1
|
Medina-Terol GJ, Chimal L, Huerta de la Cruz S, Ávila G, Aranda A, Cruz-Robles D, Centurión D, Altamirano J, Rojo R, Gómez-Viquez NL. H 2S treatment reverts cardiac hypertrophy and increases SERCA2a activity but does not fully restore cardiac Ca 2+ handling in hypertensive rats. Cell Calcium 2025; 128:103015. [PMID: 40184980 DOI: 10.1016/j.ceca.2025.103015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/15/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025]
Abstract
Hydrogen sulfide (H2S) has been proposed to play a cardioprotective role, particularly due to its ability to revert left ventricular hypertrophy (LVH) and mitigate cardiac dysfunction in various cardiomyopathies, including hypertensive heart disease. However, the extent to which cardioprotection by H2S involves improvement in Ca2+ handling remains unclear. Although H2S has been reported to influence the function of key Ca2+ handling proteins, most studies have focused on acute administration of H2S donors in isolated cardiomyocytes, rather than in a therapeutic context. In this study, we used a rat model of hypertension induced by abdominal aortic coarctation (AAC) to evaluate the therapeutic potential of NaHS, an H2S donor, on LVH and Ca2+ handling. After 8 weeks of AAC, hypertensive rats developed moderate LVH, which was accompanied by a reduction in both the amplitude and the rate of rise of systolic Ca2+ transients, as well as a decrease in sarcoplasmic reticulum (SR) Ca2+ load. Despite the reduced SR Ca2+ load, the frequency of diastolic Ca2+ sparks remained high, while the incidence and propagation rate of spontaneous Ca2+ waves significantly increased, suggesting enhanced diastolic SR Ca2+ leak, most likely due to hypersensitivity of ryanodine receptors (RyR2) to Ca2+. On the other hand, NaHS administration during the final 4 weeks of AAC reverted both LVH and hypertension, and increased SR Ca2+ reuptake mediated by the SR Ca2+ ATPase (SERCA2a). However, NaHS treatment failed to restore the amplitude and rate of rise of systolic Ca2+ transients or SR Ca2+ load. Furthermore, SR Ca2+ leak might have worsened, since spontaneous Ca2+ waves increased. In conclusion, NaHS treatment does not appear to normalize all Ca2+ handling properties during hypertensive LVH. On the contrary, NaHS may exert an arrhythmogenic effect, likely due to enhanced SERCA2a activity under conditions of unresolved RyR2 Ca2+ hypersensitivity.
Collapse
Affiliation(s)
- Grecia J Medina-Terol
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados-Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Luis Chimal
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados-Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Saúl Huerta de la Cruz
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados-Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Guillermo Ávila
- Departamento de Bioquímica, Centro de Investigación y de Estudios Avanzados-Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Alberto Aranda
- Instituto Nacional de Cardiología Ignacio Chávez, Departamento de Anatomía Patológica, Ciudad de México, Mexico
| | - David Cruz-Robles
- Instituto Nacional de Cardiología Ignacio Chávez, Departamento de Biología Molecular, Ciudad de México, Mexico
| | - David Centurión
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados-Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Julio Altamirano
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Chihuahua, Mexico
| | - Rocio Rojo
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Chihuahua, Mexico
| | - Norma Leticia Gómez-Viquez
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados-Instituto Politécnico Nacional, Ciudad de México, Mexico.
| |
Collapse
|
2
|
Oikonomou EK, Khera R. From Pixels to Patterns: Radiomic Subphenotyping of Left Ventricular Hypertrophy on Echocardiography. Circ Cardiovasc Imaging 2025:e018291. [PMID: 40326361 DOI: 10.1161/circimaging.125.018291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Affiliation(s)
- Evangelos K Oikonomou
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT. (E.K.O., R.K.)
- Cardiovascular Data Science (CarDS) Lab, Yale School of Medicine, New Haven, CT. (E.K.O., R.K.)
| | - Rohan Khera
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT. (E.K.O., R.K.)
- Cardiovascular Data Science (CarDS) Lab, Yale School of Medicine, New Haven, CT. (E.K.O., R.K.)
- Section of Biomedical Informatics and Data Science, Yale School of Medicine, New Haven, CT. (R.K.)
- Center for Outcomes Research and Evaluation, Yale-New Haven Hospital, CT (R.K.)
- Section of Health Informatics, Department of Biostatistics, Yale School of Public Health, New Haven, CT (R.K.)
| |
Collapse
|
3
|
Ogyu A, Rouzier V, Sufra R, St Sauveur R, Jean-Pierre MC, Lin JQ, Mourra N, Preval F, Jean M, Devereux RB, Pirmohamed A, Goyal P, de Las Fuentes L, Dávila-Román VG, Alexandre W, Peck RN, Deschamps MM, Pape JW, McNairy ML, Yan LD. Left ventricular hypertrophy among adults in a population-based cohort in Haiti. Sci Rep 2025; 15:12831. [PMID: 40229312 PMCID: PMC11997160 DOI: 10.1038/s41598-025-96837-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 04/01/2025] [Indexed: 04/16/2025] Open
Abstract
Left ventricular hypertrophy (LVH) is one of the strongest predictors of cardiovascular disease (CVD) and mortality; yet the means to diagnose LVH in resource-constrained settings remain limited. The objectives of this study were to determine LVH prevalence by transthoracic echocardiography (TTE) in a high-risk group, and compare TTE vs. electrocardiography (ECG-LVH) for LVH detection. We analyzed enrollment data from the Haiti cardiovascular disease cohort study on adults (≥ 18 years, n = 3,005) in Port-au-Prince between 2019 and 2021. All participants underwent questionnaires, vital signs, physical exams, and 12-lead ECGs. TTEs were acquired on those with hypertension or exhibiting CVD symptoms (n = 1040, 34.7%). TTE-LVH was defined according to the American Society of Echocardiography guidelines and ECG-LVH by Sokolow-Lyon, Cornell, and Limb-Lead Voltage criteria. The prevalence of TTE-LVH was 39.0% (95% CI 36.6-41.5%) and associated with older age. Only 26% of those with TTE-LVH and elevated blood pressure were on antihypertensives. Prevalence of ECG-LVH ranged from 1.9 to 5.0%, and compared to TTE-LVH had low agreement (κ < 0.20), low sensitivity (< 10%) and high specificity (> 90%). These findings indicate a high prevalence of TTE-LVH among high-risk Haitian adults, and poor detection using ECGs compared to TTEs. For those with TTE-LVH, treatment with antihypertensives may reduce the risk of adverse CVD outcomes.
Collapse
Affiliation(s)
- Anju Ogyu
- Center for Global Health, Weill Cornell Medicine, New York City, NY, USA.
| | - Vanessa Rouzier
- Haitian Group for the Study of Kaposi's Sarcoma and Opportunistic Infections (GHESKIO), Port-au-Prince, Haiti
| | - Rodney Sufra
- Haitian Group for the Study of Kaposi's Sarcoma and Opportunistic Infections (GHESKIO), Port-au-Prince, Haiti
| | - Reichling St Sauveur
- Haitian Group for the Study of Kaposi's Sarcoma and Opportunistic Infections (GHESKIO), Port-au-Prince, Haiti
| | - Marie Christine Jean-Pierre
- Haitian Group for the Study of Kaposi's Sarcoma and Opportunistic Infections (GHESKIO), Port-au-Prince, Haiti
| | - Joanna Q Lin
- Division of General Internal Medicine, Weill Cornell Medicine, New York City, NY, USA
| | - Nour Mourra
- Center for Global Health, Weill Cornell Medicine, New York City, NY, USA
- Division of General Internal Medicine, Weill Cornell Medicine, New York City, NY, USA
| | - Fabiola Preval
- Haitian Group for the Study of Kaposi's Sarcoma and Opportunistic Infections (GHESKIO), Port-au-Prince, Haiti
| | - Mirline Jean
- Haitian Group for the Study of Kaposi's Sarcoma and Opportunistic Infections (GHESKIO), Port-au-Prince, Haiti
| | | | - Altaf Pirmohamed
- Division of Cardiology, Weill Cornell Medicine, New York City, NY, USA
| | - Parag Goyal
- Division of Cardiology, Weill Cornell Medicine, New York City, NY, USA
| | - Lisa de Las Fuentes
- Global Health Center, Institute for Public Health and Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Victor G Dávila-Román
- Global Health Center, Institute for Public Health and Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Wheytnie Alexandre
- Center for Global Health, Weill Cornell Medicine, New York City, NY, USA
- Division of General Internal Medicine, Weill Cornell Medicine, New York City, NY, USA
| | - Robert N Peck
- Center for Global Health, Weill Cornell Medicine, New York City, NY, USA
- Division of Infectious Disease, Weill Cornell Medicine, New York City, NY, USA
| | - Marie-Marcelle Deschamps
- Haitian Group for the Study of Kaposi's Sarcoma and Opportunistic Infections (GHESKIO), Port-au-Prince, Haiti
| | - Jean W Pape
- Haitian Group for the Study of Kaposi's Sarcoma and Opportunistic Infections (GHESKIO), Port-au-Prince, Haiti
| | - Margaret L McNairy
- Center for Global Health, Weill Cornell Medicine, New York City, NY, USA
- Division of General Internal Medicine, Weill Cornell Medicine, New York City, NY, USA
| | - Lily D Yan
- Center for Global Health, Weill Cornell Medicine, New York City, NY, USA
- Division of General Internal Medicine, Weill Cornell Medicine, New York City, NY, USA
| |
Collapse
|
4
|
Bracamonte JH, Watkins L, Pat B, Dell’Italia LJ, Saucerman JJ, Holmes JW. Contributions of mechanical loading and hormonal changes to eccentric hypertrophy during volume overload: A Bayesian analysis using logic-based network models. PLoS Comput Biol 2025; 21:e1012390. [PMID: 40238825 PMCID: PMC12040246 DOI: 10.1371/journal.pcbi.1012390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 04/29/2025] [Accepted: 02/23/2025] [Indexed: 04/18/2025] Open
Abstract
Primary mitral regurgitation (MR) is a pathology that alters mechanical loading on the left ventricle, triggers an array of compensatory neurohormonal responses, and induces a distinctive ventricular remodeling response known as eccentric hypertrophy. Drug therapies may alleviate symptoms, but only mitral valve repair or replacement can provide significant recovery of cardiac function and dimensions. Questions remain about the optimal timing of surgery, with 20% of patients developing systolic dysfunction post-operatively despite being treated according to the current guidelines. Thus, better understanding of the hypertrophic process in the setting of ventricular volume overload (VO) is needed to improve and better personalize the management of MR. To address this knowledge gap, we employ a Bayesian approach to combine data from 70 studies on experimental volume overload in dogs and rats and use it to calibrate a logic-based network model of hypertrophic signaling in myocytes. The calibrated model predicts that growth in experimental VO is mostly driven by the neurohormonal response, with an initial increase in myocardial tissue stretch being compensated by subsequent remodeling fairly early in the time course of VO. This observation contrasts with a common perception that volume-overload hypertrophy is driven primarily by increased myocyte strain. The model reproduces many aspects of 43 studies not used in its calibration, including infusion of individual hypertrophic agonists alone or in combination with various drugs commonly employed to treat heart failure, as well as administration of some of those drugs in the setting of experimental volume overload. We believe this represents a promising approach to using the known structure of an intracellular signaling network to integrate information from multiple studies into quantitative predictions of the range of expected responses to potential interventions in the complex setting of cardiac hypertrophy driven by a combination of hormonal and mechanical factors.
Collapse
Affiliation(s)
- Johane H. Bracamonte
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Lionel Watkins
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Betty Pat
- Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, United States of America
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Louis J. Dell’Italia
- Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, United States of America
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jeffrey J. Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jeffrey W. Holmes
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Division of Cardiothoracic Surgery, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
5
|
Liu H, Guo X, Yang J, Xia C, Yao Y, Li X, Liu X, Yang J, Li X, Xu Y, Li J, Wang M. Structure Optimization of Natural Product Catalpol to Obtain Novel and Potent Analogs against Heart Failure. J Med Chem 2025; 68:4540-4560. [PMID: 39925333 DOI: 10.1021/acs.jmedchem.4c02591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
Heart failure (HF) is a major global health threat, characterized by high morbidity and mortality. Targeting cardiac hypertrophy has been identified as a potential therapy for HF, with current treatments showing limited efficacy. Our research aims to address this limitation by exploring new structural classes of therapeutic agents. Starting from the natural product catalpol, we designed a series of novel catalpol analogs to break through the structural limitations of natural analogs, improve the anti-HF efficacy and metabolic properties. Among these, compound JZ19 exhibited remarkable efficacy in both myocardial cell injury assays and in an isoproterenol-induced murine HF model, outperforming catalpol. Our findings indicate that JZ19 potently reversed cardiac function by modulating the PI3K-AKT-GSK3β pathway, a key regulator of hypertrophy and apoptosis. Moreover, JZ19 showed favorable pharmacokinetic properties and safety. Overall, our results provide direct pharmacologic evidence supporting the further development of JZ19 as novel HF therapeutics by inhibiting cardiac hypertrophy and apoptosis.
Collapse
Affiliation(s)
- Hanfang Liu
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xiaobo Guo
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Jin Yang
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Conglong Xia
- College of Pharmacy, Dali University, Dali 671000, China
| | - Yue Yao
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xiao Li
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xiaoyang Liu
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Junqi Yang
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xiaokang Li
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yixiang Xu
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Jian Li
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi 832003, China
- Key Laboratory of Tropical Biological Resources of Ministry of Education, College of Pharmacy, Hainan University, Haikou 570228, China
| | - Manjiong Wang
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
6
|
Warnakulasooriya D, Bondarenko VE. EAD Mechanisms in Hypertrophic Mouse Ventricular Myocytes: Insights from a Compartmentalized Mathematical Model. Bull Math Biol 2025; 87:49. [PMID: 39992477 DOI: 10.1007/s11538-025-01423-3] [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: 07/20/2024] [Accepted: 01/31/2025] [Indexed: 02/25/2025]
Abstract
Transverse aortic constriction (TAC) is one of the experimental mouse models that are designed to investigate cardiac hypertrophy and heart failure. Most of the studies with this model are devoted to the stage of developed heart failure. However, several studies of the early stages (hypertrophy after 1 week of TAC) of this disease found significant changes in the β-adrenergic system, electrical activity, and Ca2+ dynamics in mouse ventricular myocytes. To provide a quantitative description of cardiac hypertrophy, we developed a new compartmentalized mathematical model of hypertrophic mouse ventricular myocytes for the early stage after the TAC procedure. The model described the changes in cell geometry, action potentials, [Ca2+]i transients, and β1- and β2-adrenergic signaling systems. We also showed that the hypertrophic myocytes demonstrated early afterdepolarizations (EADs) upon stimulation with isoproterenol at relatively long stimulation periods. Simulation of the hypertrophic myocyte activities revealed that the synergistic effects of the late Na+ current, the L-type Ca2+ current, and the T-type Ca2+ current were responsible for the initiation of EADs. The mechanisms of EAD and its suppression were investigated and sensitivity analysis was performed. Simulation results obtained with the hypertrophic cell model were compared to those from the normal ventricular myocytes. The developed mathematical model can be used for the explanation of the existing experimental data, for the development of the models for other hypertrophic phenotypes, and to make experimentally testable predictions of a hypertrophic myocyte's behavior.
Collapse
Affiliation(s)
- Dilmini Warnakulasooriya
- Department of Mathematics and Statistics, Georgia State University, 25 Park Place, Room 1346, Atlanta, GA, 30303-3083, USA
| | - Vladimir E Bondarenko
- Department of Mathematics and Statistics, Georgia State University, 25 Park Place, Room 1346, Atlanta, GA, 30303-3083, USA.
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
7
|
Huang J, Robert C, Ling L, Chen C, Hilal S. Cross-Sectional Association Between Left Ventricular Geometric Patterns, Cortical Cerebral Microinfarcts, and Cognition. J Am Heart Assoc 2025; 14:e035522. [PMID: 39818861 PMCID: PMC12054439 DOI: 10.1161/jaha.124.035522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/09/2024] [Indexed: 01/19/2025]
Abstract
BACKGROUND Left ventricular (LV) geometric patterns are associated with cognitive impairment and cerebral small vessel disease. As a novel magnetic resonance imaging marker of cerebral small vessel disease and a risk factor for cognitive dysfunction, cortical cerebral microinfarcts (CMIs) have been associated with heart disease through mechanisms including cardioembolism and cerebral hypoperfusion. Further investigation is required to determine whether cortical CMIs could arise from hemodynamic changes related to LV geometry, thus elucidating the connection between LV geometry and cognitive function. This study aims to investigate the cross-sectional relationship between LV geometric patterns, cortical CMIs, and cognition. METHODS AND RESULTS A total of 261 patients, aged 75.3±6.8 years, were enrolled from a memory clinic study. LV dimensions were obtained using 2-dimensional echocardiography. LV mass index and relative wall thickness were determined to categorize LV geometric patterns. Cortical CMIs were graded on 3T magnetic resonance imaging. Cognition was assessed using neuropsychological tests and clinical diagnosis. Linear, zero-inflated Poisson, and logistic regression were employed for continuous, count, and binary outcomes, respectively. Mediation analysis was conducted to examine the mediation effect of cortical CMIs. A significant association was observed between concentric LV hypertrophy and cortical CMI counts (rate ratio, 3.85 [95% CI, 1.85-8.01]), as well as the odds of having dementia (odds ratio, 2.86 [95% CI, 1.07-7.89]), in the fully adjusted model. Cortical CMIs may partly explain the relationship between concentric LV hypertrophy and cognitive function. CONCLUSIONS Concentric LV hypertrophy was associated with cortical CMIs and dementia. Further verification is warranted to determine the role of cortical CMIs in the association between LV geometry and cognition.
