1
|
Peña B, Bosi S, Knight WE, Cavasin M, Ferrari I, Musani SA, Cobb TM, Kumar M, Montelongo E, Abdel-Hafiz M, Zanetti M, Farahzad N, Alegret N, McKinsey TA, Graw SL, Sbaizero O, Chi C, Vagnozzi RJ, Song K, Taylor MRG, Prato M, Park D, Mestroni L. Biocompatibility Assessment of an Injectable Carbon Nanotube-Functionalized Reverse Thermal Gel for Cardiac Tissue Engineering Applications. ACS APPLIED BIO MATERIALS 2025. [PMID: 40343469 DOI: 10.1021/acsabm.5c00125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Heart failure (HF) is a major contributor to the global burden of cardiovascular disease. Current treatments for HF do not regenerate or restore cardiac muscle function, leaving cardiac transplantation as the only definitive treatment for end-stage HF. Subsequently, there is a tremendous need for alternative HF treatments as well as methods to effectively and selectively deliver those therapies to the heart. We have engineered an injectable reverse thermal gel (RTG) functionalized with carbon nanotubes (CNTs) to create a thermoresponsive conductive hydrogel or RTG-CNT. The RTG-CNT transitions from a liquid solution to a gel-based matrix upon reaching body temperature, a unique quality that allows for rapid injection of the liquid polymeric solution followed by gel localization in situ. Previously, we demonstrated the potential use of the RTG-CNT hydrogel for cardiac tissue engineering applications using three-dimensional (3D) cocultures of primary cardiac cells. Here, we performed a preclinical study to assess the biocompatibility of our RTG-CNT hydrogel in vivo by using hydrogel intracardial injection in a mouse model and in vitro by using 3D cultures of human-induced pluripotent stem cell-derived cardiomyocytes. In this report, we present compelling results that demonstrate the RTG-CNT hydrogel biocompatibility and its potential for use in cardiac tissue engineering applications.
Collapse
Affiliation(s)
- Brisa Peña
- Bioengineering Department, University of Colorado Denver Anschutz Medical Campus, at Bioscience 2 1270 E. Montview Avenue, Suite 100, Aurora, Colorado 80045, United States
- Cardiovascular Institute, School of Medicine, Division of Cardiology, University of Colorado Denver Anschutz Medical Campus, at 12700 E.19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, at 12700 E.19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
| | - Susanna Bosi
- Department of Chemical and Pharmaceutical Sciences, INSTM Unit of Trieste, University of Trieste, Via L. Giorgieri 1, 34127 Trieste, Italy
| | - Walter E Knight
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, at 12700 E.19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
| | - Maria Cavasin
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, at 12700 E.19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
| | - Ilaria Ferrari
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, at 12700 E.19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
| | - Sara A Musani
- Bioengineering Department, University of Colorado Denver Anschutz Medical Campus, at Bioscience 2 1270 E. Montview Avenue, Suite 100, Aurora, Colorado 80045, United States
| | - Tristan M Cobb
- Bioengineering Department, University of Colorado Denver Anschutz Medical Campus, at Bioscience 2 1270 E. Montview Avenue, Suite 100, Aurora, Colorado 80045, United States
| | - Maydha Kumar
- Bioengineering Department, University of Colorado Denver Anschutz Medical Campus, at Bioscience 2 1270 E. Montview Avenue, Suite 100, Aurora, Colorado 80045, United States
| | - Efren Montelongo
- Bioengineering Department, University of Colorado Denver Anschutz Medical Campus, at Bioscience 2 1270 E. Montview Avenue, Suite 100, Aurora, Colorado 80045, United States
| | - Mostafa Abdel-Hafiz
- Bioengineering Department, University of Colorado Denver Anschutz Medical Campus, at Bioscience 2 1270 E. Montview Avenue, Suite 100, Aurora, Colorado 80045, United States
| | - Michele Zanetti
- Department of Biomedical Engineering, Yale University, 55 Prospect St., New Haven, Connecticut 06511, United States
| | - Nasim Farahzad
- Bioengineering Department, University of Colorado Denver Anschutz Medical Campus, at Bioscience 2 1270 E. Montview Avenue, Suite 100, Aurora, Colorado 80045, United States
| | - Nuria Alegret
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, 20014 Donostia-San Sebastián, Spain
