a result of extracellular collagen accumulation. Excessive collagen deposition in the extracellular matrix (ECM) in the course of obesity may increase the stiffness of the myocardium and thereby deteriorate the heart diastolic function and facilitate the occurrence of HFpEF. In this review, we include a rationale for that process, including a discussion about possible putative factors (such as increased renin–angiotensin–aldosterone activity, sympathetic overdrive, hemodynamic alterations, hypoadiponectinemia, hyperleptinemia, and concomitant heart diseases). To address the topic clearly, we include a description of the fundamentals of ECM turnover, as well as a summary of studies assessing collagen deposition in obese individuals.
Collapse
|
48
|
Zhang Z, Ding S, Wang Z, Zhu X, Zhou Z, Zhang W, Yang X, Ge J. Prmt1 upregulated by Hdc deficiency aggravates acute myocardial infarction via NETosis. Acta Pharm Sin B 2022; 12:1840-1855. [PMID: 35847488 PMCID: PMC9279636 DOI: 10.1016/j.apsb.2021.10.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/20/2021] [Accepted: 10/11/2021] [Indexed: 01/03/2023] Open
Abstract
Neutrophils are mobilized and recruited to the injured heart after myocardial infarction, and neutrophil count has been clinically implicated to be associated with coronary disease severity. Histidine decarboxylase (HDC) has been implicated in regulating reactive oxidative species (ROS) and the differentiation of myeloid cells. However, the effect of HDC on neutrophils after myocardial infarction remains unclear. Here, we found that neutrophils were disorderly recruited into the ischemic injured area of the myocardium of Hdc deficiency (Hdc−/−) mice. Moreover, Hdc deficiency led to attenuated adhesion but enhanced migration and augmented ROS/neutrophil extracellular traps (NETs) production in neutrophils. Hdc−/− mouse-derived NETs promoted cardiomyocyte death and cardiac fibroblast proliferation/migration. Furthermore, protein arginine methyltransferase 1 (PRMT1) was increased in Hdc−/− mouse-derived neutrophils but decreased with exogenous histamine treatment. Its expression could be rescued by blocking histamine receptor 1 (H1R), inhibiting ATP synthesis or reducing SWItch/sucrose non fermentable (SWI/SNF) chromatin remodeling complex. Accordingly, histamine or MS023 treatment could decrease ROS and NETs ex vivo, and ameliorated cardiac function and fibrosis, along with the reduced NETs in plasma in vivo. Together, our findings unveil the role of HDC in NETosis by histamine–H1R–ATP–SWI/SNF–PRMT1–ROS signaling and provide new biomarkers and targets for identifying and tuning the detrimental immune state in cardiovascular disease.
Collapse
|
49
|
Beneficial Effects of Dietary Nitrite on a Model of Nonalcoholic Steatohepatitis Induced by High-Fat/High-Cholesterol Diets in SHRSP5/Dmcr Rats: A Preliminary Study. Int J Mol Sci 2022; 23:ijms23062931. [PMID: 35328352 PMCID: PMC8951310 DOI: 10.3390/ijms23062931] [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: 02/05/2022] [Revised: 03/06/2022] [Accepted: 03/07/2022] [Indexed: 12/10/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is a chronic liver disease that leads to liver cirrhosis and hepatocellular carcinoma. Endothelial dysfunction caused by hepatic lipotoxicity is an underlying NASH pathology observed in the liver and the cardiovascular system. Here, we evaluated the effect of dietary nitrite on a rat NASH model. Stroke-prone, spontaneously hypertensive 5/Dmcr rats were fed a high-fat/high-cholesterol diet to develop the NASH model, with nitrite or captopril (100 mg/L, each) supplementation in drinking water for 8 weeks. The effects of nitrite and captopril were evaluated using immunohistochemical analyses of the liver and heart tissues. Dietary nitrite suppressed liver fibrosis in the rats by reducing oxidative stress, as measured using the protein levels of nicotinamide adenine dinucleotide phosphate oxidase components and inflammatory cell accumulation in the liver. Nitrite lowered the blood pressure in hypertensive NASH rats and suppressed left ventricular chamber enlargement. Similar therapeutic effects were observed in a captopril-treated rat NASH model, suggesting the possibility of a common signaling pathway through which nitrite and captopril improve NASH pathology. In conclusion, dietary nitrite attenuates the development of NASH with cardiovascular involvement in rats and provides an alternative NASH therapeutic strategy.