Collapse
Affiliation(s)
- Jiannan Huang
- Saw Swee Hock School of Public HealthNational University of Singapore and National University Health SystemSingapore
| | - Caroline Robert
- Department of Pharmacology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Lieng‐Hsi Ling
- Department of CardiologyNational University Heart CentreSingapore
- Department of Medicine, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Christopher Chen
- Department of Pharmacology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
- Memory Aging and Cognition CentreNational University Health SystemSingapore
| | - Saima Hilal
- Saw Swee Hock School of Public HealthNational University of Singapore and National University Health SystemSingapore
- Department of Pharmacology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
- Memory Aging and Cognition CentreNational University Health SystemSingapore
| |
Collapse
|
8
|
Salim AA, Kawasoe S, Kubozono T, Ojima S, Yamaguchi S, Higuchi K, Ikeda Y, Miyahara H, Tokushige K, Ohishi M. Association between serum uric acid levels and left ventricular hypertrophy based on electrocardiographic findings: a sex-specific analysis across cardiometabolic diseases. Sci Rep 2025; 15:2319. [PMID: 39833440 PMCID: PMC11746994 DOI: 10.1038/s41598-025-86497-8] [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: 06/03/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
The association between serum uric acid (UA) levels and left ventricular hypertrophy (LVH) remains unclear. We aimed to investigate this association using electrocardiographic findings. Health examination data from Kagoshima Kouseiren Hospital included 79,200 participants without cardiovascular diseases. Hypertension was defined as a blood pressure ≥ 140/90 mmHg. A Sokolow-Lyon criterion of ≥ 3.5 mm was used to define LVH. Sex-specific associations between UA and LVH were adjusted for age, body mass index, systolic blood pressure, serum triglyceride, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, fasting plasma glucose, estimated glomerular filtration rate, and lifestyle factors. The mean UA levels were 4.5 ± 1.1 and 6.1 ± 1.4 mg/dL in women and men, respectively (LVH prevalences, 4.0%, and 11.8%, respectively). Individuals without and with hypertension had mean UA levels of 5.2 ± 1.4 and 5.7 ± 1.5 mg/dL, respectively (LVH prevalences, 5.5%, and 14.4%, respectively). UA levels were significantly associated with LVH in women aged 40-49 and 50-59 years and in men aged 50-59 years. Compared with the first UA quartile, the fourth quartile showed a more significant association with LVH in individuals without hypertension. Furthermore, UA was associated with LVH in individuals without obesity, dyslipidemia, reduced kidney function, and diabetes in both sexes. Serum UA levels are associated with LVH in middle-aged women and men without cardiovascular disease risk, suggesting the potential role of UA as an LVH marker. Anwar Ahmed Salim and Shin Kawasoe contributed equally to this work.
Collapse
Affiliation(s)
- Anwar Ahmed Salim
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
- Department of Cardiology, Benjamin Mkapa Hospital, Dodoma, Tanzania
| | - Shin Kawasoe
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Takuro Kubozono
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan.
| | - Satoko Ojima
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Satoshi Yamaguchi
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Koji Higuchi
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Yoshiyuki Ikeda
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | | | | | - Mitsuru Ohishi
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
9
|
Xia X, Shan H, Jin Z, Ma T, Liu Y, Zhang J, Tian H, Dong B, Xu C, Chen S. TRIM26 exacerbates pathological cardiac hypertrophy by activating TAK1. Heliyon 2025; 11:e40653. [PMID: 39811317 PMCID: PMC11729661 DOI: 10.1016/j.heliyon.2024.e40653] [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: 06/03/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 01/16/2025] Open
Abstract
Pathological myocardial hypertrophy can induce heart failure with high mortality, it is necessary to explore its pathogenesis. Tripartite motif-containing 26 (TRIM26) belongs to the multidomain E3 ubiquitin ligase family. We observed increased expression of TRIM26 in the myocardium of C57BL/6 mice subjected to transverse aortic constriction (TAC) surgery and neonatal rat cardiomyocytes (NRCMs) treated with phenylephrine (PE). To evaluate the role of TRIM26 in pathological cardiac hypertrophy, we generated Trim26 global knockout mice and Trim26 overexpression adenoviruses. Mice with Trim26 deletion showed alleviated cardiomyocyte enlargement, inflammation, fibrosis, and cardiac dysfunction after TAC surgery. In PE-treated NRCMs, Trim26 overexpression promoted cardiomyocyte enlargement and inflammation, while Trim26 knockdown had the opposite effects. RNA sequencing and molecular biology methodologies were performed to identify targets conducive to TRIM26 function. The results showed that TRIM26 activated the transforming growth factor-beta activated kinase 1 (TAK1)-c-Jun N-terminal kinase/p38 signaling pathway in response to hypertrophic stress. Moreover, inhibition of TAK1 activation can reverse the promotion effect of TRIM26 overexpression on cardiomyocyte hypertrophy induced by PE stimulation in vitro. Our study demonstrated that TRIM26 plays an active role in pathological cardiac hypertrophy, and the TRIM26-TAK1 pathway may represent a therapeutic target for treating pathological cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- Xiaochuang Xia
- Department of Cardiology, Huanggang central Hospital of Yangtze University, Huanggang, China
| | - Huajing Shan
- Department of Cardiology, Huanggang central Hospital of Yangtze University, Huanggang, China
| | - Zhaoxia Jin
- Department of Cardiology, Huanggang central Hospital of Yangtze University, Huanggang, China
| | - Tengfei Ma
- Department of Neurosurgery, Huanggang central Hospital of Yangtze University, Huanggang, China
- Huanggang Institute of Translational Medicine, Huanggang, China
| | - Yemao Liu
- Department of Cardiology, Huanggang central Hospital of Yangtze University, Huanggang, China
- Huanggang Institute of Translational Medicine, Huanggang, China
| | - Jianqing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Han Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bizhen Dong
- Huanggang Institute of Translational Medicine, Huanggang, China
| | - Chengsheng Xu
- Department of Cardiology, Huanggang central Hospital of Yangtze University, Huanggang, China
| | - Shaoze Chen
- Department of Cardiology, Huanggang central Hospital of Yangtze University, Huanggang, China
| |
Collapse
|
10
|
Bracamonte JH, Watkins L, Pat B, Dell’Italia LJ, Saucerman JJ, Holmes JW. Contributions of mechanical loading and hormonal changes to eccentric hypertrophy during volume overload: a Bayesian analysis using logic-based network models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.612768. [PMID: 39345523 PMCID: PMC11429691 DOI: 10.1101/2024.09.12.612768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Primary mitral regurgitation (MR) is a pathology that alters mechanical loading on the left ventricle, triggers an array of compensatory neurohormonal responses, and induces a distinctive ventricular remodeling response known as eccentric hypertrophy. Drug therapies may alleviate symptoms, but only mitral valve repair or replacement can provide significant recovery of cardiac function and dimensions. Questions remain about the optimal timing of surgery, with 20% of patients developing systolic dysfunction post-operatively despite being treated according to the current guidelines. Thus, better understanding of the hypertrophic process in the setting of ventricular volume overload (VO) is needed to improve and better personalize the management of MR. To address this knowledge gap, we employ a Bayesian approach to combine data from 70 studies on experimental volume overload in dogs and rats and use it to calibrate a logic-based network model of hypertrophic signaling in myocytes. The calibrated model predicts that growth in experimental VO is mostly driven by the neurohormonal response, with an initial increase in myocardial tissue stretch being compensated by subsequent remodeling fairly early in the time course of VO. This observation contrasts with a common perception that volume-overload hypertrophy is driven primarily by increased myocyte strain. The model reproduces many aspects of 43 studies not used in its calibration, including infusion of individual hypertrophic agonists alone or in combination with various drugs commonly employed to treat heart failure, as well as administration of some of those drugs in the setting of experimental volume overload. We believe this represents a promising approach to using the known structure of an intracellular signaling network to integrate information from multiple studies into quantitative predictions of the range of expected responses to potential interventions in the complex setting of cardiac hypertrophy driven by a combination of hormonal and mechanical factors.
Collapse
Affiliation(s)
- Johane H. Bracamonte
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Lionel Watkins
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Betty Pat
- Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, United States of America
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Louis J. Dell’Italia
- Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, United States of America
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jeffrey J. Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jeffrey W. Holmes
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Division of Cardiothoracic Surgery, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
11
|
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
|
12
|
Abbas N, Bentele M, Waleczek FJG, Fuchs M, Just A, Pfanne A, Pich A, Linke S, Neumüller S, Stucki-Koch A, Jordan M, Perbellini F, Werlein C, Korte W, Ius F, Ruhparwar A, Weber N, Fiedler J, Thum T. Ex vivo modelling of cardiac injury identifies ferroptosis-related pathways as a potential therapeutic avenue for translational medicine. J Mol Cell Cardiol 2024; 196:125-140. [PMID: 39341589 PMCID: PMC7617241 DOI: 10.1016/j.yjmcc.2024.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 09/13/2024] [Accepted: 09/22/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Heart failure (HF) is a burgeoning health problem worldwide. Often arising as a result of cardiac injury, HF has become a major cause of mortality with limited availability of effective treatments. Ferroptotic pathways, triggering an iron-dependent form of cell death, are known to be potential key players in heart disease. This form of cell death does not exhibit typical characteristics of programmed cell death, and is mediated by impaired iron metabolism and lipid peroxidation signalling. OBJECTIVES The aim of this study is to establish an ex-vivo model of myocardial injury in living myocardial slices (LMS) and to identify novel underlying mechanisms and potential therapeutic druggable target(s). METHODS AND RESULTS In this study, we employed LMS as an ex vivo model of cardiac injury to investigate underlying mechanisms and potential therapeutic targets. Cryoinjury was induced in adult rat LMS, resulting in 30 % tissue damage. Cryoinjured LMS demonstrated impaired contractile function, cardiomyocyte hypertrophy, inflammation, and cardiac fibrosis, closely resembling in vivo cardiac injury characteristics. Proteomic analysis revealed an enrichment of factors associated with ferroptosis in the injured LMS, suggesting a potential causative role. To test this hypothesis, we pharmacologically inhibited ferroptotic pathways using ferrostatin (Fer-1) in the cryoinjured rat LMS, resulting in attenuation of structural changes and repression of pro-fibrotic processes. Furthermore, LMS generated from failing human hearts were used as a model of chronic heart failure. In this model, Fer-1 treatment was observed to reduce the expression of ferroptotic genes, enhances contractile function and improves tissue viability. Blocking ferroptosis-associated pathways in human cardiac fibroblasts (HCFs) resulted in a downregulation of fibroblast activation genes, a decrease in fibroblast migration capacity, and a reduction in reactive oxygen species production. RNA sequencing analysis of Fer-1-treated human LMS implicated metallothioneins as a potential underlying mechanism for the inhibition of these pathways. This effect is possibly mediated through the replenishment of glutathione reserves. CONCLUSIONS Our findings highlight the potential of targeting ferroptosis-related pathways and metallothioneins as a promising strategy for the treatment of heart disease.
Collapse
Affiliation(s)
- Naisam Abbas
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany; Fraunhofer Institute of Toxicology and Experimental Medicine (ITEM), Hannover, Germany; Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Hannover, Germany
| | - Marco Bentele
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany; Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Hannover, Germany
| | - Florian J G Waleczek
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany; Fraunhofer Institute of Toxicology and Experimental Medicine (ITEM), Hannover, Germany; Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Hannover, Germany
| | - Maximilian Fuchs
- Fraunhofer Institute of Toxicology and Experimental Medicine (ITEM), Hannover, Germany; Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Hannover, Germany
| | - Annette Just
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Angelika Pfanne
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Andreas Pich
- Institute of Toxicology and Core Unit Proteomics, Hannover Medical School, Hannover, Germany
| | - Sophie Linke
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Susanne Neumüller
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Angelika Stucki-Koch
- Fraunhofer Institute of Toxicology and Experimental Medicine (ITEM), Hannover, Germany; Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Hannover, Germany
| | - Maria Jordan
- Fraunhofer Institute of Toxicology and Experimental Medicine (ITEM), Hannover, Germany; Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Hannover, Germany
| | - Filippo Perbellini
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | | | - Wilhelm Korte
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Fabio Ius
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Arjang Ruhparwar
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Natalie Weber
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Jan Fiedler
- Fraunhofer Institute of Toxicology and Experimental Medicine (ITEM), Hannover, Germany; Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany; Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
13
|
Duse DA, Schröder NH, Srivastava T, Benkhoff M, Vogt J, Nowak MK, Funk F, Semleit N, Wollnitzke P, Erkens R, Kötter S, Meuth SG, Keul P, Santos W, Polzin A, Kelm M, Krüger M, Schmitt J, Levkau B. Deficiency of the sphingosine-1-phosphate (S1P) transporter Mfsd2b protects the heart against hypertension-induced cardiac remodeling by suppressing the L-type-Ca 2+ channel. Basic Res Cardiol 2024; 119:853-868. [PMID: 39110173 PMCID: PMC11461684 DOI: 10.1007/s00395-024-01073-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 10/09/2024]
Abstract
The erythrocyte S1P transporter Mfsd2b is also expressed in the heart. We hypothesized that S1P transport by Mfsd2b is involved in cardiac function. Hypertension-induced cardiac remodeling was induced by 4-weeks Angiotensin II (AngII) administration and assessed by echocardiography. Ca2+ transients and sarcomere shortening were examined in adult cardiomyocytes (ACM) from Mfsd2b+/+ and Mfsd2b-/- mice. Tension and force development were measured in skinned cardiac fibers. Myocardial gene expression was determined by real-time PCR, Protein Phosphatase 2A (PP2A) by enzymatic assay, and S1P by LC/MS, respectively. Msfd2b was expressed in the murine and human heart, and its deficiency led to higher cardiac S1P. Mfsd2b-/- mice had regular basal cardiac function but were protected against AngII-induced deterioration of left-ventricular function as evidenced by ~ 30% better stroke volume and cardiac index, and preserved ejection fraction despite similar increases in blood pressure. Mfsd2b-/- ACM exhibited attenuated Ca2+ mobilization in response to isoprenaline whereas contractility was unchanged. Mfsd2b-/- ACM showed no changes in proteins responsible for Ca2+ homeostasis, and skinned cardiac fibers exhibited reduced passive tension generation with preserved contractility. Verapamil abolished the differences in Ca2+ mobilization between Mfsd2b+/+ and Mfsd2b-/- ACM suggesting that S1P inhibits L-type-Ca2+ channels (LTCC). In agreement, intracellular S1P activated the inhibitory LTCC phosphatase PP2A in ACM and PP2A activity was increased in Mfsd2b-/- hearts. We suggest that myocardial S1P protects from hypertension-induced left-ventricular remodeling by inhibiting LTCC through PP2A activation. Pharmacologic inhibition of Mfsd2b may thus offer a novel approach to heart failure.
Collapse
Affiliation(s)
- Dragos Andrei Duse
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany
- Department of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
| | - Nathalie Hannelore Schröder
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany
| | - Tanu Srivastava
- Institute of Pharmacology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Marcel Benkhoff
- Department of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Jens Vogt
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany
| | - Melissa Kim Nowak
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany
| | - Florian Funk
- Institute of Pharmacology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Nina Semleit
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany
| | - Philipp Wollnitzke
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany
| | - Ralf Erkens
- Department of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
| | - Sebastian Kötter
- Institute of Cardiovascular Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Sven Günther Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Petra Keul
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany
| | - Webster Santos
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA, 24060, USA
| | - Amin Polzin
- Department of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
| | - Malte Kelm
- Department of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
| | - Martina Krüger
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
- Institute of Cardiovascular Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Joachim Schmitt
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
- Institute of Pharmacology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Bodo Levkau
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany.
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany.
| |
Collapse
|
14
|
Mkhize SA, Manilall A, Mokotedi L, Gunter S, Michel FS. Involvement of pentraxin-3 in the development of hypertension but not left ventricular hypertrophy in male spontaneously hypertensive rats. Physiol Rep 2024; 12:e70086. [PMID: 39414396 PMCID: PMC11483509 DOI: 10.14814/phy2.70086] [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: 03/21/2024] [Revised: 09/26/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024] Open
Abstract
Hypertension drives the development of concentric left ventricular hypertrophy (LVH). However, the relative contribution of pentraxin-3 (PTX-3), a novel marker for inflammatory cardiovascular disease, in the hypertrophic response to pressure overload has not been adequately elucidated. We investigated the role of PTX-3 in the development of LVH in spontaneously hypertensive rats (SHR), untreated and treated with either captopril (an ACE inhibitor) or hydralazine (a non-specific vasodilator). Three-month-old SHR received either 20 mg/kg/day hydralazine (SHR + H, n = 6), 40 mg/kg/day captopril (SHR + C, n = 6), or plain gelatine cubes (untreated SHR, n = 7) orally for 4 months. Wistar Kyoto rats (WKY, n = 7) were used as the normotensive controls. Blood pressure (BP) was measured using the tail-cuff method. Cardiac geometry and function were determined using M-mode echocardiography. Circulating concentrations of inflammatory markers were measured in plasma by ELISA. LV fibrosis and cardiomyocyte width were assessed by histology. Relative mRNA expression of PTX-3 was determined in the LV by RT-PCR. Untreated SHR exhibited greater systolic BP and relative wall thickness (RWT) compared to WKY. Captopril and hydralazine normalized BP but only captopril reversed RWT in SHR. Circulating PTX-3 and VCAM-1 levels were elevated in untreated SHR and reduced with captopril and hydralazine. Circulating PTX-3 was positively associated with systolic BP but lacked independent relations with indices of LVH. LV relative mRNA expression of PTX-3 was similar between the groups. PTX-3 may not be involved in the development of LVH in SHR, but plausibly reflects the localized inflammatory milieu associated with hypertension.
Collapse
Affiliation(s)
- Siluleko A. Mkhize
- Integrated Molecular Physiology Research Initiative, Faculty of Health Sciences, School of PhysiologyUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Ashmeetha Manilall
- Integrated Molecular Physiology Research Initiative, Faculty of Health Sciences, School of PhysiologyUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Lebogang Mokotedi
- Integrated Molecular Physiology Research Initiative, Faculty of Health Sciences, School of PhysiologyUniversity of the WitwatersrandJohannesburgSouth Africa
- Department of Physiology, School of MedicineSefako Makgatho Health Sciences UniversityPretoriaSouth Africa
| | - Sule Gunter
- Integrated Molecular Physiology Research Initiative, Faculty of Health Sciences, School of PhysiologyUniversity of the WitwatersrandJohannesburgSouth Africa
- Department of Health Sciences and MedicineBond UniversityGold CoastQueenslandAustralia
| | - Frederic S. Michel
- Integrated Molecular Physiology Research Initiative, Faculty of Health Sciences, School of PhysiologyUniversity of the WitwatersrandJohannesburgSouth Africa
| |
Collapse
|
15
|
Yoon JJ, Tai AL, Kim HY, Han BH, Shin S, Lee HS, Kang DG. TongGuanWan Alleviates Doxorubicin- and Isoproterenol-Induced Cardiac Hypertrophy and Fibrosis by Modulating Apoptotic and Fibrotic Pathways. Int J Mol Sci 2024; 25:10573. [PMID: 39408900 PMCID: PMC11476530 DOI: 10.3390/ijms251910573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Heart failure, a major public health issue, often stems from prolonged stress or damage to the heart muscle, leading to cardiac hypertrophy. This can progress to heart failure and other cardiovascular problems. Doxorubicin (DOX), a common chemotherapy drug, and isoproterenol (ISO), a β-adrenergic agonist, both induce cardiac hypertrophy through different mechanisms. This study investigates TongGuanWan (TGW,), a traditional herbal remedy, for its effects on cardiac hypertrophy and fibrosis in DOX-induced H9c2 cells and ISO-induced mouse models. TGW was found to counteract DOX-induced increases in H9c2 cell surface area (n = 8, p < 0.01) and improve biomarkers like ANP (n = 3, p < 0.01)) and BNP (n = 3, p < 0.01). It inhibited the MAPK pathway (n = 4, p < 0.01) and GATA-4/calcineurin/NFAT-3 signaling, reduced inflammation by decreasing NF-κB p65 translocation, and enhanced apoptosis-related factors such as caspase-3 (n = 3, p < 0.01), caspase-9 (n = 3, p < 0.01), Bax (n = 3, p < 0.01), and Bcl-2 (n = 3, p < 0.01). Flow cytometry showed TGW reduced apoptotic cell populations. In vivo, TGW reduced heart (n = 8~10, p < 0.01), and left ventricle weights (n = 6~7), cardiac hypertrophy markers (n = 3, p < 0.01), and perivascular fibrosis in ISO-induced mice, with Western blot analysis confirming decreased levels of fibrosis-related factors like fibronectin, α-SMA (n = 3, p < 0.05), and collagen type I (n = 3, p < 0.05). These findings suggest TGW has potential as a therapeutic option for cardiac hypertrophy and fibrosis.