| | - Timothy A McKinsey
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, at 12700 E.19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, at 12700 E.19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
| | - Sharon L Graw
- Cardiovascular Institute, School of Medicine, Division of Cardiology, University of Colorado Denver Anschutz Medical Campus, at 12700 E.19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, at 12700 E.19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
| | - Orfeo Sbaizero
- Department of Engineering and Architecture, University of Trieste, at Via Valerio 2, Trieste 34127, Italy
| | - Congwu Chi
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, at 12700 E.19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
| | - Ronald J Vagnozzi
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, at 12700 E.19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, at 12700 E.19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
| | - Kunhua Song
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, at 12700 E.19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
| | - Matthew R G Taylor
- Cardiovascular Institute, School of Medicine, Division of Cardiology, University of Colorado Denver Anschutz Medical Campus, at 12700 E.19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, at 12700 E.19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
| | - Maurizio Prato
- Department of Chemical and Pharmaceutical Sciences, INSTM Unit of Trieste, University of Trieste, Via L. Giorgieri 1, 34127 Trieste, Italy
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, 20014 Donostia-San Sebastián, Spain
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Daewon Park
- Bioengineering Department, University of Colorado Denver Anschutz Medical Campus, at Bioscience 2 1270 E. Montview Avenue, Suite 100, Aurora, Colorado 80045, United States
| | - Luisa Mestroni
- Cardiovascular Institute, School of Medicine, Division of Cardiology, University of Colorado Denver Anschutz Medical Campus, at 12700 E.19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, at 12700 E.19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
| |
Collapse
|
2
|
Wang J, Verkerk AO, Wilders R, Zhang Y, Zhang K, Prakosa A, Rivaud MR, Marsman EMJ, Boender AR, Klerk M, Fokkert L, de Jonge B, Neef K, Kirzner OF, Bezzina CR, Remme CA, Tan HL, Boukens BJ, Devalla HD, Trayanova NA, Christoffels VM, Barnett P, Boink GJJ. SCN10A-short gene therapy to restore conduction and protect against malignant cardiac arrhythmias. Eur Heart J 2025; 46:1747-1762. [PMID: 39973098 PMCID: PMC12055233 DOI: 10.1093/eurheartj/ehaf053] [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: 10/23/2023] [Revised: 05/27/2024] [Accepted: 01/23/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND AND AIMS Life-threatening arrhythmias are a well-established consequence of reduced cardiac sodium current (INa). Gene therapy approaches to increase INa have demonstrated potential benefits to prevent arrhythmias. However, the development of such therapies is hampered by the large size of sodium channels. In this study, SCN10A-short (S10s), a short transcript encoding the carboxy-terminal domain of the human neuronal sodium channel, was evaluated as a gene therapy target to increase INa and prevent arrhythmias. METHODS Adeno-associated viral vector overexpressing S10s was injected into wild type and Scn5a-haploinsufficient mice on which patch-clamp studies, optical mapping, electrocardiogram analyses, and ischaemia reperfusion were performed. In vitro and in silico studies were conducted to further explore the effect of S10s gene therapy in the context of human hearts. RESULTS Cardiac S10s overexpression increased cellular INa, maximal action potential upstroke velocity, and action potential amplitude in Scn5a-haploinsufficient cardiomyocytes. S10s gene therapy rescues conduction slowing in Scn5a-haploinsufficient mice and prevented ventricular tachycardia induced by ischaemia-reperfusion in wild type mice. S10s overexpression increased maximal action potential upstroke velocity in human inducible pluripotent stem cell-derived cardiomyocytes and prevented inducible arrhythmias in simulated human heart models. CONCLUSIONS S10s gene therapy may be effective to treat cardiac conduction abnormalities and associated arrhythmias.