Collapse
|
50
|
Figueiredo AM, Cardoso AC, Pereira BLB, Silva RAC, Ripa AFGD, Pinelli TFB, Oliveira BC, Rafacho BPM, Ishikawa LLW, Azevedo PS, Okoshi K, Fernandes AAH, Zornoff LAM, Minicucci MF, Polegato BF, Paiva SAR. Açai supplementation (Euterpe oleracea Mart.) attenuates cardiac remodeling after myocardial infarction in rats through different mechanistic pathways. PLoS One 2022; 17:e0264854. [PMID: 35245316 PMCID: PMC8896726 DOI: 10.1371/journal.pone.0264854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 02/17/2022] [Indexed: 11/18/2022] Open
Abstract
Myocardial infarction has a high mortality rate worldwide. Therefore, clinical intervention in cardiac remodeling after myocardial infarction is essential. Açai pulp is a natural product and has been considered a functional food because of its antioxidant/anti-inflammatory properties. The aim of the present study was to analyze the effect of açai pulp supplementation on cardiac remodeling after myocardial infarction in rats. After 7 days of surgery, male Wistar rats were assigned to six groups: sham animals fed standard chow (SA0, n = 14), fed standard chow with 2% açai pulp (SA2, n = 12) and fed standard chow with 5% açai pulp (SA5, n = 14), infarcted animals fed standard chow (IA0, n = 12), fed standard chow with 2% açai pulp (IA2, n = 12), and fed standard chow with 5% açai pulp (IA5, n = 12). After 3 months of supplementation, echocardiography and euthanasia were performed. Açai pulp supplementation, after myocardial infarction, improved energy metabolism, attenuated oxidative stress (lower concentration of malondialdehyde, P = 0.023; dose-dependent effect), modulated the inflammatory process (lower concentration of interleukin-10, P<0.001; dose-dependent effect) and decreased the deposit of collagen (lower percentage of interstitial collagen fraction, P<0.001; dose-dependent effect). In conclusion, açai pulp supplementation attenuated cardiac remodeling after myocardial infarction in rats. Also, different doses of açai pulp supplementation have dose-dependent effects on cardiac remodeling.
Collapse
Affiliation(s)
- Amanda Menezes Figueiredo
- Internal Medicine Department, Botucatu Medical School, Universidade Estadual Paulista (UNESP), Botucatu, São Paulo, Brazil
- * E-mail:
| | - Ana Carolina Cardoso
- Internal Medicine Department, Botucatu Medical School, Universidade Estadual Paulista (UNESP), Botucatu, São Paulo, Brazil
| | - Bruna Leticia Buzati Pereira
- Internal Medicine Department, Botucatu Medical School, Universidade Estadual Paulista (UNESP), Botucatu, São Paulo, Brazil
| | - Renata Aparecida Candido Silva
- Internal Medicine Department, Botucatu Medical School, Universidade Estadual Paulista (UNESP), Botucatu, São Paulo, Brazil
| | | | | | - Bruna Camargo Oliveira
- Internal Medicine Department, Botucatu Medical School, Universidade Estadual Paulista (UNESP), Botucatu, São Paulo, Brazil
| | - Bruna Paola Murino Rafacho
- Internal Medicine Department, Botucatu Medical School, Universidade Estadual Paulista (UNESP), Botucatu, São Paulo, Brazil
| | - Larissa Lumi Watanabe Ishikawa
- Chemistry and Biochemistry Department, Institute of Biosciences, Universidade Estadual Paulista (UNESP), Botucatu, São Paulo, Brazil
| | - Paula Schmidt Azevedo
- Internal Medicine Department, Botucatu Medical School, Universidade Estadual Paulista (UNESP), Botucatu, São Paulo, Brazil
| | - Katashi Okoshi
- Internal Medicine Department, Botucatu Medical School, Universidade Estadual Paulista (UNESP), Botucatu, São Paulo, Brazil
| | - Ana Angelica Henrique Fernandes
- Chemistry and Biochemistry Department, Institute of Biosciences, Universidade Estadual Paulista (UNESP), Botucatu, São Paulo, Brazil
| | | | - Marcos Ferreira Minicucci
- Internal Medicine Department, Botucatu Medical School, Universidade Estadual Paulista (UNESP), Botucatu, São Paulo, Brazil
| | - Bertha Furlan Polegato
- Internal Medicine Department, Botucatu Medical School, Universidade Estadual Paulista (UNESP), Botucatu, São Paulo, Brazil
| | - Sergio Alberto Rupp Paiva
- Internal Medicine Department, Botucatu Medical School, Universidade Estadual Paulista (UNESP), Botucatu, São Paulo, Brazil
| |
Collapse
|