Collapse
Affiliation(s)
- Jung-Joo Yoon
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, 460, Iksan-daero, Iksan 54538, Republic of Korea; (J.-J.Y.); (A.-L.T.); (H.-Y.K.); (B.-H.H.)
| | - Ai-Lin Tai
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, 460, Iksan-daero, Iksan 54538, Republic of Korea; (J.-J.Y.); (A.-L.T.); (H.-Y.K.); (B.-H.H.)
- College of Oriental Medicine and Professional Graduate School of Oriental Medicine, Wonkwang University, 460, Iksan-daero, Iksan 54538, Republic of Korea
| | - Hye-Yoom Kim
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, 460, Iksan-daero, Iksan 54538, Republic of Korea; (J.-J.Y.); (A.-L.T.); (H.-Y.K.); (B.-H.H.)
| | - Byung-Hyuk Han
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, 460, Iksan-daero, Iksan 54538, Republic of Korea; (J.-J.Y.); (A.-L.T.); (H.-Y.K.); (B.-H.H.)
| | - Sarah Shin
- KM Science Research Division, Korea Institute of Oriental Medicine, 1672, Yuseong-daero, Yuseong-gu, Daejeon 34054, Republic of Korea;
| | - Ho-Sub Lee
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, 460, Iksan-daero, Iksan 54538, Republic of Korea; (J.-J.Y.); (A.-L.T.); (H.-Y.K.); (B.-H.H.)
| | - Dae-Gill Kang
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, 460, Iksan-daero, Iksan 54538, Republic of Korea; (J.-J.Y.); (A.-L.T.); (H.-Y.K.); (B.-H.H.)
- College of Oriental Medicine and Professional Graduate School of Oriental Medicine, Wonkwang University, 460, Iksan-daero, Iksan 54538, Republic of Korea
| |
Collapse
|
16
|
Agbaje AO. Accelerometer-based sedentary time and physical activity from childhood through young adulthood with progressive cardiac changes: a 13-year longitudinal study. Eur J Prev Cardiol 2024; 31:1480-1492. [PMID: 38711312 PMCID: PMC11378265 DOI: 10.1093/eurjpc/zwae129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/27/2024] [Accepted: 04/04/2024] [Indexed: 05/08/2024]
Abstract
AIMS Longitudinal evidence on the relationship of sedentary time (ST), light-intensity physical activity (LPA), and moderate-to-vigorous-intensity physical activity (MVPA) with changes in cardiac structure and function in the paediatric population is scarce. This evidence is clinically important due to the impact ST can have on the long-term prognosis of healthy young population in the lifetime continuum. This prospective observational study examined the relationships of cumulative ST, LPA, and MVPA from childhood with longitudinal changes in cardiac structure and function. METHODS AND RESULTS This is a secondary analysis from the Avon Longitudinal Study of Parents and Children, UK birth cohort of 1682 children aged 11 years. Participants who had at least one follow-up timepoints accelerometer-measured ST, LPA, and MVPA over a period of 13 years and repeated echocardiography-measured cardiac structure and function at ages 17- and 24-year clinic visit were included. Left ventricular mass indexed for height2.7 (LVMI2.7) and left ventricular (LV) diastolic function from mitral E/A ratio (LVDF) were computed. Among 1682 children (mean [SD] age, 11.75 [0.24] years; 1054 [62.7%] females), the cumulative one-min/day increase in ST from ages 11 to 24 years was associated with progressively increased LVMI2.7 {effect estimate 0.002 g/m2.7 [confidence interval (CI) 0.001-0.003], P < 0.001}, irrespective of sex, obesity, and hypertensive status. Cumulative one-min/day increase in LPA was associated with a decreased LVMI2.7 (-0.005 g/m2.7 [-0.006 to -0.003], P < 0.0001) but an increased LVDF. Cumulative one-minute/day increase in MVPA was associated with progressively increased LVMI2.7 (0.003 g/m2.7 [0.001-0.006], P = 0.015). CONCLUSION ST contributed +40% to the 7-year increase in cardiac mass, MVPA increased cardiac mass by +5%, but LPA reduced cardiac mass by -49%. Increased ST may have long-term pathologic effects on cardiac structure and function during growth from childhood through young adulthood; however, engaging in LPA may enhance cardiac health in the young population.
Collapse
Affiliation(s)
- Andrew O Agbaje
- Clinical Epidemiology and Child Health Unit, Institute of Public Health and Clinical Nutrition, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 8, P.O. Box 1627, 70211 Kuopio, Finland
- Children’s Health and Exercise Research Centre, Department of Public Health and Sports Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| |
Collapse
|
17
|
Kageyama S, Taylor CA, Updegrove A, Garg S, Masuda S, Revaiah PC, Kageyama M, Tsai TY, Miyashita K, Tobe A, Tanaka K, De Mey J, La Meir M, Schneider U, Doenst T, Teichgräber U, Saima M, Pompilio G, Andreini D, Pontone G, Puskas JD, Gupta H, Morel MA, Serruys PW, Onuma Y. Cardiac computed tomography-derived coronary artery volume to myocardial mass in patients with severe coronary artery disease. J Cardiovasc Comput Tomogr 2024; 18:478-488. [PMID: 38944640 DOI: 10.1016/j.jcct.2024.06.010] [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: 03/14/2024] [Revised: 05/27/2024] [Accepted: 06/14/2024] [Indexed: 07/01/2024]
Abstract
BACKGROUND Coronary artery lumen volume (V) to myocardial mass (M) ratio (V/M) can show the mismatch between epicardial coronary arteries and the underlying myocardium. METHODS The V, M and V/M were obtained from the coronary computed tomography angiography (CCTA) of patients in the FAST-TRACK CABG study, the first-in-human trial of coronary artery bypass grafting (CABG) guided solely by CCTA and fractional flow reserve derived from CCTA (FFRCT) in patients with complex coronary artery disease (CAD). The correlations between V/M ratios and baseline characteristics were determined and compared with those from the ADVANCE registry, an unselected cohort of historical controls with chronic CAD. RESULTS The V/M ratio was obtained in 106 of the 114 pre-CABG CCTAs. Mean age was 65.6 years and 87% of them were male. The anatomical SYNTAX score from CCTA was significantly higher than the functional SYNTAX score derived using FFRCT [43.1 (15.2) vs 41.1 (16.5), p < 0.001]. Mean V, M, and V/M were 2204 mm3, 137 g, and 16.5 mm3/g, respectively. There were weak negative correlations between V and anatomical and functional SYNTAX scores (Pearson's r = -0.26 and -0.34). V and V/M had a strong correlation (r = 0.82). The V/M ratio in the current study was significantly lower than that in the ADVANCE registry (median 16.1 vs. 24.8 [1st quartile 20.1]). CONCLUSION Systematically smaller V/M ratios were found in this population with severe CAD requiring CABG compared to an unselected cohort with chronic CAD. The V/M ratio could provide additional non-invasive assessment of CAD especially when combined with FFRCT.
Collapse
Affiliation(s)
| | | | | | - Scot Garg
- Department of Cardiology, Royal Blackburn Hospital, Blackburn, United Kingdom
| | | | | | | | | | | | | | - Kaoru Tanaka
- Department of Radiology, University Hospital Brussels, Belgium
| | - Johan De Mey
- Universitair Ziekenhuis Brussel, VUB, Brussels, Belgium
| | - Mark La Meir
- Universitair Ziekenhuis Brussel, VUB, Brussels, Belgium
| | - Ulrich Schneider
- Department of Cardiothoracic Surgery, University Hospital Jena, Germany
| | - Torsten Doenst
- Department of Cardiothoracic Surgery, University Hospital Jena, Germany
| | | | | | - Giulio Pompilio
- Centro Cardiologico Monzino, IRCCS Monzino, Italy; Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milano, Italy
| | - Daniele Andreini
- Division of Cardiology and Cardiac Imaging, IRCCS Galeazzi Sant'Ambrogio, Milan, Italy; Department of Biomedical and Clinical Sciences, University of Milan, Italy
| | - Gianluca Pontone
- Centro Cardiologico Monzino, IRCCS Monzino, Italy; Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milano, Italy
| | - John D Puskas
- Department of Cardiothoracic Surgery, Emory University Hospital Midtown, USA
| | - Himanshu Gupta
- Department of Radiology, The Valley Hospital, Ridgewood, NJ, USA
| | | | | | | |
Collapse
|
18
|
Coelho P, Mascarenhas K, Rodrigues J, Rodrigues F. Investigation of Electrocardiographic Changes in Individuals with Three or More Cardiovascular Risk Factors on Santiago Island-The Cross-Sectional PrevCardio.CV Study. J Pers Med 2024; 14:876. [PMID: 39202067 PMCID: PMC11355878 DOI: 10.3390/jpm14080876] [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: 07/25/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
Cerebrocardiovascular diseases represent one of the greatest public health concerns globally. In Cabo Verde, non-communicable diseases, such as cerebrocardiovascular diseases, have become leading causes of morbidity and mortality. This study aimed to correlate risk factors with cardiac electrical changes in adult individuals residing on Santiago Island-Cabo Verde. A cross-sectional population-based study using simple random sampling was conducted in 2021 with individuals aged 18 and over, of both sexes, having authorization 35/2021 from the Cabo Verde Ethics Commission. The sample size was calculated based on Santiago Island's projected population for 2021, considering an estimated prevalence of 50%, a 95% confidence interval, and a standard error of 4%, resulting in a sample of 599 individuals. The data were collected through a questionnaire on risk factors and cerebrocardiovascular diseases, blood pressure measurement, capillary blood glucose evaluation, and a 12-lead electrocardiogram. The study sample was predominantly female (54.8%), with the largest age group being 18-27 years (21%). Among the sample, 9.3% had no risk factors, 27.5% had one risk factor, 36.2% had two risk factors, and 26.9% had three or more risk factors. Of those who underwent electrocardiography, 60.24% showed electrocardiographic changes, with the most prevalent being ventricular repolarization changes, nonspecific repolarization changes, and early repolarization. A relationship was observed between cerebrocardiovascular disease risk factors and the electrocardiographic changes found in the study participants.
Collapse
Affiliation(s)
- Patrícia Coelho
- Sport Physical Activity and Health Research & Innovation Center (Sprint), Polytechnic Institute of Castelo Branco, 6000-084 Castelo Branco, Portugal;
| | - Kelly Mascarenhas
- Polytechnic Institute of Castelo Branco, 6000-084 Castelo Branco, Portugal;
| | - Júlio Rodrigues
- Instituto Nacional de Saúde Pública de Cabo Verde, Cidade da Praia 7600, Cape Verde;
| | - Francisco Rodrigues
- Sport Physical Activity and Health Research & Innovation Center (Sprint), Polytechnic Institute of Castelo Branco, 6000-084 Castelo Branco, Portugal;
| |
Collapse
|
19
|
Perez-Bonilla P, LaViolette B, Bhandary B, Ullas S, Chen X, Hirenallur-Shanthappa D. Isoproterenol induced cardiac hypertrophy: A comparison of three doses and two delivery methods in C57BL/6J mice. PLoS One 2024; 19:e0307467. [PMID: 39038017 PMCID: PMC11262646 DOI: 10.1371/journal.pone.0307467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 07/05/2024] [Indexed: 07/24/2024] Open
Abstract
Heart Failure (HF) continues to be a complex public health issue with increasing world population prevalence. Although overall mortality has decreased for HF and hypertrophic cardiomyopathy (HCM), a precursor for HF, their prevalence continues to increase annually. Because the etiology of HF and HCM is heterogeneous, it has been difficult to identify novel therapies to combat these diseases. Isoproterenol (ISP), a non-selective β-adrenoreceptor agonist, is commonly used to induce cardiotoxicity and cause acute and chronic HCM and HF in mice. However, the variability in dose and duration of ISP treatment used in studies has made it difficult to determine the optimal combination of ISP dose and delivery method to develop a reliable ISP-induced mouse model for disease. Here we examined cardiac effects induced by ISP via subcutaneous (SQ) and SQ-minipump (SMP) infusions across 3 doses (2, 4, and 10mg/kg/day) over 2 weeks to determine whether SQ and SMP ISP delivery induced comparable disease severity in C57BL/6J mice. To assess disease, we measured body and heart weight, surface electrocardiogram (ECG), and echocardiography recordings. We found all 3 ISP doses comparably increase heart weight, but these increases are more pronounced when ISP was administered via SMP. We also found that the combination of ISP treatment and delivery method induces contrasting heart rate, RR interval, and R and S amplitudes that may place SMP treated mice at higher risk for sustained disease burden. Mice treated via SMP also had increased heart wall thickness and LV Mass, but mice treated via SQ showed greater increase in gene markers for hypertrophy and fibrosis. Overall, these data suggest that at 2 weeks, mice treated with 2, 4, or 10mg/kg/day ISP via SQ and SMP routes cause similar pathological heart phenotypes but highlight the importance of drug delivery method to induce differing disease pathways.
Collapse
Affiliation(s)
- Patricia Perez-Bonilla
- Global Discovery, Investigative & Translational Sciences–Animal Models and Imaging, Pfizer Inc, Cambridge, Massachusetts, United States of America
| | - Brianna LaViolette
- Global Discovery, Investigative & Translational Sciences–Animal Models and Imaging, Pfizer Inc, Cambridge, Massachusetts, United States of America
| | - Bidur Bhandary
- Rare Diseases Research Unit, Pfizer Inc, Cambridge, Massachusetts, United States of America
| | - Soumya Ullas
- Global Discovery, Investigative & Translational Sciences–Animal Models and Imaging, Pfizer Inc, Cambridge, Massachusetts, United States of America
| | - Xian Chen
- Global Discovery, Investigative & Translational Sciences–Animal Models and Imaging, Pfizer Inc, Cambridge, Massachusetts, United States of America
| | - Dinesh Hirenallur-Shanthappa
- Global Discovery, Investigative & Translational Sciences–Animal Models and Imaging, Pfizer Inc, Cambridge, Massachusetts, United States of America
| |
Collapse
|
20
|
DeFreitas MJ, Shelton EL, Schmidt AF, Ballengee S, Tian R, Chen P, Sharma M, Levine A, Katz ED, Rojas C, Abitbol CL, Hunter J, Kulandavelu S, Wu S, Young KC, Benny M. Neonatal hyperoxia exposure leads to developmental programming of cardiovascular and renal disease in adult rats. Sci Rep 2024; 14:16742. [PMID: 39033222 PMCID: PMC11271593 DOI: 10.1038/s41598-024-65844-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 06/25/2024] [Indexed: 07/23/2024] Open
Abstract
Premature infants are often exposed to hyperoxia. However, there is limited data regarding the mechanistic underpinnings linking neonatal hyperoxia exposure and its contribution to cardio-renal dysfunction in adults born preterm. Our objective was to determine whether neonatal hyperoxia induces systemic vascular stiffness and cardio-renal dysfunction in adulthood. Newborn rats were randomly assigned to room air (RA) or hyperoxia (85% O2) from postnatal day 1 to 14, then recovered in RA until 1 year of life. Arterial stiffness, cardio-renal histomorphometry, and fibrosis in the aorta, heart, and kidney were assessed. RNA-sequencing (RNA-seq) of the aorta and kidney was also done. Adult rats exposed to neonatal hyperoxia had increased aortic and mesenteric artery stiffness as demonstrated by wire and pressure myography. They also had cardiomyocyte hypertrophy, glomerulomegaly, and tubular injury. Hyperoxia exposure altered the transcriptome profile associated with fibrosis and matrix remodeling in the aorta and kidney. There was also increased TGF-β1 levels and fibrosis in the aorta, left ventricle, and kidney. In conclusion, neonatal hyperoxia exposure was associated with systemic vascular and cardio-renal alterations in 1-year-old rats. Further studies to determine how targeted therapies could reprogram cardio-renal injury after neonatal hyperoxia exposure are indicated.
Collapse
Affiliation(s)
- Marissa J DeFreitas
- Department of Pediatrics/Division of Nephrology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Elaine L Shelton
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Augusto F Schmidt
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Sydne Ballengee
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Runxia Tian
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - PingPing Chen
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Mayank Sharma
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Amanda Levine
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Emily Davidovic Katz
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Claudia Rojas
- Department of Pathology, Memorial Healthcare Systems, Hollywood, FL, USA
| | - Carolyn L Abitbol
- Department of Pediatrics/Division of Nephrology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juanita Hunter
- Department of Pediatrics/Division of Cardiology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Shathiyah Kulandavelu
- Department of Pediatrics/Division of Nephrology, University of Miami Miller School of Medicine, Miami, FL, USA
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Shu Wu
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Karen C Young
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Merline Benny
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA.
| |
Collapse
|
21
|
Guler GB, Guler A, Tanboga IH, Turkmen I, Atmaca S, Sahin H, Tekin M, Karakurt ST, Erin F, Inan D, Cinli TA, Akkas BE, Cansever AT, Erturk M. Ongoing assertion of two-dimensional measurements on differentiation type of left ventricular hypertrophy: Focus on inferior vena cava. Echocardiography 2024; 41:e15880. [PMID: 38979714 DOI: 10.1111/echo.15880] [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: 05/11/2024] [Revised: 06/11/2024] [Accepted: 06/16/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND Left ventricular hypertrophy (LVH), including hypertensive LVH, hypertrophic cardiomyopathy (HCM) and cardiac amyloidosis (CA), is a commonly encountered condition in cardiology practice, presenting challenges in differential diagnosis. In this study, we aimed to investigate the importance of echocardiographic evaluation of the inferior vena cava (IVC) in distinguishing LVH subtypes including hypertensive LVH, HCM, and CA. METHODS In this retrospective study, patients with common causes of LVH including hypertensive LVH, HCM, and CA were included. The role of echocardiographic evaluation of IVC diameter and collapsibility in distinguishing these causes of LVH was assessed in conjunction with other echocardiographic, clinical, and imaging methods. RESULTS A total of 211 patients (45% HCM, 43% hypertensive heart disease, and 12% CA) were included in our study. Their mean age was 56.6 years and 62% of them were male. While mean IVC diameter was significantly dilated in CA patients (13.4 mm in hypertensive LVH, 16.0 mm in HCM, and 21.1 mm in CA, p < .001), its collapsibility was reduced (IVC collapsible in 95% of hypertensive patients, 72% of HCM patients, and 12% of CA patients, p < .001). In the analysis of diagnostic probabilities, the presence of both hypovoltage and IVC dilation is significant for CA patients. Although it is not statistically significant, the presence of IVC dilation along with atrial fibrillation supports the diagnosis of HCM. CONCLUSION In conclusion, although advances in imaging techniques facilitate the diagnosis of LVH, simple echocardiographic methods should never be overlooked. Our study supports the notion that IVC assessment could play an important role in the differential diagnosis of LVH.