Collapse
Affiliation(s)
- Jianan Wang
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, Amsterdam 1105 AZ, The Netherlands
| | - Arie O Verkerk
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, Amsterdam 1105 AZ, The Netherlands
- Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, Amsterdam 1105 AZ, The Netherlands
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, Amsterdam 1105 AZ, The Netherlands
| | - Yingnan Zhang
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kelly Zhang
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Adityo Prakosa
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Mathilde R Rivaud
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, Amsterdam 1105 AZ, The Netherlands
| | - E Madelief J Marsman
- Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, Amsterdam 1105 AZ, The Netherlands
| | - Arie R Boender
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, Amsterdam 1105 AZ, The Netherlands
- PacingCure B.V., Roetersstraat 35, Amsterdam 1018 WB, The Netherlands
| | - Mischa Klerk
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, Amsterdam 1105 AZ, The Netherlands
| | - Lianne Fokkert
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, Amsterdam 1105 AZ, The Netherlands
| | - Berend de Jonge
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, Amsterdam 1105 AZ, The Netherlands
| | - Klaus Neef
- PacingCure B.V., Roetersstraat 35, Amsterdam 1018 WB, The Netherlands
- Netherlands Heart Institute, Moreelsepark 1, Utrecht 3511 EP, The Netherlands
| | - Osne F Kirzner
- PacingCure B.V., Roetersstraat 35, Amsterdam 1018 WB, The Netherlands
- Department of Anaesthesiology, Amsterdam University Medical Centers, De Boelelaan 1117, Amsterdam 1081 HV, The Netherlands
| | - Connie R Bezzina
- Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, Amsterdam 1105 AZ, The Netherlands
| | - Carol Ann Remme
- Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, Amsterdam 1105 AZ, The Netherlands
| | - Hanno L Tan
- Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, Amsterdam 1105 AZ, The Netherlands
- PacingCure B.V., Roetersstraat 35, Amsterdam 1018 WB, The Netherlands
- Netherlands Heart Institute, Moreelsepark 1, Utrecht 3511 EP, The Netherlands
| | - Bastiaan J Boukens
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, Amsterdam 1105 AZ, The Netherlands
- Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, Maastricht 6229 ER, The Netherlands
| | - Harsha D Devalla
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, Amsterdam 1105 AZ, The Netherlands
| | - Natalia A Trayanova
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Alliance for Cardiovascular Diagnostic and Treatment Innovation, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, Amsterdam 1105 AZ, The Netherlands
| | - Phil Barnett
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, Amsterdam 1105 AZ, The Netherlands
| | - Gerard J J Boink
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, Amsterdam 1105 AZ, The Netherlands
- PacingCure B.V., Roetersstraat 35, Amsterdam 1018 WB, The Netherlands
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
| |
Collapse
|
3
|
Hafez OA, Chang RB. Regulation of Cardiac Function by the Autonomic Nervous System. Physiology (Bethesda) 2025; 40:0. [PMID: 39585760 DOI: 10.1152/physiol.00018.2024] [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: 04/05/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/27/2024] Open
Abstract
The autonomic nervous system is critical for regulating cardiovascular physiology. The neurocardiac axis encompasses multiple levels of control, including the motor circuits of the sympathetic and parasympathetic nervous systems, sensory neurons that contribute to cardiac reflexes, and the intrinsic cardiac nervous system that provides localized sensing and regulation of the heart. Disruption of these systems can lead to significant clinical conditions. Recent advances have enhanced our understanding of the autonomic control of the heart, detailing the specific neuronal populations involved and their physiologic roles. In this review, we discuss this research at each level of the neurocardiac axis. We conclude by discussing the clinical field of neurocardiology and attempts to translate this new understanding of neurocardiac physiology to the clinic. We highlight the contributions of autonomic dysfunction in prevalent cardiovascular diseases and assess the current status of novel neuroscience-based treatment approaches.