Collapse
Affiliation(s)
- Gamze Babur Guler
- Department of Cardiology, University of Health Sciences, Mehmet Akif Ersoy Thoracic Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Arda Guler
- Department of Cardiology, University of Health Sciences, Mehmet Akif Ersoy Thoracic Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Ibrahim Halil Tanboga
- Department of Cardiology & Biostatistics, Istanbul Nisantasi University Medical School, Istanbul, Turkey
| | - Irem Turkmen
- Department of Cardiology, University of Health Sciences, Mehmet Akif Ersoy Thoracic Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Sezgin Atmaca
- Department of Cardiology, University of Health Sciences, Mehmet Akif Ersoy Thoracic Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Hasan Sahin
- Department of Cardiology, University of Health Sciences, Mehmet Akif Ersoy Thoracic Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Meltem Tekin
- Department of Cardiology, University of Health Sciences, Mehmet Akif Ersoy Thoracic Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Seda Tukenmez Karakurt
- Department of Cardiology, University of Health Sciences, Mehmet Akif Ersoy Thoracic Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Faruk Erin
- Bahcesehir University Faculty of Medicine, Istanbul, Turkey
| | - Duygu Inan
- Department of Cardiology, University of Health Sciences, Basaksehir Cam Sakura City Hospital, Istanbul, Turkey
| | - Tahir Alper Cinli
- Department of Hematology, University of Health Sciences, Van Training and Research Hospital, Van, Turkey
| | - Burcu Esen Akkas
- Department of Nuclear Medicine, University of Health Sciences, Basaksehir Cam Sakura City Hospital, Istanbul, Turkey
| | - Aysel Turkvatan Cansever
- Department of Radiology, University of Health Sciences, Mehmet Akif Ersoy Thoracic Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Mehmet Erturk
- Department of Cardiology, University of Health Sciences, Mehmet Akif Ersoy Thoracic Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
22
|
Pardon G, Vander Roest AS, Chirikian O, Birnbaum F, Lewis H, Castillo EA, Wilson R, Denisin AK, Blair CA, Holbrook C, Koleckar K, Chang ACY, Blau HM, Pruitt BL. Tracking single hiPSC-derived cardiomyocyte contractile function using CONTRAX an efficient pipeline for traction force measurement. Nat Commun 2024; 15:5427. [PMID: 38926342 PMCID: PMC11208611 DOI: 10.1038/s41467-024-49755-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) are powerful in vitro models to study the mechanisms underlying cardiomyopathies and cardiotoxicity. Quantification of the contractile function in single hiPSC-CMs at high-throughput and over time is essential to disentangle how cellular mechanisms affect heart function. Here, we present CONTRAX, an open-access, versatile, and streamlined pipeline for quantitative tracking of the contractile dynamics of single hiPSC-CMs over time. Three software modules enable: parameter-based identification of single hiPSC-CMs; automated video acquisition of >200 cells/hour; and contractility measurements via traction force microscopy. We analyze >4,500 hiPSC-CMs over time in the same cells under orthogonal conditions of culture media and substrate stiffnesses; +/- drug treatment; +/- cardiac mutations. Using undirected clustering, we reveal converging maturation patterns, quantifiable drug response to Mavacamten and significant deficiencies in hiPSC-CMs with disease mutations. CONTRAX empowers researchers with a potent quantitative approach to develop cardiac therapies.
Collapse
Grants
- R00 HL153679 NHLBI NIH HHS
- K99 HL153679 NHLBI NIH HHS
- RM1 GM131981 NIGMS NIH HHS
- 20POST35211011 American Heart Association (American Heart Association, Inc.)
- 17CSA33590101 American Heart Association (American Heart Association, Inc.)
- 18CDA34110411 American Heart Association (American Heart Association, Inc.)
- 1R21HL13099301 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 18POST34080160 American Heart Association (American Heart Association, Inc.)
- 1F31HL158227 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- F31 HL158227 NHLBI NIH HHS
- 201411MFE-338745-169197 Gouvernement du Canada | Canadian Institutes of Health Research (Instituts de Recherche en Santé du Canada)
- P2SKP2_164954 Schweizerischer Nationalfonds zur Förderung der Wissenschaftlichen Forschung (Swiss National Science Foundation)
- 13POST14480004 American Heart Association (American Heart Association, Inc.)
- RM1GM131981 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 82070248 National Natural Science Foundation of China (National Science Foundation of China)
- P400PM_180825 Schweizerischer Nationalfonds zur Förderung der Wissenschaftlichen Forschung (Swiss National Science Foundation)
- U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- Shanghai Pujiang Program 19PJ1407000 Program for Professor of Special Appointment (Eastern Scholar) at Shanghai Institutions of Higher Learning 0900000024 to A.C.Y.C. Innovative Research Team of High-Level Local Universities in Shanghai (A.C.Y.C.)
- the Baxter Foundation, Li Ka Shing Foundation and The Stanford Cardiovascular Institute
Collapse
Affiliation(s)
- Gaspard Pardon
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Departments of Bioengineering and Mechanical Engineering, University of California, Santa Barbara, CA, USA
- School of Life Sciences, EPFL École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Alison S Vander Roest
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Engineering, Michigan Engineering, University of Michigan Ann Arbor, MI, USA
| | - Orlando Chirikian
- Biomolecular Science and Engineering Program, University of California, Santa Barbara, CA, USA
| | - Foster Birnbaum
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Henry Lewis
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA
| | - Erica A Castillo
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA
- Departments of Bioengineering and Mechanical Engineering, University of California, Santa Barbara, CA, USA
| | - Robin Wilson
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA
| | - Aleksandra K Denisin
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA
| | - Cheavar A Blair
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA
- Departments of Bioengineering and Mechanical Engineering, University of California, Santa Barbara, CA, USA
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Colin Holbrook
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kassie Koleckar
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Alex C Y Chang
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Shanghai Institute of Precision Medicine and Department of Cardiology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Helen M Blau
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Beth L Pruitt
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Departments of Bioengineering and Mechanical Engineering, University of California, Santa Barbara, CA, USA.
- Biomolecular Science and Engineering Program, University of California, Santa Barbara, CA, USA.
| |
Collapse
|
23
|
Kim HY, Shin S, Yoon JJ, Ahn YM, Song JH, Lee DS, Park JY, Lee HS, Jung J. Exploring the potential effect of electroacupuncture on cardiovascular function and lipid profiles in spontaneously hypertensive rats. Integr Med Res 2024; 13:101041. [PMID: 38948488 PMCID: PMC11214362 DOI: 10.1016/j.imr.2024.101041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 07/02/2024] Open
Abstract
Background Investigating the effects of electroacupuncture (EA) treatment on cardiovascular function and aortic lipid profiles in spontaneously hypertensive rats (SHR) constitutes the foundational focus of this study. The overarching goal is to comprehensively elucidate the alterations brought about by EA treatment and to assess its potential as an alternative therapy for hypertension. Methods Consecutive EA treatments were administered to SHR, and the effects on systolic blood pressure, cardiac function, and hypertension-related neuronal signals were assessed. Aortic lipid profiles in vehicle-treated SHR and EA-treated SHR groups were analyzed using mass spectrometry-based lipid profiling. Additionally, the expression of Cers2 and GNPAT, enzymes involved in the synthesis of specific aortic lipids, was examined. Results The study demonstrated that consecutive EA treatments restored systolic blood pressure, improved cardiovascular function, and normalized hypertension-related neuronal signals in SHR. Analysis of the aortic lipid profiles revealed distinct differences between the vehicle-treated SHR group and the EA-treated SHR group. Specifically, EA treatment significantly altered the levels of aortic sphingomyelin and phospholipids, including very long-chain fatty acyl-ceramides and ether phosphatidylcholines. These changes in aortic lipid profiles correlated significantly with systolic blood pressure and cardiac function indicators. Furthermore, EA treatment significantly altered the expression of Cers2 and GNPAT. Conclusions The findings suggest that EA may influence cardiovascular functions and aortic lipid profiles in SHR.
Collapse
Affiliation(s)
- Hye-Yoom Kim
- Hanbang Cardio-renal Research Center & Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan, South Korea
| | - Sarah Shin
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Jung-Joo Yoon
- Hanbang Cardio-renal Research Center & Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan, South Korea
| | - You-Mee Ahn
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Ji-Hye Song
- College of Korean Medicine, Daejeon University, Daejeon, South Korea
| | - Da-Som Lee
- College of Korean Medicine, Daejeon University, Daejeon, South Korea
| | - Ji-Yeun Park
- College of Korean Medicine, Daejeon University, Daejeon, South Korea
| | - Ho-Sub Lee
- Hanbang Cardio-renal Research Center & Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan, South Korea
| | - Jeeyoun Jung
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea
| |
Collapse
|
24
|
Brown K, Williams Louie E, Pinn-Woodcock T, Pearson E, Pearson GB, Marr J, Hackett ES, Rath Brown L, Mitchell KJ. Cardiac Disease Related to Primary Hyperthyroidism in a 20-Year-Old Mule. Animals (Basel) 2024; 14:1660. [PMID: 38891707 PMCID: PMC11171152 DOI: 10.3390/ani14111660] [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: 04/02/2024] [Revised: 05/22/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Primary hyperthyroidism is a rarely diagnosed endocrinopathy in equids and there have been no previous reports of structural and functional cardiac changes associated with hyperthyroidism in these species. This case report investigates a 20-year-old mule gelding that presented for a three-month history of thin body condition despite polyphagia, with a heart murmur and elevated free and total thyroid hormone concentrations. On presentation, physical exam revealed a body condition score of two out of nine, persistent tachycardia, pansystolic heart murmur and firm bilateral ventral proximal cervical masses. Bloodwork confirmed markedly elevated free T4, total T4 and T3 concentrations. Echocardiogram demonstrated left ventricular concentric hypertrophy with increased ventricular and atrial systolic function. Bilateral thyroidectomy was performed under standing sedation without complications. Histopathology demonstrated adenocarcinoma of the left thyroid gland and multiple adenomas with osseous metaplasia within the right thyroid. The mule was supplemented with levothyroxine sodium two weeks post-op after a thyroid panel demonstrated undetectable concentrations. Polyphagia resolved following surgery and the mule began gaining weight. Echocardiographic changes improved but did not resolve at two years post-operative. Continued bi-annual follow up and monitoring of thyroid levels was recommended. This case represents the first documentation of hemodynamically relevant cardiac remodeling in an equid associated with primary hyperthyroidism.
Collapse
Affiliation(s)
- Kaitlin Brown
- Department of Clinical Sciences, Cornell University, Ithaca, NY 14850, USA; (K.B.); (E.W.L.); (T.P.-W.); (E.P.); (G.B.P.); (E.S.H.)
| | - Elizabeth Williams Louie
- Department of Clinical Sciences, Cornell University, Ithaca, NY 14850, USA; (K.B.); (E.W.L.); (T.P.-W.); (E.P.); (G.B.P.); (E.S.H.)
| | - Toby Pinn-Woodcock
- Department of Clinical Sciences, Cornell University, Ithaca, NY 14850, USA; (K.B.); (E.W.L.); (T.P.-W.); (E.P.); (G.B.P.); (E.S.H.)
| | - Erin Pearson
- Department of Clinical Sciences, Cornell University, Ithaca, NY 14850, USA; (K.B.); (E.W.L.); (T.P.-W.); (E.P.); (G.B.P.); (E.S.H.)
| | - Garett B. Pearson
- Department of Clinical Sciences, Cornell University, Ithaca, NY 14850, USA; (K.B.); (E.W.L.); (T.P.-W.); (E.P.); (G.B.P.); (E.S.H.)
| | - Jacqueline Marr
- New Hampshire Veterinary Diagnostic Laboratory, University of New Hampshire, Durham, NH 03824, USA;
| | - Eileen S. Hackett
- Department of Clinical Sciences, Cornell University, Ithaca, NY 14850, USA; (K.B.); (E.W.L.); (T.P.-W.); (E.P.); (G.B.P.); (E.S.H.)
| | | | - Katharyn J. Mitchell
- Department of Clinical Sciences, Cornell University, Ithaca, NY 14850, USA; (K.B.); (E.W.L.); (T.P.-W.); (E.P.); (G.B.P.); (E.S.H.)
| |
Collapse
|
25
|
Cheng T, Liu C, Wang Y, Li G, Feng L, Zhang S, Qi B, Cui J, Guo L, Cao L, Wang Y, Qi Z, Yang L. A novel histone deacetylase inhibitor Se-SAHA attenuates isoproterenol-induced heart failure via antioxidative stress and autophagy inhibition. Toxicol Appl Pharmacol 2024; 487:116957. [PMID: 38735590 DOI: 10.1016/j.taap.2024.116957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/27/2024] [Accepted: 05/06/2024] [Indexed: 05/14/2024]
Abstract
Heart failure is associated with histone deacetylase (HDAC) regulation of gene expression, the inhibition of which is thought to be beneficial for heart failure therapy. Here, we explored the cardioprotective effects and underlying mechanism of a novel selenium-containing HDAC inhibitor, Se-SAHA, on isoproterenol (ISO)-induced heart failure. We found that pretreatment with Se-SAHA attenuated ISO-induced cardiac hypertrophy and fibrosis in neonatal rat ventricular myocytes (NRVMs). Se-SAHA significantly attenuated the generation of ISO-induced reactive oxygen species (ROS) and restored the expression levels of superoxide dismutase 2 (SOD2) and heme oxygenase-1 (HO-1) in vitro. Furthermore, Se-SAHA pretreatment prevented the accumulation of autophagosomes. Se-SAHA reversed the high expression of HDAC1 and HDAC6 induced by ISO incubation. However, after the addition of the HDAC agonist, the effect of Se-SAHA on blocking autophagy was inhibited. Using ISO-induced mouse models, cardiac ventricular contractile dysfunction, hypertrophy, and fibrosis was reduced treated by Se-SAHA. In addition, Se-SAHA inhibited HDAC1 and HDAC6 overexpression in ISO-treated mice. Se-SAHA treatment significantly increased the activity of SOD2 and improved the ability to eliminate free radicals. Se-SAHA hindered the excessive levels of the microtubule-associated protein 1 light chain 3 (LC3)-II and Beclin-1 in heart failure mice. Collectively, our results indicate that Se-SAHA exerts cardio-protection against ISO-induced heart failure via antioxidative stress and autophagy inhibition.
Collapse
Affiliation(s)
- Tianwei Cheng
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Chang Liu
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Yufei Wang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Guangru Li
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Lifeng Feng
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Shengzheng Zhang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Bing Qi
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Jianlin Cui
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Lihong Guo
- Institute of Digestive Disease, Shengli Oilfield Central Hospital, Dongying 257000, China
| | - Lei Cao
- Department of General Surgery, Tianjin Union Medical Center, Tianjin 300122, China
| | - Yanming Wang
- College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, China.
| | - Zhi Qi
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China; Institute of Digestive Disease, Shengli Oilfield Central Hospital, Dongying 257000, China; Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin 300122, China.
| | - Liang Yang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China; Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin 300122, China.
| |
Collapse
|
26
|
Chichareon P, Methavigul K, Lip GYH, Krittayaphong R. Systolic blood pressure visit-to-visit variability and outcomes in Asian patients with atrial fibrillation. Hypertens Res 2024; 47:1479-1489. [PMID: 38438726 DOI: 10.1038/s41440-024-01592-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 12/27/2023] [Accepted: 01/13/2024] [Indexed: 03/06/2024]
Abstract
We aimed to assess the association between SBP-VVV and outcomes in Asian patients with atrial fibrillation (AF). AF patients in the COOL-AF registry with SBP measured at baseline, and at least two other visits were studied. We defined SBP-VVV using the standard deviation (SD) of average SBP. Patients were categorized according to the quartiles of SBP SD. The associations between SBP-VVV and outcomes were assessed in the adjusted Cox model. We studied 3172 patients (mean age 67.7 years; 41.8% female), with the prevalence of hypertension being 69%. Warfarin was used in 69% of patients, whereas 7% received non-vitamin K antagonist oral anticoagulants. The minimum and maximum SD of average SBP in the study population was 0.58 and 56.38 mmHg respectively. The cutoff of SD of average SBP for each quartile in our study were 9.09, 12.15, and 16.21 mmHg. The rates of all-cause mortality, ischemic stroke or systemic embolization (SSE), major bleeding, and intracranial hemorrhage (ICH) were 3.10, 1.42, 2.09, and 0.64 per 100 person-years, respectively. Compared with the first quartile, patients in the fourth quartile had a significantly higher risk of mortality (adjusted HR 1.60, 95%CI 1.13-2.25), bleeding (aHR 1.92, 95%CI 1.25-2.96) and ICH (aHR 3.51, 95%CI 1.40-8.76). The risk of SSE was not significantly different among the quartiles. SBP-VVV had a significant impact on the long-term outcomes of Asian patients with AF, particularly mortality and bleeding. Adequate SBP control and maintaining SBP stability over time may improve outcomes for AF patients.