Collapse
Affiliation(s)
- Omar A Hafez
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, United States
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, United States
- M.D.-Ph.D. Program, Yale University School of Medicine, New Haven, Connecticut, United States
| | - Rui B Chang
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, United States
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, United States
| |
Collapse
|
4
|
Pramanda AN, Farabi F, Prameswari HS, Achmad C, Tiksnadi BB. Myocardial-alternation index (MMI) is correlated with soluble suppression of tumorigenecity-2 (sST2) in patients with ischemic cardiomyopathy. Egypt Heart J 2025; 77:39. [PMID: 40261549 PMCID: PMC12014887 DOI: 10.1186/s43044-025-00634-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 03/23/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Ischemic cardiomyopathy is a condition that represents myocardial dysfunction due to obstructive coronary artery disease. In ischemic cardiomyopathy, both structural and electrical remodeling occur. Myocardial biomarker, soluble ST2 (sST2) is able to predict patient's mortality and morbidity, and structural remodeling of the heart is responsible for its expression. ECG dispersion mapping (ECG-DM) as evaluated by myocardial micro-alternation index (MMI) may predict alteration of the myocardial electrophysiology with high sensitivity and specificity. The association between structural and electrical remodeling in ischemic cardiomyopathy is not fully understood. This study aims to evaluate the correlation between MMI and sST2 level in patients with ischemic cardiomyopathy. RESULT Total patients who met for the inclusion criteria were 30 patients. Mean age was 57.97 ± 10.04 years; most patients were male (80%). 27 (90%) patients had class II NYHA functional class. The most common risk factors were smoking (20 (66,7%)) and hypertension (17 (56,7%)). Median MMI was 34.0% (IQR: 23.0-42.3%) and median sST2 was 5.6 ng/mL (IQR: 2.0-11.5 ng/mL). This study found that MMI had a significant correlation with sST2, indicating a link between structural and electrical remodeling in ischemic cardiomyopathy (r = 0.583, p < 0,05). CONCLUSION There was a correlation between MMI and sST2 in patients with ischemic cardiomyopathy.
Collapse
Affiliation(s)
- Andra Naufal Pramanda
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Bandung, Indonesia
| | - Fatih Farabi
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Bandung, Indonesia
| | - Hawani Sasmaya Prameswari
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Bandung, Indonesia
| | - Chaerul Achmad
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Bandung, Indonesia
| | - Badai Bhatara Tiksnadi
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Bandung, Indonesia.
| |
Collapse
|
5
|
Agrawal N, Afzal M, Almalki WH, Ballal S, Sharma GC, Krithiga T, Panigrahi R, Saini S, Ali H, Goyal K, Rana M, Abida Khan. Longevity mechanisms in cardiac aging: exploring calcium dysregulation and senescence. Biogerontology 2025; 26:94. [PMID: 40259024 DOI: 10.1007/s10522-025-10229-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 03/20/2025] [Indexed: 04/23/2025]
Abstract
Cardiac aging is a multistep process that results in a loss of various structural and functional heart abilities, increasing the risk of heart disease. Since its remarkable discovery in the early 1800s, when limestone is heated, calcium's importance has been defined in numerous ways. It can help stiffen shells and bones, function as a reducing agent in chemical reactions, and play a central role in cellular signalling. The movement of calcium ions in and out of cells and between those is referred to as calcium signalling. It influences the binding of the ligand, enzyme activity, electrochemical gradients, and other cellular processes. Calcium signalling is critical for both contraction and relaxation under the sliding filament model of heart muscle. However, with age, the heart undergoes changes that lead to increases in cardiac dysfunction, such as myocardial fibrosis, decreased cardiomyocyte function, and noxious disturbances in calcium homeostasis. Additionally, when cardiac tissues age, cellular senescence, a state of irreversible cell cycle arrest, accumulates and begins to exacerbate tissue inflammation and fibrosis. This review explores the most recent discoveries regarding the role of senescent cell accumulation and calcium signalling perturbances in cardiac aging. Additionally, new treatment strategies are used to reduce aged-related heart dysfunction by targeting senescent cells and modulating calcium homeostasis.