Collapse
Affiliation(s)
- Ply Chichareon
- Cardiology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Komsing Methavigul
- Department of Cardiology, Central Chest Institute of Thailand, Nonthaburi, Thailand
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University, and Liverpool Heart & Chest Hospital, Liverpool, UK
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Rungroj Krittayaphong
- Division of Cardiology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
27
|
Chu HW, Hwang IC, Kim HM, Park J, Choi H, Choi HM, Yoon YE, Cho GY. Age-dependent implications of left ventricular hypertrophy regression in patients with hypertension. Hypertens Res 2024; 47:1144-1156. [PMID: 38238511 DOI: 10.1038/s41440-023-01571-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 11/27/2023] [Accepted: 12/16/2023] [Indexed: 03/13/2024]
Abstract
Left ventricular hypertrophy (LVH) is a significant risk factor for cardiovascular mortality and morbidity in patients with hypertension. However, the effect of age on LVH regression or persistence and its differential prognostic value remain unclear. Therefore, we investigated the clinical implications of LVH regression in 1847 patients with hypertension and echocardiography data (at baseline and during antihypertensive treatment at an interval of 6-18 months) according to age. LVH was defined as a left ventricular mass index (LVMI) > 115 g/m2 and >95 g/m2 in men and women, respectively. LVH prevalence at baseline was not different according to age (age < 65 years: 42.6%; age ≥65 years: 45.7%; p = 0.187), but LVH regression was more frequently observed in the younger group (36.4% vs. 27.5%; p = 0.008). Spline curves and multiple linear regression analysis showed a significant relationship between reductions in systolic blood pressure and LVMI in the younger group (β = 0.425; p < 0.001), but not the elderly group (β = 0.044; p = 0.308). LVH regression was associated with a lower risk of the study outcome (composite of cardiovascular death and hospitalization for heart failure) regardless of age. In conclusion, the association between the reduction in blood pressure and LVH regression was prominent in patients with age < 65 years, but not in those with age ≥65 years. However, an association between LVH regression and lower risk of cardiovascular death and hospitalization for heart failure was observed regardless of patient age, suggesting the prognostic value of the LVH regression not only in the younger patients but also in elderly patients.
Collapse
Affiliation(s)
- Hyun-Wook Chu
- Department of Cardiology, Cardiovascular Center, Seoul National University Bundang Hospital, Seongnam, Gyeonggi, South Korea
| | - In-Chang Hwang
- Department of Cardiology, Cardiovascular Center, Seoul National University Bundang Hospital, Seongnam, Gyeonggi, South Korea.
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea.
| | - Hyue Mee Kim
- Division of Cardiology, Department of Internal Medicine, Chung-Ang University Hospital, Seoul, South Korea.
| | - Jiesuck Park
- Department of Cardiology, Cardiovascular Center, Seoul National University Bundang Hospital, Seongnam, Gyeonggi, South Korea
| | - Hyejung Choi
- Department of Cardiology, Cardiovascular Center, Seoul National University Bundang Hospital, Seongnam, Gyeonggi, South Korea
| | - Hong-Mi Choi
- Department of Cardiology, Cardiovascular Center, Seoul National University Bundang Hospital, Seongnam, Gyeonggi, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Yeonyee E Yoon
- Department of Cardiology, Cardiovascular Center, Seoul National University Bundang Hospital, Seongnam, Gyeonggi, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Goo-Yeong Cho
- Department of Cardiology, Cardiovascular Center, Seoul National University Bundang Hospital, Seongnam, Gyeonggi, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
28
|
Garg P, Grafton-Clarke C, Matthews G, Swoboda P, Zhong L, Aung N, Thomson R, Alabed S, Demirkiran A, Vassiliou VS, Swift AJ. Sex-specific cardiac magnetic resonance pulmonary capillary wedge pressure. EUROPEAN HEART JOURNAL OPEN 2024; 4:oeae038. [PMID: 38751456 PMCID: PMC11095051 DOI: 10.1093/ehjopen/oeae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/23/2024] [Accepted: 04/28/2024] [Indexed: 05/18/2024]
Abstract
Aims Heart failure (HF) with preserved ejection fraction disproportionately affects women. There are no validated sex-specific tools for HF diagnosis despite widely reported differences in cardiac structure. This study investigates whether sex, as assigned at birth, influences cardiac magnetic resonance (CMR) assessment of left ventricular filling pressure (LVFP), a hallmark of HF agnostic to ejection fraction. Methods and results A derivation cohort of patients with suspected pulmonary hypertension and HF from the Sheffield centre underwent invasive right heart catheterization and CMR within 24 h of each other. A sex-specific CMR model to estimate LVFP, measured as pulmonary capillary wedge pressure (PCWP), was developed using multivariable regression. A validation cohort of patients with confirmed HF from the Leeds centre was used to evaluate for the primary endpoints of HF hospitalization and major adverse cardiovascular events (MACEs). Comparison between generic and sex-specific CMR-derived PCWP was undertaken. A total of 835 (60% female) and 454 (36% female) patients were recruited into the derivation and validation cohorts respectively. A sex-specific model incorporating left atrial volume and left ventricular mass was created. The generic CMR PCWP showed significant differences between males and females (14.7 ± 4 vs. 13 ± 3.0 mmHg, P > 0.001), not present with the sex-specific CMR PCWP (14.1 ± 3 vs. 13.8 mmHg, P = 0.3). The sex-specific, but not the generic, CMR PCWP was associated with HF hospitalization (hazard ratio 3.9, P = 0.0002) and MACE (hazard ratio 2.5, P = 0.001) over a mean follow-up period of 2.4 ± 1.2 years. Conclusion Accounting for sex improves precision and prognostic performance of CMR biomarkers for HF.
Collapse
Affiliation(s)
- Pankaj Garg
- Norwich Medical School, University of East Anglia, Norwich Research Park, Rosalind Franklin Road, Norwich NR4 7UQ, UK
- Department of Cardiology, Norfolk and Norwich University NHS Foundation Trust, Colney Lane, Norwich NR4 7UY, UK
| | - Ciaran Grafton-Clarke
- Norwich Medical School, University of East Anglia, Norwich Research Park, Rosalind Franklin Road, Norwich NR4 7UQ, UK
- Department of Cardiology, Norfolk and Norwich University NHS Foundation Trust, Colney Lane, Norwich NR4 7UY, UK
| | - Gareth Matthews
- Norwich Medical School, University of East Anglia, Norwich Research Park, Rosalind Franklin Road, Norwich NR4 7UQ, UK
- Department of Cardiology, Norfolk and Norwich University NHS Foundation Trust, Colney Lane, Norwich NR4 7UY, UK
| | - Peter Swoboda
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Liang Zhong
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore
- Signature Programme of Cardiovascular Metabolic and Disorders, Duke-NUS Medical School, 8 College Road, Singapore
| | - Nay Aung
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London, UK
| | - Ross Thomson
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London, UK
| | - Samer Alabed
- National Institute for Health and Care Research, Sheffield Biomedical Research Centre, Sheffield, UK
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Ahmet Demirkiran
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Department of Cardiology, Kocaeli City Hospital, Kocaeli, Turkey
| | - Vassilios S Vassiliou
- Norwich Medical School, University of East Anglia, Norwich Research Park, Rosalind Franklin Road, Norwich NR4 7UQ, UK
- Department of Cardiology, Norfolk and Norwich University NHS Foundation Trust, Colney Lane, Norwich NR4 7UY, UK
| | - Andrew J Swift
- National Institute for Health and Care Research, Sheffield Biomedical Research Centre, Sheffield, UK
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, Sheffield, UK
- INSIGNEO, Institute for in silico Medicine, University of Sheffield, Sheffield, UK
| |
Collapse
|
29
|
Chuah SH, Tan LK, Md Sari NA, Chan BT, Hasikin K, Lim E, Ung NM, Abdul Aziz YF, Jayabalan J, Liew YM. Remodeling in Aortic Stenosis With Reduced and Preserved Ejection Fraction: Insight on Motion Abnormality Via 3D + Time Personalized LV Modeling in Cardiac MRI. J Magn Reson Imaging 2024; 59:1242-1255. [PMID: 37452574 DOI: 10.1002/jmri.28915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND Increased afterload in aortic stenosis (AS) induces left ventricle (LV) remodeling to preserve a normal ejection fraction. This compensatory response can become maladaptive and manifest with motion abnormality. It is a clinical challenge to identify contractile and relaxation dysfunction during early subclinical stage to prevent irreversible deterioration. PURPOSE To evaluate the changes of regional wall dynamics in 3D + time domain as remodeling progresses in AS. STUDY TYPE Retrospective. POPULATION A total of 31 AS patients with reduced and preserved ejection fraction (14 AS_rEF: 7 male, 66.5 [7.8] years old; 17 AS_pEF: 12 male, 67.0 [6.0] years old) and 15 healthy (6 male, 61.0 [7.0] years old). FIELD STRENGTH/SEQUENCE 1.5 T Magnetic resonance imaging/steady state free precession and late-gadolinium enhancement sequences. ASSESSMENT Individual LV models were reconstructed in 3D + time domain and motion metrics including wall thickening (TI), dyssynchrony index (DI), contraction rate (CR), and relaxation rate (RR) were automatically extracted and associated with the presence of scarring and remodeling. STATISTICAL TESTS Shapiro-Wilk: data normality; Kruskal-Wallis: significant difference (P < 0.05); ICC and CV: variability; Mann-Whitney: effect size. RESULTS AS_rEF group shows distinct deterioration of cardiac motions compared to AS_pEF and healthy groups (TIAS_rEF : 0.92 [0.85] mm, TIAS_pEF : 5.13 [1.99] mm, TIhealthy : 3.61 [1.09] mm, ES: 0.48-0.83; DIAS_rEF : 17.11 [7.89]%, DIAS_pEF : 6.39 [4.04]%, DIhealthy : 5.71 [1.87]%, ES: 0.32-0.85; CRAS_rEF : 8.69 [6.11] mm/second, CRAS_pEF : 16.48 [6.70] mm/second, CRhealthy : 10.82 [4.57] mm/second, ES: 0.29-0.60; RRAS_rEF : 8.45 [4.84] mm/second; RRAS_pEF : 13.49 [8.56] mm/second, RRhealthy : 9.31 [2.48] mm/second, ES: 0.14-0.43). The difference in the motion metrics between healthy and AS_pEF groups were insignificant (P-value = 0.16-0.72). AS_rEF group was dominated by eccentric hypertrophy (47.1%) with concomitant scarring. Conversely, AS_pEF group was dominated by concentric remodeling and hypertrophy (71.4%), which could demonstrate hyperkinesia with slight wall dyssynchrony than healthy. Dysfunction of LV mechanics corresponded to the presence of myocardial scarring (54.9% in AS), which reverted the compensatory mechanisms initiated and performed by LV remodeling. DATA CONCLUSION The proposed 3D + time modeling technique may distinguish regional motion abnormalities between AS_pEF, AS_rEF, and healthy cohorts, aiding clinical diagnosis and monitoring of AS progression. Subclinical myocardial dysfunction is evident in early AS despite of normal EF. LEVEL OF EVIDENCE 4 TECHNICAL EFFICACY: Stage 1.
Collapse
Affiliation(s)
- Shoon Hui Chuah
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Li Kuo Tan
- Department of Biomedical Imaging, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- University Malaya Research Imaging Centre, Department of Biomedical Imaging, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Nor Ashikin Md Sari
- Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Bee Ting Chan
- Department of Mechanical, Materials and Manufacturing Engineering, Faculty of Science and Engineering, University of Nottingham Malaysia, Semenyih, Malaysia
| | - Khairunnisa Hasikin
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Einly Lim
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Ngie Min Ung
- Clinical Oncology Unit, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Yang Faridah Abdul Aziz
- University Malaya Research Imaging Centre, Department of Biomedical Imaging, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Jeyaraaj Jayabalan
- Department of Biomedical Imaging, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Yih Miin Liew
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
30
|
Wu Q, Liu WJ, Ma XY, Chang JS, Zhao XY, Liu YH, Yu XY. Zonisamide attenuates pressure overload-induced myocardial hypertrophy in mice through proteasome inhibition. Acta Pharmacol Sin 2024; 45:738-750. [PMID: 38097716 PMCID: PMC10943222 DOI: 10.1038/s41401-023-01191-7] [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: 07/09/2023] [Accepted: 11/02/2023] [Indexed: 03/17/2024]
Abstract
Myocardial hypertrophy is a pathological thickening of the myocardium which ultimately results in heart failure. We previously reported that zonisamide, an antiepileptic drug, attenuated pressure overload-caused myocardial hypertrophy and diabetic cardiomyopathy in murine models. In addition, we have found that the inhibition of proteasome activates glycogen synthesis kinase 3 (GSK-3) thus alleviates myocardial hypertrophy, which is an important anti-hypertrophic strategy. In this study, we investigated whether zonisamide prevented pressure overload-caused myocardial hypertrophy through suppressing proteasome. Pressure overload-caused myocardial hypertrophy was induced in mice by trans-aortic constriction (TAC) surgery. Two days after the surgery, the mice were administered zonisamide (10, 20, 40 mg·kg-1·d-1, i.g.) for four weeks. We showed that zonisamide administration significantly mitigated impaired cardiac function. Furthermore, zonisamide administration significantly inhibited proteasome activity as well as the expression levels of proteasome subunit beta types (PSMB) of the 20 S proteasome (PSMB1, PSMB2 and PSMB5) and proteasome-regulated particles (RPT) of the 19 S proteasome (RPT1, RPT4) in heart tissues of TAC mice. In primary neonatal rat cardiomyocytes (NRCMs), zonisamide (0.3 μM) prevented myocardial hypertrophy triggered by angiotensin II (Ang II), and significantly inhibited proteasome activity, proteasome subunits and proteasome-regulated particles. In Ang II-treated NRCMs, we found that 18α-glycyrrhetinic acid (18α-GA, 2 mg/ml), a proteasome inducer, eliminated the protective effects of zonisamide against myocardial hypertrophy and proteasome. Moreover, zonisamide treatment activated GSK-3 through inhibiting the phosphorylated AKT (protein kinase B, PKB) and phosphorylated liver kinase B1/AMP-activated protein kinase (LKB1/AMPKα), the upstream of GSK-3. Zonisamide treatment also inhibited GSK-3's downstream signaling proteins, including extracellular signal-regulated kinase (ERK) and GATA binding protein 4 (GATA4), both being the hypertrophic factors. Collectively, this study highlights the potential of zonisamide as a new therapeutic agent for myocardial hypertrophy, as it shows potent anti-hypertrophic potential through the suppression of proteasome.
Collapse
Affiliation(s)
- Qian Wu
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Wan-Jie Liu
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xin-Yu Ma
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ji-Shuo Chang
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xiao-Ya Zhao
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ying-Hua Liu
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Xi-Yong Yu
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
31
|
Frenzel S, Bülow R, Dörr M, Felix SB, Friedrich N, Völzke H, Wittfeld K, Grabe HJ, Bahls M. Left ventricular hypertrophy as a risk factor for accelerated brain aging: Results from the Study of Health in Pomerania. Hum Brain Mapp 2024; 45:e26567. [PMID: 38391110 PMCID: PMC10885183 DOI: 10.1002/hbm.26567] [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: 08/28/2023] [Revised: 11/16/2023] [Accepted: 11/30/2023] [Indexed: 02/24/2024] Open
Abstract
Previous studies provided evidence for the importance of cardiac structure abnormalities, in particular greater left ventricular (LV) mass, for brain aging, but longitudinal studies are lacking to date. We included 926 individuals (median age 48 years; 53% women) from the TREND cohort of the Study of Health in Pomerania (SHIP) without reduced ejection fraction or a history of myocardial infarction. LV mass index (LVMI) was determined by echocardiography at baseline. Brain morphometric measurements were derived from magnetic resonance images at baseline and 7-year follow-up. Direct effects of baseline LVMI on brain morphometry at follow-up were estimated using linear regression models with adjustment for baseline brain morphometry. At baseline, median LVMI was 40 g/m2.7 and 241 individuals (26%) met the criterion of LV hypertrophy. After correction for multiple testing, baseline LVMI was directly associated with reduced global cortical thickness and increased cortical brain age at follow-up independent from hypertension and blood pressure. Exposure-outcome relations were nonlinear and significantly stronger in the upper half of the exposure distribution. Specifically, an increase in baseline LVMI from the 50% quantile to the 95% quantile was associated additional 2.7 years (95% confidence interval = [1.5 years, 3.8 years]) of cortical brain age at follow-up. Additional regional analyses yielded bilateral effects on multiple frontal cortical regions. Our findings highlight the role of cardiac structure in brain aging. LVMI constitutes an easily measurable marker that might help to identify persons at risk for cognitive impairment and dementia.
Collapse
Affiliation(s)
- Stefan Frenzel
- Department of Psychiatry and PsychotherapyUniversity Medicine GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
| | - Robin Bülow
- Institute of Diagnostic Radiology and NeuroradiologyUniversity Medicine GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
| | - Marcus Dörr
- Department of Internal Medicine BUniversity Medicine GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
- German Centre for Cardiovascular Research (DZHK), Partner Site GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
| | - Stephan B. Felix
- Department of Internal Medicine BUniversity Medicine GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
- German Centre for Cardiovascular Research (DZHK), Partner Site GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
| | - Nele Friedrich
- German Centre for Cardiovascular Research (DZHK), Partner Site GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medicine GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
| | - Henry Völzke
- German Centre for Cardiovascular Research (DZHK), Partner Site GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
- Institute for Community MedicineUniversity Medicine GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
| | - Katharina Wittfeld
- Department of Psychiatry and PsychotherapyUniversity Medicine GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
- German Center for Neurodegenerative Disease (DZNE), Partner Site Rostock/GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
| | - Hans J. Grabe
- Department of Psychiatry and PsychotherapyUniversity Medicine GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
- German Center for Neurodegenerative Disease (DZNE), Partner Site Rostock/GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
| | - Martin Bahls
- Department of Internal Medicine BUniversity Medicine GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
- German Centre for Cardiovascular Research (DZHK), Partner Site GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
| |
Collapse
|
32
|
Bhullar SK, Dhalla NS. Adaptive and maladaptive roles of different angiotensin receptors in the development of cardiac hypertrophy and heart failure. Can J Physiol Pharmacol 2024; 102:86-104. [PMID: 37748204 DOI: 10.1139/cjpp-2023-0226] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Angiotensin II (Ang II) is formed by the action of angiotensin-converting enzyme (ACE) in the renin-angiotensin system. This hormone is known to induce cardiac hypertrophy and heart failure and its actions are mediated by the interaction of both pro- and antihypertrophic Ang II receptors (AT1R and AT2R). Ang II is also metabolized by ACE 2 to Ang-(1-7), which elicits the activation of Mas receptors (MasR) for inducing antihypertrophic actions. Since heart failure under different pathophysiological situations is preceded by adaptive and maladaptive cardiac hypertrophy, we have reviewed the existing literature to gain some information regarding the roles of AT1R, AT2R, and MasR in both acute and chronic conditions of cardiac hypertrophy. It appears that the activation of AT1R may be involved in the development of adaptive and maladaptive cardiac hypertrophy as well as subsequent heart failure because both ACE inhibitors and AT1R antagonists exert beneficial effects. On the other hand, the activation of both AT2R and MasR may prevent the occurrence of maladaptive cardiac hypertrophy and delay the progression of heart failure, and thus therapy with different activators of these antihypertrophic receptors under chronic pathological stages may prove beneficial. Accordingly, it is suggested that a great deal of effort should be made to develop appropriate activators of both AT2R and MasR for the treatment of heart failure subjects.
Collapse
Affiliation(s)
- Sukhwinder K Bhullar
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre and Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Naranjan S Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre and Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
33
|
Magruder JT, Holst KA, Stewart J, Yadav PK, Thourani VH. Early Intervention in Asymptomatic Aortic Stenosis: What Are We Waiting For? Can J Cardiol 2024; 40:201-209. [PMID: 38036025 DOI: 10.1016/j.cjca.2023.11.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/06/2023] [Accepted: 11/25/2023] [Indexed: 12/02/2023] Open
Abstract
Aortic stenosis (AS) contributes to significant cardiovascular morbidity and mortality worldwide, and the natural history from symptoms to ventricular decompensation, heart failure, and death has been well documented. For more than 2 decades, technologies including imaging and biomarkers have shown a promising ability to detect myocardial damage associated with AS before symptoms arise. Current treatment guidelines rely heavily on symptoms or ventricular decompensation as triggers for aortic valve intervention. There is increasing appreciation of the relationship between myocardial damage due to AS before the emergence of symptoms, and a number of published randomised trials suggest a benefit to early intervention in asymptomatic AS, with additional trials actively enrolling. Future treatment paradigms may incorporate early detection of ventricular damage by noninvasive new technologies as triggers for asymptomatic intervention. Enthusiasm for early aortic valve replacement should be tempered by consideration of the competing risks of early valve intervention, but an increasing preponderance of evidence continues to suggest that earlier intervention in AS is warranted.
Collapse
Affiliation(s)
- J Trent Magruder
- Department of Cardiovascular Surgery, Piedmont Heart Institute, Athens, Georgia, USA
| | - Kimberly A Holst
- Department of Cardiovascular Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Jim Stewart
- Department of Cardiology, Marcus Heart Valve Center, Piedmont Heart Institute, Atlanta, Georgia, USA
| | - Pradeep K Yadav
- Department of Cardiology, Marcus Heart Valve Center, Piedmont Heart Institute, Atlanta, Georgia, USA
| | - Vinod H Thourani
- Department of Cardiovascular Surgery, Marcus Heart Valve Center, Piedmont Heart Institute, Marcus Valve Center, Atlanta, Georgia, USA.
| |
Collapse
|
34
|
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
|
35
|
De la Garza Salazar F, Egenriether B. Exploring vectorcardiography: An extensive vectocardiogram analysis across age, sex, BMI, and cardiac conditions. J Electrocardiol 2024; 82:100-112. [PMID: 38113771 DOI: 10.1016/j.jelectrocard.2023.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/03/2023] [Accepted: 12/07/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND The vectocardiogram (VCG) offers a three-dimensional view of the heart's electrical activity, yet many VCG parameters remain unexplored in diverse clinical contexts. OBJECTIVES This study aims to explore the relationships between various VCG parameters and specific patient characteristics. METHODS ECG signals from adults were transformed into VCGs utilizing the Kors matrix, yielding 315 parameters per patient from the P, QRS and T loops. Univariable analysis, circular statistics, and stepwise logistic regression were employed to examine the relationships between VCG parameters and factors such as age, sex, BMI, hypertension, echocardiographic ischemic heart disease (Echo-IHD), and left ventricular hypertrophy (Echo-LVH). RESULTS We included 664 adults and considered an alpha value of 0.05 and a power of 90%. The study revealed significant associations, such as age with P loop roundness index (RI) (OR = 3.825, 95% confidence interval [95%CI] = 2.079-7.04), male sex with QRS loop RI (OR = 6.08, 95%CI = 1.835-20.153), abnormal BMI with the T loop's RI (OR = 0.544, 95%CI = 0.325-0.909), hypertension with the T loop planarity index (PI) (OR = 8.01, 95%CI = 2.134-30.117), Echo-IHD with QRS loop curvature at the 4/10th segment (OR = 7.58, 95%CI = 1.954-29.458), and Echo-LVH with the T loop lag-1/10 dihedral angle (OR = 10.3, 95%CI = 1.822-58.101). In the study, several additional VCG parameters demonstrated statistically significant, albeit smaller, associations with patient demographics and cardiovascular conditions. CONCLUSIONS The findings enhance our understanding of the intricate relationships between VCG parameters and patient characteristics, emphasizing the potential role of VCG analysis in assessing cardiovascular diseases. These insights may guide future research and clinical applications in cardiology.
Collapse
Affiliation(s)
| | - Brian Egenriether
- Monte Blanco #605 Col. Residencial San Agustín 2o Sector, 66260 San Pedro Garza García, Nuevo León, México
| |
Collapse
|
36
|
Laenens D, Stassen J, Galloo X, Ewe SH, Singh GK, Ammanullah MR, Hirasawa K, Sia CH, Butcher SC, Chew NWS, Kong WKF, Poh KK, Ding ZP, Ajmone Marsan N, Bax JJ. The impact of atrial fibrillation on prognosis in aortic stenosis. EUROPEAN HEART JOURNAL. QUALITY OF CARE & CLINICAL OUTCOMES 2023; 9:778-784. [PMID: 36669758 PMCID: PMC10745267 DOI: 10.1093/ehjqcco/qcad004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/11/2023] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
BACKGROUND Atrial fibrillation (AF) and aortic stenosis (AS) are both highly prevalent and often coexist. Various studies have focused on the prognostic value of AF in patients with AS, but rarely considered left ventricular (LV) diastolic function as a prognostic factor. OBJECTIVE To evaluate the prognostic impact of AF in patients with AS while correcting for LV diastolic function. METHODS Patients with first diagnosis of significant AS were selected and stratified according to history of AF. The endpoint was all-cause mortality. RESULTS In total, 2849 patients with significant AS (mean age 72 ± 12 years, 54.8% men) were evaluated, and 686 (24.1%) had a history of AF. During a median follow-up of 60 (30-97) months, 1182 (41.5%) patients died. Ten-year mortality rate in patients with AF was 46.8% compared to 36.8% in patients with sinus rhythm (SR) (log-rank P < 0.001). On univariable (HR: 1.42; 95% CI: 1.25-1.62; P < 0.001) and multivariable Cox regression analysis (HR: 1.19; 95% CI: 1.02-1.38; P = 0.026), AF was independently associated with mortality. However, when correcting for indexed left atrial volume, E/e' or both, AF was no longer independently associated with all-cause mortality. CONCLUSION Patients with significant AS and AF have a reduced survival as compared to patients with SR. Nonetheless, when correcting for markers of LV diastolic function, AF was not independently associated with outcomes in patients with significant AS.
Collapse
Affiliation(s)
- Dorien Laenens
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Jan Stassen
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Xavier Galloo
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - See Hooi Ewe
- Department of Cardiology, National Heart Center Singapore, 5 Hospital Drive, Singapore 169609, Singapore, Singapore
| | - Gurpreet K Singh
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Mohammed R Ammanullah
- Department of Cardiology, National Heart Center Singapore, 5 Hospital Drive, Singapore 169609, Singapore, Singapore
| | - Kensuke Hirasawa
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Ching-Hui Sia
- Department of Cardiology, National University Heart Center Singapore, 5 Lower Kent Ridge Rd, Singapore 119074, Singapore
| | - Steele C Butcher
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Department of Cardiology, Royal Perth Hospital, 197 Wellington St, Perth, WA 6000, Australia
| | - Nicholas W S Chew
- Department of Cardiology, National University Heart Center Singapore, 5 Lower Kent Ridge Rd, Singapore 119074, Singapore
| | - William K F Kong
- Department of Cardiology, National University Heart Center Singapore, 5 Lower Kent Ridge Rd, Singapore 119074, Singapore
| | - Kian Keong Poh
- Department of Cardiology, National University Heart Center Singapore, 5 Lower Kent Ridge Rd, Singapore 119074, Singapore
| | - Zee P Ding
- Department of Cardiology, National Heart Center Singapore, 5 Hospital Drive, Singapore 169609, Singapore, Singapore
| | - Nina Ajmone Marsan
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Jeroen J Bax
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Department of Cardiology, Turku Heart Center, University of Turku and Turku Unviersity Hospital, Kiinamyllynkatu 4-8, 20521 Turku, Finland
| |
Collapse
|
37
|
A.F. de Souza I, M.H. Padrao E, R. Marques I, A. Miyawaki I, Riceto Loyola Júnior JE, Caporal S. Moreira V, Gomes C, H.A. Silva C, Oprysko C, Barreto do Amaral Neto A, Cardoso R, Samesiana N, Alberto Pastore C, Tavares CA. Diagnostic Accuracy of ECG to Detect Left Ventricular Hypertrophy in Patients with Left Bundle Branch Block: A Systematic Review and Meta-analysis. CJC Open 2023; 5:971-980. [PMID: 38204852 PMCID: PMC10774079 DOI: 10.1016/j.cjco.2023.08.010] [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: 05/22/2023] [Accepted: 08/24/2023] [Indexed: 01/12/2024] Open
Abstract
Background Electrocardiographic (ECG) criteria to detect left ventricular hypertrophy (LVH) in patients with left bundle branch block (LBBB) remain under debate. We conducted a systematic review and meta-analysis to evaluate the diagnostic accuracy of different ECG criteria for diagnosing LVH in patients with LBBB. Methods We searched PubMed, Embase, Cochrane, and LILACS for articles evaluating the diagnostic accuracy of ECG criteria for LVH in patients with LBBB published between 1984 and 2023. Echocardiogram, magnetic resonance imaging, or autopsy were used as the reference standard for diagnosis of LVH. Risk of bias was assessed using the Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) tool. The co-primary outcomes were sensitivity, specificity, the diagnostic odds ratio, and likelihood ratios, estimated using a bivariate generalized linear mixed model for each ECG criterion. The prespecified protocol was registered in the International Prospective Register of Systematic Reviews (PROSPERO). Results We included 12 studies with a total of 1023 patients. We analyzed 10 criteria for LVH on ECG, including the Sokolow-Lyon criterion, the Cornell criterion, the RaVL (R wave in aVL) criterion, the Gubner-Ungerleider criterion, and the Dálfo criterion, among others. The Dalfó criterion was used for 487 patients and had the highest pooled sensitivity of 86% (95% confidence interval [CI] 57%-97%). All the other criteria had poor sensitivities. The Gubner-Ungerleider criterion and the RV5 or RV6 > 25 mm criterion had the highest specificities, with the former being used for 805 patients, obtaining a specificity of 99% (95% CI 80%-100%) and the latter being used for 355 patients, obtaining a specificity of 99% (95% CI 94%-100%). Conclusions In patients with LBBB, the use of ECG criteria had poor performance for ruling out LVH, mostly due to low sensitivities. None of the criteria analyzed demonstrated a balanced tradeoff between sensitivity and specificity, suggesting that ECG should not be used routinely to screen for LVH.
Collapse
Affiliation(s)
| | - Eduardo M.H. Padrao
- Department of Pulmonary and Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | - Cintia Gomes
- Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Caroliny H.A. Silva
- Universidade Federal do Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Carson Oprysko
- Department of Internal Medicine, University of Connecticut, Farmington, Connecticut, USA
| | | | - Rhanderson Cardoso
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nelson Samesiana
- Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, São Paulo, SP, Brazil
| | | | - Caio A.M. Tavares
- Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, São Paulo, SP, Brazil
- Academic Research Organization (ARO), Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| |
Collapse
|
38
|
Yan X, Gong J, Wang Z, Wang F, Qi C. Association of lipoprotein(a) with left ventricular hypertrophy assessed by electrocardiogram in adults: a large cross-sectional study. Front Endocrinol (Lausanne) 2023; 14:1260050. [PMID: 38098866 PMCID: PMC10720892 DOI: 10.3389/fendo.2023.1260050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/27/2023] [Indexed: 12/17/2023] Open
Abstract
Background and aims Increasing evidence supports a causal relationship between lipoprotein(a) [Lp(a)] and atherosclerotic cardiovascular disease, yet its association with left ventricular hypertrophy (LVH) assessed by electrocardiogram (ECG) remains unknown. The aim of this study was to explore the relationship between Lp(a) and LVH assessed by ECG in general population. Methods and results In this cross-sectional study, we screened 4,052 adults from the participants of the third National Health and Nutrition Examination Survey for analysis. Lp(a) was regarded as an exposure variable. LVH defined by the left ventricular mass index estimated from ECG was considered as an outcome variable. Multivariate logistic regression and restricted cubic spline (RCS) were used to assess the relationship between Lp(a) and LVH. Individuals with LVH had higher Lp(a) compared to individuals without LVH (P< 0.001). In the fully adjusted model, Lp(a) was strongly associated with LVH when as a continuous variable (per 1-unit increment, OR: 1.366, 95% CI: 1.043-1.789, P = 0.024), and higher Lp(a) remained independently associated with a higher risk of LVH when participants were divided into four groups according to quartiles of Lp(a) (Q4 vs Q1, OR: 1.508, 95% CI: 1.185-1.918, P = 0.001). And in subgroup analysis, this association remained significant among participants< 60 years, ≥ 60 years, male, with body mass index< 30 kg/m2, with hypertension and without diabetes (P< 0.05). In addition, we did not observe a nonlinear and threshold effect of Lp(a) with LVH in the RCS analysis (P for nonlinearity = 0.113). Conclusion Lp(a) was closely associated with LVH assessed by ECG in general population.
Collapse
Affiliation(s)
- Xuejiao Yan
- Department of Cardiology, The Affiliated Changzhou No.2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Jing Gong
- Department of Geriatrics, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhenwei Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fangfang Wang
- Department of Cardiology, The Affiliated Changzhou No.2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Chunjian Qi
- Medical Research Center, The Affiliated Changzhou No.2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| |
Collapse
|
39
|
Chang Y, Kang MK, Park MS, Leem GH, Song TJ. Resolved Proteinuria May Attenuate the Risk of Heart Failure: A Nationwide Population-Based Cohort Study. J Pers Med 2023; 13:1662. [PMID: 38138889 PMCID: PMC10744716 DOI: 10.3390/jpm13121662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/14/2023] [Accepted: 11/26/2023] [Indexed: 12/24/2023] Open
Abstract
Although proteinuria is a risk factor for heart failure (HF), proteinuria can be reversible or persistent. Our objective was to explore the link between changes in the proteinuria status and the risk of HF. We included participants from a Korean national health screening cohort who underwent health examinations in 2003-2004 and 2005-2006 and had no history of HF. Participants were categorized into four groups: proteinuria-free, proteinuria-resolved, proteinuria-developed, and proteinuria-persistent. The outcome of interest was the occurrence of HF. The study included 1,703,651 participants, among whom 17,543 (1.03%) were in the proteinuria-resolved group and 4585 (0.27%) were in the proteinuria-persistent group. After a median follow-up period of 14.04 years (interquartile range 14.19-15.07), HF occurred in 75,064 (4.41%) participants. A multivariable Cox proportional hazards regression analysis indicated that the proteinuria-persistent group had a higher risk of HF compared with the proteinuria-free group (hazard ratio (HR): 2.19, 95% confidence interval (CI): 2.03-2.36, p < 0.001). In a further pairwise comparison analysis, participants in the proteinuria-resolved group had a relatively low risk of HF compared with those in the proteinuria-persistent group (HR: 0.64, 95% CI: 0.58-0.70, p < 0.001). In conclusion, the risk of HF can change with alterations in the proteinuria status.
Collapse
Affiliation(s)
- Yoonkyung Chang
- Department of Neurology, Mokdong Hospital, Ewha Womans University College of Medicine, Seoul 08209, Republic of Korea;
| | - Min Kyoung Kang
- Department of Neurology, Seoul Hospital, Ewha Womans University College of Medicine, Seoul 07804, Republic of Korea; (M.K.K.); (M.-S.P.)
| | - Moo-Seok Park
- Department of Neurology, Seoul Hospital, Ewha Womans University College of Medicine, Seoul 07804, Republic of Korea; (M.K.K.); (M.-S.P.)
| | - Gwang-Hyun Leem
- Ewha Medical Research Institute, Ewha Womans University, Seoul 07804, Republic of Korea;
| | - Tae-Jin Song
- Department of Neurology, Seoul Hospital, Ewha Womans University College of Medicine, Seoul 07804, Republic of Korea; (M.K.K.); (M.-S.P.)
| |
Collapse
|
40
|
Yang Y, Hao Z, An N, Han Y, Miao W, Storey KB, Lefai E, Liu X, Wang J, Liu S, Xie M, Chang H. Integrated transcriptomics and metabolomics reveal protective effects on heart of hibernating Daurian ground squirrels. J Cell Physiol 2023; 238:2724-2748. [PMID: 37733616 DOI: 10.1002/jcp.31123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/02/2023] [Accepted: 09/08/2023] [Indexed: 09/23/2023]
Abstract
Hibernating mammals are natural models of resistance to ischemia, hypoxia-reperfusion injury, and hypothermia. Daurian ground squirrels (spermophilus dauricus) can adapt to endure multiple torpor-arousal cycles without sustaining cardiac damage. However, the molecular regulatory mechanisms that underlie this adaptive response are not yet fully understood. This study investigates morphological, functional, genetic, and metabolic changes that occur in the heart of ground squirrels in three groups: summer active (SA), late torpor (LT), and interbout arousal (IBA). Morphological and functional changes in the heart were measured using hematoxylin-eosin (HE) staining, Masson staining, echocardiography, and enzyme-linked immunosorbent assay (ELISA). Results showed significant changes in cardiac function in the LT group as compared with SA or IBA groups, but no irreversible damage occurred. To understand the molecular mechanisms underlying these phenotypic changes, transcriptomic and metabolomic analyses were conducted to assess differential changes in gene expression and metabolite levels in the three groups of ground squirrels, with a focus on GO and KEGG pathway analysis. Transcriptomic analysis showed that differentially expressed genes were involved in the remodeling of cytoskeletal proteins, reduction in protein synthesis, and downregulation of the ubiquitin-proteasome pathway during hibernation (including LT and IBA groups), as compared with the SA group. Metabolomic analysis revealed increased free amino acids, activation of the glutathione antioxidant system, altered cardiac fatty acid metabolic preferences, and enhanced pentose phosphate pathway activity during hibernation as compared with the SA group. Combining the transcriptomic and metabolomic data, active mitochondrial oxidative phosphorylation and creatine-phosphocreatine energy shuttle systems were observed, as well as inhibition of ferroptosis signaling pathways during hibernation as compared with the SA group. In conclusion, these results provide new insights into cardio-protection in hibernators from the perspective of gene and metabolite changes and deepen our understanding of adaptive cardio-protection mechanisms in mammalian hibernators.