Collapse
Affiliation(s)
- Neetu Agrawal
- Institute of Pharmaceutical Research, GLA University, Mathura, UP, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Girish Chandra Sharma
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - T Krithiga
- Department of Chemistry, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Rajashree Panigrahi
- Department of Microbiology IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Suman Saini
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to Be University), Clement Town, Dehradun, 248002, India
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, Uttarakhand, 248007, India
| | - Abida Khan
- Center For Health Research, Northern Border University, Arar 73213, Saudi Arabia
| |
Collapse
|
6
|
Tonon CR, Pereira AG, Ferreira NF, Monte MG, Vieira NM, Fujimori ASS, Ballin PDS, de Paiva SAR, Zornoff LAM, Minicucci MF, Polegato BF. The Gut-Heart Axis and Its Role in Doxorubicin-Induced Cardiotoxicity: A Narrative Review. Microorganisms 2025; 13:855. [PMID: 40284691 PMCID: PMC12029146 DOI: 10.3390/microorganisms13040855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/04/2025] [Accepted: 04/06/2025] [Indexed: 04/29/2025] Open
Abstract
Doxorubicin is a widely used chemotherapy for the treatment of several types of cancer. However, its application is restricted due to adverse effects, particularly cardiotoxicity, which can progress to heart failure-a chronic and debilitating condition. Several mechanisms have been identified in the pathophysiology of doxorubicin-induced cardiotoxicity, including oxidative stress, mitochondrial dysfunction, inflammation, and disruption of collagen homeostasis. More recently, dysbiosis of the gut microbiota has been implicated in the development and perpetuation of cardiac injury. Studies have reported alterations in the composition and abundance of the microbiota during doxorubicin treatment. Therefore, as of recent, there is a new field of research in order to develop strategies involving the gut microbiota to prevent or attenuate cardiotoxicity since there is no effective therapy at the moment. This narrative review aims to provide an update on the role of gut microbiota and intestinal permeability in the pathophysiology of cardiovascular diseases, and more specifically doxorubicin-induced cardiotoxicity. Additionally, it seeks to establish a foundation for future research targeting gut microbiota to alleviate cardiotoxicity.
Collapse
Affiliation(s)
- Carolina Rodrigues Tonon
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, Brazil (B.F.P.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Boichenko V, Noakes VM, Reilly-O’Donnell B, Luciani GB, Emanueli C, Martelli F, Gorelik J. Circulating Non-Coding RNAs as Indicators of Fibrosis and Heart Failure Severity. Cells 2025; 14:553. [PMID: 40214506 PMCID: PMC11989213 DOI: 10.3390/cells14070553] [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: 03/22/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
Heart failure (HF) is a leading cause of morbidity and mortality worldwide, representing a complex clinical syndrome in which the heart's ability to pump blood efficiently is impaired. HF can be subclassified into heart failure with reduced ejection fraction (HFrEF) and heart failure with preserved ejection fraction (HFpEF), each with distinct pathophysiological mechanisms and varying levels of severity. The progression of HF is significantly driven by cardiac fibrosis, a pathological process in which the extracellular matrix undergoes abnormal and uncontrolled remodelling. Cardiac fibrosis is characterized by excessive matrix protein deposition and the activation of myofibroblasts, increasing the stiffness of the heart, thus disrupting its normal structure and function and promoting lethal arrythmia. MicroRNAs, long non-coding RNAs, and circular RNAs, collectively known as non-coding RNAs (ncRNAs), have recently gained significant attention due to a growing body of evidence suggesting their involvement in cardiac remodelling such as fibrosis. ncRNAs can be found in the peripheral blood, indicating their potential as biomarkers for assessing HF severity. In this review, we critically examine recent advancements and findings related to the use of ncRNAs as biomarkers of HF and discuss their implication in fibrosis development.