Collapse
Affiliation(s)
- Yingyu Yang
- Shaanxi Key Laboratory for Animal Conservation, College of Life Sciences, Northwest University, Xi'an, China
| | - Ziwei Hao
- Shaanxi Key Laboratory for Animal Conservation, College of Life Sciences, Northwest University, Xi'an, China
| | - Ning An
- Shaanxi Key Laboratory for Animal Conservation, College of Life Sciences, Northwest University, Xi'an, China
| | - Yuting Han
- Shaanxi Key Laboratory for Animal Conservation, College of Life Sciences, Northwest University, Xi'an, China
| | - Weilan Miao
- Shaanxi Key Laboratory for Animal Conservation, College of Life Sciences, Northwest University, Xi'an, China
| | - Kenneth B Storey
- Department of Biology, Carleton University, Ottawa, Ontario, Canada
| | - Etienne Lefai
- INRAE, Unité de Nutrition Humaine, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Xiaoxuan Liu
- Shaanxi Key Laboratory for Animal Conservation, College of Life Sciences, Northwest University, Xi'an, China
| | - Junshu Wang
- Shaanxi Key Laboratory for Animal Conservation, College of Life Sciences, Northwest University, Xi'an, China
| | - Shuo Liu
- Shaanxi Key Laboratory for Animal Conservation, College of Life Sciences, Northwest University, Xi'an, China
| | - Manjiang Xie
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, Shaanxi, China
| | - Hui Chang
- Shaanxi Key Laboratory for Animal Conservation, College of Life Sciences, Northwest University, Xi'an, China
| |
Collapse
|
41
|
Dweck MR, Loganath K, Bing R, Treibel TA, McCann GP, Newby DE, Leipsic J, Fraccaro C, Paolisso P, Cosyns B, Habib G, Cavalcante J, Donal E, Lancellotti P, Clavel MA, Otto CM, Pibarot P. Multi-modality imaging in aortic stenosis: an EACVI clinical consensus document. Eur Heart J Cardiovasc Imaging 2023; 24:1430-1443. [PMID: 37395329 DOI: 10.1093/ehjci/jead153] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 06/16/2023] [Indexed: 07/04/2023] Open
Abstract
In this EACVI clinical scientific update, we will explore the current use of multi-modality imaging in the diagnosis, risk stratification, and follow-up of patients with aortic stenosis, with a particular focus on recent developments and future directions. Echocardiography is and will likely remain the key method of diagnosis and surveillance of aortic stenosis providing detailed assessments of valve haemodynamics and the cardiac remodelling response. Computed tomography (CT) is already widely used in the planning of transcutaneous aortic valve implantation. We anticipate its increased use as an anatomical adjudicator to clarify disease severity in patients with discordant echocardiographic measurements. CT calcium scoring is currently used for this purpose; however, contrast CT techniques are emerging that allow identification of both calcific and fibrotic valve thickening. Additionally, improved assessments of myocardial decompensation with echocardiography, cardiac magnetic resonance, and CT will become more commonplace in our routine assessment of aortic stenosis. Underpinning all of this will be widespread application of artificial intelligence. In combination, we believe this new era of multi-modality imaging in aortic stenosis will improve the diagnosis, follow-up, and timing of intervention in aortic stenosis as well as potentially accelerate the development of the novel pharmacological treatments required for this disease.
Collapse
Affiliation(s)
- Marc R Dweck
- Centre for Cardiovascular Science, University of Edinburgh, Chancellors Building, Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Krithika Loganath
- Centre for Cardiovascular Science, University of Edinburgh, Chancellors Building, Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Rong Bing
- Centre for Cardiovascular Science, University of Edinburgh, Chancellors Building, Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Thomas A Treibel
- Barts Heart Centre, Bart's Health NHS Trust, W Smithfield, EC1A 7BE, London, UK
- University College London Institute of Cardiovascular Science, 62 Huntley St, WC1E 6DD, London, UK
| | - Gerry P McCann
- Department of Cardiovascular Sciences, University of Leicester, University Rd, Leicester LE1 7RH, UK
- The NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester, LE3 9QP, UK
| | - David E Newby
- Centre for Cardiovascular Science, University of Edinburgh, Chancellors Building, Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Jonathon Leipsic
- Centre for Cardiovascular Innovation, St Paul's and Vancouver General Hospital, 1081 Burrard St Room 166, Vancouver, British Columbia V6Z 1Y6, Canada
| | - Chiara Fraccaro
- Department of Cardiac, Thoracic and Vascular Science and Public Health, Via Giustiniani, 2 - 35128, Padua, Italy
| | - Pasquale Paolisso
- Cardiovascular Center Aalst, OLV Clinic, Moorselbaan 164, 9300 Aalst, Belgium
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80125 Naples, Italy
| | - Bernard Cosyns
- Department of Cardiology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Jette, Belgium
| | - Gilbert Habib
- Cardiology Department, Hôpital La Timone, 264 Rue Saint-Pierre, 13005 Marseille, France
| | - João Cavalcante
- Allina Health Minneapolis Heart Institute, Abbott Northwestern Hospital, 800 E 28th St, Minneapolis, MN 55407, USA
| | - Erwan Donal
- Cardiology and CIC, Université Rennes, 2 Rue Henri Le Guilloux, 35033 Rennes, France
| | - Patrizio Lancellotti
- GIGA Cardiovascular Sciences, Department of Cardiology, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
- Gruppo Villa Maria Care and Research, Corso Giuseppe Garibaldi, 11, 48022 Lugo RA, Italy
| | - Marie-Annick Clavel
- Institut Universitaire de Cardiologie et de Pneumologie de Québec/Québec Heart and Lung Institute, 2725 Ch Ste-Foy, Québec, QC G1V 4G5, Canada
- Faculté de Médecine-Département de Médecine, Université Laval, Ferdinand Vandry Pavillon, 1050 Av. de la Médecine, Québec City, Quebec G1V 0A6, Canada
| | - Catherine M Otto
- Division of Cardiology, Department of Medicine, University of Washington School of Medicine, 4333 Brooklyn Ave NE Box 359458, Seattle, WA 98195-9458, USA
| | - Phillipe Pibarot
- Institut Universitaire de Cardiologie et de Pneumologie de Québec/Québec Heart and Lung Institute, 2725 Ch Ste-Foy, Québec, QC G1V 4G5, Canada
| |
Collapse
|
42
|
Zoico E, Giani A, Saatchi T, Rizzatti V, Mazzali G, Fantin F, Benfari G, Onorati F, Urbani S, Zamboni M. Myocardial Fibrosis and Steatosis in Patients with Aortic Stenosis: Roles of Myostatin and Ceramides. Int J Mol Sci 2023; 24:15508. [PMID: 37958492 PMCID: PMC10648018 DOI: 10.3390/ijms242115508] [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: 09/07/2023] [Revised: 10/16/2023] [Accepted: 10/20/2023] [Indexed: 11/15/2023] Open
Abstract
Aortic stenosis (AS) involves progressive valve obstruction and a remodeling response of the left ventriculum (LV) with systolic and diastolic dysfunction. The roles of interstitial fibrosis and myocardial steatosis in LV dysfunction in AS have not been completely characterized. We enrolled 31 patients (19 women and 12 men) with severe AS undergoing elective aortic valve replacement. The subjects were clinically evaluated, and transthoracic echocardiography was performed pre-surgery. LV septal biopsies were obtained to assess fibrosis and apoptosis and fat deposition in myocytes (perilipin 5 (PLIN5)), or in the form of adipocytes within the heart (perilipin 1 (PLIN1)), the presence of ceramides and myostatin were assessed via immunohistochemistry. After BMI adjustment, we found a positive association between fibrosis and apoptotic cardiomyocytes, as well as fibrosis and the area covered by PLIN5. Apoptosis and PLIN5 were also significantly interrelated. LV fibrosis increased with a higher medium gradient (MG) and peak gradient (PG). Ceramides and myostatin levels were higher in patients within the higher MG and PG tertiles. In the linear regression analysis, increased fibrosis correlated with increased apoptosis and myostatin, independent from confounding factors. After adjustment for age and BMI, we found a positive relationship between PLIN5 and E/A and a negative correlation between septal S', global longitudinal strain (GLS), and fibrosis. Myostatin was inversely correlated with GLS and ejection fraction. Fibrosis and myocardial steatosis altogether contribute to ventricular dysfunction in severe AS. The association of myostatin and fibrosis with systolic dysfunction, as well as between myocardial steatosis and diastolic dysfunction, highlights potential therapeutic targets.
Collapse
Affiliation(s)
- Elena Zoico
- Division of Geriatric Medicine, Department of Medicine, University of Verona, 37126 Verona, Italy; (A.G.)
| | - Anna Giani
- Division of Geriatric Medicine, Department of Medicine, University of Verona, 37126 Verona, Italy; (A.G.)
| | - Tanaz Saatchi
- Division of Geriatric Medicine, Department of Medicine, University of Verona, 37126 Verona, Italy; (A.G.)
| | - Vanni Rizzatti
- Division of Geriatric Medicine, Department of Medicine, University of Verona, 37126 Verona, Italy; (A.G.)
| | - Gloria Mazzali
- Division of Geriatric Medicine, Department of Medicine, University of Verona, 37126 Verona, Italy; (A.G.)
| | - Francesco Fantin
- Division of Geriatric Medicine, Department of Medicine, University of Verona, 37126 Verona, Italy; (A.G.)
| | - Giovanni Benfari
- Division of Cardiology, Department of Medicine, University of Verona, 37126 Verona, Italy
| | - Francesco Onorati
- Division of Cardiac Surgery, Department of Surgery, Dentistry, Pediatric and Gynecology, University of Verona, 37126 Verona, Italy
| | - Silvia Urbani
- Division of Geriatric Medicine, Department of Medicine, University of Verona, 37126 Verona, Italy; (A.G.)
| | - Mauro Zamboni
- Division of Geriatric Medicine, Department of Surgery, Dentistry, Pediatric and Gynecology, University of Verona, 37126 Verona, Italy
| |
Collapse
|
43
|
Somsura R, Kamkajon K, Chaimongkolnukul K, Chantip S, Teerapornpuntakit J, Wongdee K, Kamonsutthipaijit N, Tangtrongsup S, Panupinthu N, Tiyasatkulkovit W, Charoenphandhu N. Tissue-specific expression of senescence biomarkers in spontaneously hypertensive rats: evidence of premature aging in hypertension. PeerJ 2023; 11:e16300. [PMID: 37872946 PMCID: PMC10590574 DOI: 10.7717/peerj.16300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/25/2023] [Indexed: 10/25/2023] Open
Abstract
Background Cellular senescence is an age-related physiological process that contributes to tissue dysfunction and accelerated onset of chronic metabolic diseases including hypertension. Indeed, elevation of blood pressure in hypertension coincides with premature vascular aging and dysfunction. In addition, onsets of metabolic disturbance and osteopenia in patients with hypertension have also been reported. It is possible that hypertension enhances premature aging and causes progressive loss of function in multiple organs. However, the landscape of cellular senescence in critical tissues affected by hypertension remains elusive. Materials and Methods Heart, liver, bone, hypothalamus, and kidney were collected from spontaneously hypertensive rats (SHR) and age- and sex-matched normotensive Wistar rats (WT) at 6, 12, 24 and 36 weeks of age (n = 10 animals/group). Changes in mRNA levels of senescence biomarkers namely cyclin-dependent kinase (CDK) inhibitors (CDKIs), i.e., Cdkn2a (encoding p16Ink4a) and Cdkn1a (encoding p21cip1) as well as senescence-associated secretory phenotypes (SASPs), i.e., Timp1, Mmp12, Il6 and Cxcl1, were determined. Additionally, bone collagen alignment and hydroxy apatite crystal dimensions were determined by synchrotron radiation small- and wide-angle X-ray scattering (SAXS/WAXS) techniques. Results Real-time PCR revealed that transcript levels of genes encoding CDKIs and SASPs in the heart and liver were upregulated in SHR from 6 to 36 weeks of age. Expression of Timp1 and Cxcl1 was increased in bone tissues isolated from 36-week-old SHR. In contrast, we found that expression levels of Timp1 and Il6 mRNA were decreased in hypothalamus and kidney of SHR in all age groups. Simultaneous SAXS/WAXS analysis also revealed misalignment of bone collagen fibers in SHR as compared to WT. Conclusion Premature aging was identified in an organ directly affected by high blood pressure (i.e., heart) and those with known functional defects in SHR (i.e., liver and bone). Cellular senescence was not evident in organs with autoregulation of blood pressure (i.e., brain and kidney). Our study suggested that cellular senescence is induced by persistently elevated blood pressure and in part, leading to organ dysfunction. Therefore, interventions that can both lower blood pressure and prevent cellular senescence should provide therapeutic benefits for treatment of cardiovascular and metabolic consequences.
Collapse
Affiliation(s)
- Ratthapon Somsura
- Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Master of Science Program in Zoology, Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Kanokwan Kamkajon
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | - Surachai Chantip
- National Laboratory Animal Center, Mahidol University, Nakhon Pathom, Thailand
| | - Jarinthorn Teerapornpuntakit
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Kannikar Wongdee
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
- Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand
| | | | - Suwimol Tangtrongsup
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Nattapon Panupinthu
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | - Narattaphol Charoenphandhu
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
- The Academy of Science, The Royal Society of Thailand, Dusit, Bangkok, Thailand
| |
Collapse
|
44
|
Koga M, Izumo M, Yoneyama K, Akashi YJ, Yashima F, Tada N, Yamawaki M, Shirai S, Naganuma T, Yamanaka F, Ueno H, Tabata M, Mizutani K, Takagi K, Watanabe Y, Yamamoto M, Hayashida K. Prognostic Value of Electrocardiographic Left Ventricular Hypertrophy After Transcatheter Aortic Valve Implantation: Insights from the OCEAN-TAVI Registry. Am J Cardiol 2023; 204:130-139. [PMID: 37541149 DOI: 10.1016/j.amjcard.2023.07.101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 08/06/2023]
Abstract
Electrocardiogram (ECG) left ventricular hypertrophy (LVH) is associated with the prognosis of patients with aortic stenosis. However, the impact of the presence or absence of ECG-LVH on the clinical outcomes after transcatheter aortic valve implantation (TAVI) is limited. This study aimed to assess the prognostic value of ECG-LVH among patients with aortic stenosis treated by TAVI. A total of 1,667 patients who underwent TAVI were prospectively enrolled into the OCEAN-TAVI (Optimized CathEter vAlvular iNtervention-Transcatheter Aortic Valve Implantation) registry. A total of 1,446 patients (mean age 84 years; 29.9% men) were analyzed. The Sokolow-Lyon index was used to determine the presence of ECG-LVH. LVH was also assessed using transthoracic echocardiography (TTE). We investigated the association between ECG-LVH and all-cause and cardiovascular mortality. This study identified ECG-LVH and TTE-LVH in 743 (51.5%) and 1,242 patients (86.0%), respectively. The Kaplan-Meier analysis revealed that all-cause mortality was significantly higher among patients without ECG-LVH than among those with ECG-LVH (log-rank p <0.001). In the multivariable analysis, the absence of ECG-LVH was independently associated with all-cause mortality (hazard ratio 1.98, 95% confidence interval 1.39 to 2.82, p <0.001), regardless of the presence or absence of TTE-LVH. Furthermore, the presence of TTE-LVH with the absence of ECG-LVH was observed in 575 patients (40%), which was associated with cardiovascular mortality (hazard ratio 2.84, 95% confidence interval 1.56 to 5.17, p <0.001). In conclusion, the absence of ECG-LVH was independently associated with an increased risk of all-cause mortality after TAVI. Risk stratification using both ECG-LVH and TTE-LVH is a useful predictor of adverse clinical outcomes after TAVI.
Collapse
Affiliation(s)
- Masashi Koga
- Division of Cardiology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Masaki Izumo
- Division of Cardiology, St. Marianna University School of Medicine, Kawasaki, Japan.
| | - Kihei Yoneyama
- Division of Cardiology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yoshihiro J Akashi
- Division of Cardiology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Fumiaki Yashima
- Department of Cardiology, Saiseikai Utsunomiya Hospital, Utsunomiya, Japan
| | - Norio Tada
- Department of Cardiology, Sendai Kosei Hospital, Sendai, Japan
| | - Masahiro Yamawaki
- Department of Cardiology, Saiseikai Yokohama City Eastern Hospital, Yokohama, Japan
| | - Shinichi Shirai
- Department of Cardiovascular Medicine, Kokura Memorial Hospital, Kitakyushu, Japan
| | - Toru Naganuma
- Department of Cardiology, New Tokyo Hospital, Matsudo, Japan
| | - Futoshi Yamanaka
- Department of Cardiology, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Hiroshi Ueno
- Department of Cardiovascular Medicine, Toyama University Hospital, Toyama, Japan
| | - Minoru Tabata
- Department of Cardiovascular Surgery, Tokyo Bay Urayasu Ichikawa Medical Center, Urayasu, Japan
| | - Kazuki Mizutani
- Department of Cardiology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Kensuke Takagi
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yusuke Watanabe
- Department of Cardiology, Teikyo University School of Medicine, Tokyo, Japan
| | - Masanori Yamamoto
- Department of Cardiology, Toyohashi Heart Center, Toyohashi, Japan; Department of Cardiology, Nagoya Heart Center, Nagoya, Japan
| | - Kentaro Hayashida
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
45
|
Hjertaas JJ, Einarsen E, Gerdts E, Kokorina M, Moen CA, Urheim S, Saeed S, Matre K. Impact of aortic valve stenosis on myocardial deformation in different left ventricular levels: A three-dimensional speckle tracking echocardiography study. Echocardiography 2023; 40:1028-1039. [PMID: 37543718 DOI: 10.1111/echo.15668] [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: 12/29/2022] [Revised: 07/05/2023] [Accepted: 07/27/2023] [Indexed: 08/07/2023] Open
Abstract
BACKGROUND Global systolic left ventricular (LV) myocardial function progressively declines as degenerative aortic valve stenosis (AS) progresses. Whether this results in uniformly distributed deformation changes from base to apex has not been investigated. METHODS Eighty-five AS patients underwent three-dimensional (3D) echocardiography in this cross-sectional study. Patients were grouped by peak jet velocity into mild (n = 32), moderate (n = 31), and severe (n = 22) AS. 3D speckle tracking derived strain, rotation, twist, and torsion were obtained to assess global LV function and myocardial function at the apical, mid, and basal levels. RESULTS Global longitudinal strain (GLS) was lower in patients with severe AS (-16.1 ± 2.4% in mild, -15.5 ± 2.5% in moderate, and -13.5 ± 3.0% in severe AS [all p < .01]). Peak basal and mid longitudinal strain (LS), basal rotation and twist from apical to basal level followed the same pattern, while peak apical LS was higher in moderate AS compared to severe AS (all p < .05). In multivariate analyses, lower GLS was particularly associated with male sex, higher body mass index and peak aortic jet velocity, lower basal LS with higher filling pressure (E/e') and LV mass, lower mid LS with higher RWT and presence of AS symptoms, and lower apical LS with male sex and higher systolic blood pressure, respectively (all p < .05). CONCLUSION Using 3D speckle tracking echocardiography reveals regional and global changes in LV mechanics in AS related to the severity of AS, LV remodeling and presence of cardiovascular risk factors.
Collapse
Affiliation(s)
| | - Eigir Einarsen
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Eva Gerdts
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Marina Kokorina
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | | | - Stig Urheim
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Sahrai Saeed
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Knut Matre
- Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
46
|
Conley JM, Lambright CS, Evans N, Farraj AK, Smoot J, Grindstaff RD, Hill D, McCord J, Medlock-Kakaley E, Dixon A, Hines E, Gray LE. Dose additive maternal and offspring effects of oral maternal exposure to a mixture of three PFAS (HFPO-DA, NBP2, PFOS) during pregnancy in the Sprague-Dawley rat. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 892:164609. [PMID: 37271399 PMCID: PMC10681034 DOI: 10.1016/j.scitotenv.2023.164609] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/30/2023] [Accepted: 05/30/2023] [Indexed: 06/06/2023]
Abstract
Simultaneous exposure to multiple per- and polyfluoroalkyl substances (PFAS) is common in humans across the globe. Individual PFAS are associated with adverse health effects, yet the nature of mixture effects after exposure to two or more PFAS remains unclear. Previously we reported that oral administration of hexafluoropropylene oxide-dimer acid (HFPO-DA, or GenX), Nafion byproduct 2 (NBP2), or perfluorooctane sulfonate (PFOS) individually during pregnancy produced maternal and F1 effects. Here, we hypothesized that responses to the combined exposure to these three PFAS would be dose additive. Pregnant Sprague-Dawley rats were exposed to a fixed-ratio equipotent mixture where the top dose contained each PFAS at their ED50 for neonatal mortality (100 % dose = PFOS 3 mg/kg; NBP2 10 mg/kg; HFPO-DA 110 mg/kg), followed by a dilution series (33.3, 10, 3.3, and 1 %) and vehicle controls (0 % dose). Consistent with the single chemical studies, dams were exposed from gestation day (GD)14-18 or from GD8-postnatal day (PND2). Fetal and maternal livers on GD18 displayed multiple significantly upregulated genes associated with lipid and carbohydrate metabolism at all dose levels, while dams displayed significantly increased liver weight (≥3.3 % dose) and reduced serum thyroid hormones (≥33.3 % dose). Maternal exposure from GD8-PND2 significantly reduced pup bodyweights at birth (≥33.3 % dose) and PND2 (all doses), increased neonatal liver weights (≥3.3 % dose), increased pup mortality (≥3.3 % dose), and reduced maternal bodyweights and weight gain at the top dose. Echocardiography of adult F1 males and females identified significantly increased left ventricular anterior wall thickness (~10 % increase), whereas other cardiac morphological, functional, and transcriptomic measures were unaffected. Mixture effects in maternal and neonatal animals conformed to dose addition using a relative potency factor (RPF) analysis. Results support dose addition-based cumulative assessment approaches for estimating combined effects of PFAS co-exposure.
Collapse
Affiliation(s)
- Justin M Conley
- U.S. Environmental Protection Agency, Office of Research & Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Christy S Lambright
- U.S. Environmental Protection Agency, Office of Research & Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Nicola Evans
- U.S. Environmental Protection Agency, Office of Research & Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Aimen K Farraj
- U.S. Environmental Protection Agency, Office of Research & Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Jacob Smoot
- ORISE Participant, U.S. Environmental Protection Agency, Office of Research & Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Rachel D Grindstaff
- U.S. Environmental Protection Agency, Office of Research & Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA
| | - Donna Hill
- U.S. Environmental Protection Agency, Office of Research & Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - James McCord
- U.S. Environmental Protection Agency, Office of Research & Development, Center for Environmental Measurement and Modeling, Research Triangle Park, NC, USA.
| | - Elizabeth Medlock-Kakaley
- U.S. Environmental Protection Agency, Office of Research & Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Aaron Dixon
- U.S. Environmental Protection Agency, Office of Research & Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Erin Hines
- U.S. Environmental Protection Agency, Office of Research & Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - L Earl Gray
- U.S. Environmental Protection Agency, Office of Research & Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| |
Collapse
|
47
|
Kanwischer L, Xu X, Saifuddin AB, Maamari S, Tan X, Alnour F, Tampe B, Meyer T, Zeisberg M, Hasenfuss G, Puls M, Zeisberg EM. Low levels of circulating methylated IRX3 are related to worse outcome after transcatheter aortic valve implantation in patients with severe aortic stenosis. Clin Epigenetics 2023; 15:149. [PMID: 37697352 PMCID: PMC10496273 DOI: 10.1186/s13148-023-01561-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 09/04/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND Aortic stenosis (AS) is one of the most common cardiac diseases and major cause of morbidity and mortality in the elderly. Transcatheter aortic valve implantation (TAVI) is performed in such patients with symptomatic severe AS and reduces mortality for the majority of these patients. However, a significant percentage dies within the first two years after TAVI, such that there is an interest to identify parameters, which predict outcome and could guide pre-TAVI patient selection. High levels of cardiac fibrosis have been identified as such independent predictor of cardiovascular mortality after TAVI. Promoter hypermethylation commonly leads to gene downregulation, and the Iroquois homeobox 3 (IRX3) gene was identified in a genome-wide transcriptome and methylome to be hypermethylated and downregulated in AS patients. In a well-described cohort of 100 TAVI patients in which cardiac fibrosis levels were quantified histologically in cardiac biopsies, and which had a follow-up of up to two years, we investigated if circulating methylated DNA of IRX3 in the peripheral blood is associated with cardiac fibrosis and/or mortality in AS patients undergoing TAVI and thus could serve as a biomarker to add information on outcome after TAVI. RESULTS Patients with high levels of methylation in circulating IRX3 show a significantly increased survival as compared to patients with low levels of IRX3 methylation indicating that high peripheral IRX3 methylation is associated with an improved outcome. In the multivariable setting, peripheral IRX3 methylation acts as an independent predictor of all-cause mortality. While there is no significant correlation of levels of IRX3 methylation with cardiac death, there is a significant but very weak inverse correlation between circulating IRX3 promoter methylation level and the amount of cardiac fibrosis. Higher levels of peripheral IRX3 methylation further correlated with decreased cardiac IRX3 expression and vice versa. CONCLUSIONS High levels of IRX3 methylation in the blood of AS patients at the time of TAVI are associated with better overall survival after TAVI and at least partially reflect myocardial IRX3 expression. Circulating methylated IRX3 might aid as a potential biomarker to help guide both pre-TAVI patient selection and post-TAVI monitoring.
Collapse
Affiliation(s)
- Leon Kanwischer
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August-University, Robert-Koch-Str. 40, 37075, Göttingen, Germany
- DZHK German Center for Cardiovascular Research, Partner Site Göttingen, Göttingen, Germany
| | - Xingbo Xu
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August-University, Robert-Koch-Str. 40, 37075, Göttingen, Germany
- DZHK German Center for Cardiovascular Research, Partner Site Göttingen, Göttingen, Germany
| | - Afifa Binta Saifuddin
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August-University, Robert-Koch-Str. 40, 37075, Göttingen, Germany
- DZHK German Center for Cardiovascular Research, Partner Site Göttingen, Göttingen, Germany
| | - Sabine Maamari
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August-University, Robert-Koch-Str. 40, 37075, Göttingen, Germany
- DZHK German Center for Cardiovascular Research, Partner Site Göttingen, Göttingen, Germany
| | - Xiaoying Tan
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
- DZHK German Center for Cardiovascular Research, Partner Site Göttingen, Göttingen, Germany
| | - Fouzi Alnour
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August-University, Robert-Koch-Str. 40, 37075, Göttingen, Germany
- DZHK German Center for Cardiovascular Research, Partner Site Göttingen, Göttingen, Germany
| | - Björn Tampe
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Thomas Meyer
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
- DZHK German Center for Cardiovascular Research, Partner Site Göttingen, Göttingen, Germany
| | - Michael Zeisberg
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
- DZHK German Center for Cardiovascular Research, Partner Site Göttingen, Göttingen, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August-University, Robert-Koch-Str. 40, 37075, Göttingen, Germany
- DZHK German Center for Cardiovascular Research, Partner Site Göttingen, Göttingen, Germany
| | - Miriam Puls
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August-University, Robert-Koch-Str. 40, 37075, Göttingen, Germany
- DZHK German Center for Cardiovascular Research, Partner Site Göttingen, Göttingen, Germany
| | - Elisabeth M Zeisberg
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August-University, Robert-Koch-Str. 40, 37075, Göttingen, Germany.
- DZHK German Center for Cardiovascular Research, Partner Site Göttingen, Göttingen, Germany.
| |
Collapse
|
48
|
Nikorowitsch J, Bei der Kellen R, Haack A, Magnussen C, Prochaska J, Wild PS, Dörr M, Twerenbold R, Schnabel RB, Kirchhof P, Blankenberg S, Markus MRP, Wenzel JP. Correlation of systolic and diastolic blood pressure with echocardiographic phenotypes of cardiac structure and function from three German population-based studies. Sci Rep 2023; 13:14525. [PMID: 37666935 PMCID: PMC10477248 DOI: 10.1038/s41598-023-41571-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: 05/02/2023] [Accepted: 08/29/2023] [Indexed: 09/06/2023] Open
Abstract
Arterial hypertension is considered a risk factor for the development of heart failure. Here we investigate cross-sectional associations of systolic and diastolic blood pressure with subtle functional and morphological changes of left ventricular echocardiographic parameters representing early dysfunction in three representative German population-based studies. We assessed 26,719 individuals without symptomatic heart failure from the Hamburg City Health Study (HCHS, n = 7396, derivation cohort), the Gutenberg Health Study (GHS, 14,715, validation cohort) and the Study of Health in Pomerania (SHIP, 4608, validation cohort). Multivariable linear regression analyses with systolic and diastolic blood pressure as continuous exposure variables were adjusted for common cardiovascular risk factors and antihypertensive medication. Both systolic and diastolic blood pressure were consistently associated with measures of left ventricular hypertrophy (β per standard deviation (SD) for LV mass (g) and systolic blood pressure: 5.09 (p < 0.001); diastolic blood pressure: 2.29 (p < 0.001) in HCHS). Systolic blood pressure correlated with declining diastolic function (β per SD for E/e': 0.29, p < 0.001 in HCHS) and diastolic blood pressure with declining systolic function (β per SD for LVEF, in %: - 0.15; p = 0.041 in HCHS) in all cohorts. Pending further validation, our results from three independent German population samples suggest differential effects of systolic versus diastolic blood pressure on left ventricular structure and function.
Collapse
Affiliation(s)
- Julius Nikorowitsch
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany.
| | | | - Alena Haack
- Epidemiological Study Center, Hamburg, Germany
| | - Christina Magnussen
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany
| | - Jürgen Prochaska
- Preventive Cardiology and Preventive Medicine, Department of Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine Main, Mainz, Germany
| | - Philipp S Wild
- Preventive Cardiology and Preventive Medicine, Department of Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine Main, Mainz, Germany
| | - Marcus Dörr
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Raphael Twerenbold
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany
- Epidemiological Study Center, Hamburg, Germany
- Center for Population Health Innovation (POINT Institute), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Renate B Schnabel
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany
- Epidemiological Study Center, Hamburg, Germany
| | - Paulus Kirchhof
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Stefan Blankenberg
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany
- Epidemiological Study Center, Hamburg, Germany
- Center for Population Health Innovation (POINT Institute), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Cardio-CARE, Medizincampus Davos, Davos, Switzerland
| | - Marcello Ricardo Paulista Markus
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
- German Center for Diabetes Research (DZD), Partner Site Greifswald, Greifswald, Germany
| | - Jan-Per Wenzel
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany
- Epidemiological Study Center, Hamburg, Germany
| |
Collapse
|
49
|
Agbaje AO, Zachariah JP, Tuomainen TP. Arterial stiffness but not carotid intima-media thickness progression precedes premature structural and functional cardiac damage in youth: A 7-year temporal and mediation longitudinal study. Atherosclerosis 2023; 380:117197. [PMID: 37582328 DOI: 10.1016/j.atherosclerosis.2023.117197] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/26/2023] [Accepted: 07/26/2023] [Indexed: 08/17/2023]
Abstract
BACKGROUND AND AIMS The longitudinal relations of cardiac indices with the aorta and carotid vessel and the time sequence for early cardiac disease development are uncharacterized in youth. We examined the temporal longitudinal associations of carotid-femoral pulse wave velocity (cfPWV) and carotid intima-media thickness (cIMT) with left ventricular hypertrophy (LVH) and diastolic dysfunction (LVDD). METHODS From the Avon Longitudinal Study of Parents and Children, UK birth cohort, 1856 adolescents (1011 females) at a mean (SD) age 17.7 (0.3) years were followed up for 7 years. Vicorder-measured cfPWV and ultrasound-measured cIMT were grouped in tertiles as low (reference), moderate, and high. Echocardiography measured cardiac abnormalities are left ventricular mass indexed for height2.7 (LVMI2.7) ≥51 g/m2.7 as LVH; relative wall thickness ≥44 as hiRWT; LVD function E/A <1.5 as LVD dysfunction (LVDD); and LV filling pressure E/e' ≥8 as hiLVFP. Data were analysed with generalized logit mixed-effect models, cross-lagged path, and mediation structural equation models adjusting for cardiometabolic and lifestyle factors. RESULTS Over follow-up, LVH prevalence increased from 3.6% to 7.2% and LVDD from 11.1 to 16.3%. High cfPWV progression was associated with worsening LVH [Odds ratio 1.23 (1.13-1.35); p < 0.001] in the total cohort, males, overweight/obese, and normotensive. High cfPWV progression was associated with worsening hiLVFP in the total cohort, females, and normal weight. Likewise, high cIMT progression was associated with worsening LVH [1.27 (1.26-1.27); p < 0.0001] in the total cohort, overweight/obese and elevated BP/hypertensive. Neither cfPWV nor cIMT progression was associated with worsening hiRWT in the total cohort. In cross-lagged models, higher baseline cfPWV was associated with future LVMI2.7 (β = 0.06, SE, 5.14, p = 0.035), RWT, LVDF, and LVFP. However, baseline LVMI2.7, RWT, LVDF, and LVFP were not associated with follow-up cfPWV. Baseline cIMT was not associated with follow-up cardiac indices and vice versa. Cumulative increased systolic blood pressure (34.3% mediation) and insulin resistance (15.1% mediation) mediated the direct associations of cumulative cfPWV with cumulative LVMI2.7. CONCLUSIONS Arterial stiffness progression temporally preceded worsening structural and functional cardiac damage in youth with increased systolic blood pressure and insulin resistance partly mediating the relationships. Future interventions aimed at attenuating premature cardiac damage in adolescents and young adults may consider a simultaneous treatment of both arterial stiffness, elevated blood pressure and insulin resistance.
Collapse
Affiliation(s)
- Andrew O Agbaje
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland; Children's Health and Exercise Research Centre, Department of Public Health and Sports Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK.
| | - Justin P Zachariah
- Section of Pediatric Cardiology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, United States
| | - Tomi-Pekka Tuomainen
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
50
|
Vasconcelos MM, Ganan CS, da Silveira CFDSMP, Malagutte KNDS, Poiati JR, Nunes HRDC, Martin LC, Bazan R, Borges VTM, Bazan SGZ. Evolution of Myocardial Hypertrophy Associated With Pregnancy in Hypertensive Women Six Months Postpartum. Curr Probl Cardiol 2023; 48:101772. [PMID: 37121455 DOI: 10.1016/j.cpcardiol.2023.101772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/02/2023]
Abstract
Systemic arterial hypertension (SAH) is one of the principal risk factors for developing cardiovascular disease. When a hypertensive woman becomes pregnant, new hemodynamic condition is installed, with addition from chronic pressure overload to chronic volume overload. This new hemodynamic condition can provide greater myocardial hypertrophy(LVH), whose postpartum evolution has been little studied in the literature. To evaluate LVH in hypertensive women in the third trimester of pregnancy and 6 months postpartum and to establish which clinical variables are associated with elevated risk of LVH. Prospective longitudinal study including 41 pregnant women beyond 35 gestational weeks and with previous SAH. They were submitted to clinical and echocardiographic evaluation at the gestational period and 6 months postpartum. Statistical analysis: multivariate logistic regression with the exposures most strongly associated with maintenance of hypertrophy in univariate analysis. Significance level: P<0.05. The mean age was 29±6.2 years. The majority of the women were white(85.4%). Before pregnancy 23(59%) women used antihypertensive drugs and 28(71.8%) used during pregnancy. At the end of gestation, all women presented LVH, 79% maintained hypertrophy 6 months postpartum. In multivariate analysis, exposures significantly associated with hypertrophy maintenance: systolic blood pressure(SBP) at the end of gestation, OR=1.16(1.03-1.30);P=0.013 and SBP increase at 6 months postpartum in relation to end of gestation, OR=22.9(1.8-294);P=0.016. In hypertensive pregnant women, LVH frequency is elevated at the end of pregnancy, and recovery frequency of this hypertrophy, at 6 months postpartum, is very low. The increase of SBP 6 months postpartum was associated with maintenance of hypertrophy.
Collapse
Affiliation(s)
- Milena Miranda Vasconcelos
- Department of Internal Medicine, Botucatu Medical School - UNESP, São Paulo State University, Botucatu, Brazil
| | - Camilla Sousa Ganan
- Department of Internal Medicine, Botucatu Medical School - UNESP, São Paulo State University, Botucatu, Brazil
| | | | | | - Juliane Rosa Poiati
- Department of Gynecology and Obstetrics, Botucatu Medical School - UNESP, São Paulo State University, Botucatu, Brazil
| | | | - Luis Cuadrado Martin
- Department of Internal Medicine, Botucatu Medical School - UNESP, São Paulo State University, Botucatu, Brazil
| | - Rodrigo Bazan
- Department of Internal Medicine, Botucatu Medical School - UNESP, São Paulo State University, Botucatu, Brazil
| | | | - Silméia Garcia Zanati Bazan
- Department of Internal Medicine, Botucatu Medical School - UNESP, São Paulo State University, Botucatu, Brazil
| |
Collapse
|