Collapse
Affiliation(s)
- Veronika Boichenko
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Cardiovascular and Surgical Sciences, The University of Verona, Policlinico G. B. Rossi, P.le. La Scuro 10, 37134 Verona, Italy
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Via Morandi 30, San Donato Milanese, 20097 Milano, Italy
| | - Victoria Maria Noakes
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Benedict Reilly-O’Donnell
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Giovanni Battista Luciani
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Cardiovascular and Surgical Sciences, The University of Verona, Policlinico G. B. Rossi, P.le. La Scuro 10, 37134 Verona, Italy
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Via Morandi 30, San Donato Milanese, 20097 Milano, Italy
| | - Julia Gorelik
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
8
|
Kozhevnikova MV, Belenkov YN, Shestakova KM, Ageev AA, Markin PA, Kakotkina AV, Korobkova EO, Moskaleva NE, Kuznetsov IV, Khabarova NV, Kukharenko AV, Appolonova SA. Metabolomic profiling in heart failure as a new tool for diagnosis and phenotyping. Sci Rep 2025; 15:11849. [PMID: 40195403 PMCID: PMC11976976 DOI: 10.1038/s41598-025-95553-2] [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: 11/27/2024] [Accepted: 03/21/2025] [Indexed: 04/09/2025] Open
Abstract
Classifying heart failure (HF) by stages and ejection fraction (EF) remains a debated topic in cardiology. Metabolomic profiling (MP) offers a means to identify unique pathophysiological changes across different phenotypes, presenting a promising approach for the diagnosis and prognosis of HF, as well as for the development of targeted therapies. In our study, MP was performed on 408 HF patients (54.9% male). The mean ages of patients were 62 [53;68], 67 [65;74], 68 [61;72], and 69 [65;73] years for stages A, B, C, and D, respectively. This study demonstrates high accuracy in HF stage classification, distinguishing Stage A from Stage B with an AUC ROC of 0.91 and Stage B from Stage C with an AUC ROC of 0.97, by integrating chromatography-mass spectrometry data through multiparametric machine learning models. The observed metabolic similarities between HF with mildly reduced EF and HF with reduced EF phenotypes (AUC ROC 0.96) once again highlight the fundamental differences at the cellular and molecular levels between HF with preserved EF and HF with EF < 50%. Hierarchical clustering based on MP identified four distinct HF phenotypes and 26 key metabolites, including metabolites of tryptophan catabolism, glutamine, riboflavin, norepinephrine, serine, and long- and medium-chain acylcarnitines. The average follow-up period was 542.37 [16;1271] days. A downward change in the trajectory of EF [HR 3,008, 95% CI 1,035 to 8,743, p = 0,043] and metabolomic cluster 3 [HR 2,880; 95% CI 1,062 to 7,810, p = 0,0376] were associated with increased risk of all-cause mortality. MP can refine HF phenotyping and deepen the understanding of its underlying mechanisms. Metabolomic analysis illuminates the biochemical landscape of HF, aiding in its classification and suggesting new therapeutic pathways.
Collapse
Affiliation(s)
- Maria V Kozhevnikova
- Hospital Therapy No. 1 Department, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, 119435, Russia.
- I.M. Sechenov First Moscow State Medical University, 2-4 Bolshaya Pirogovskaya St., 119991, Moscow, Russia.
| | - Yuri N Belenkov
- Hospital Therapy No. 1 Department, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, 119435, Russia
| | - Ksenia M Shestakova
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, 119435, Russia
| | - Anton A Ageev
- Hospital Therapy No. 1 Department, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, 119435, Russia
| | - Pavel A Markin
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, 119435, Russia
| | - Anastasiia V Kakotkina
- Hospital Therapy No. 1 Department, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, 119435, Russia
| | - Ekaterina O Korobkova
- Hospital Therapy No. 1 Department, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, 119435, Russia
| | - Natalia E Moskaleva
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, 119435, Russia
| | - Ivan V Kuznetsov
- Hospital Therapy No. 1 Department, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, 119435, Russia
| | - Natalia V Khabarova
- Hospital Therapy No. 1 Department, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, 119435, Russia
| | - Alexey V Kukharenko
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, 119435, Russia
| | - Svetlana A Appolonova
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, 119435, Russia
| |
Collapse
|
9
|
Fahmy MI, Sadek MA, Abdou K, El-Dessouki AM, El-Shiekh RA, Khalaf SS. Orientin: a comprehensive review of a promising bioactive flavonoid. Inflammopharmacology 2025; 33:1713-1728. [PMID: 40056319 PMCID: PMC11991976 DOI: 10.1007/s10787-025-01690-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 02/10/2025] [Indexed: 03/10/2025]
Abstract
Medicinal herbs continue to play an important part in modern drugs and healthcare because customers think that most of them have fewer or milder side effects than traditional modern medicines. Bioactive compounds are typically isolated from plants before being used as a source of therapeutic medicines. As a result, extracting bioactive compounds from medicinal plants is an important step in developing plant-based medications. Orientin is a flavonoid C-glycoside found in many plants, is frequently used in bioactivity studies due to its numerous beneficial properties, which include antioxidants, antiaging, anti-inflammation, vasodilation and cardioprotective, neuroprotective, antidiabetic, hepatoprotective, and adaptogenic effects. In this review, the comprehensive search for the health benefits of orientin was traced. The findings reflected that orientin could be considered one of the important natural candidates as a potential nutraceutical. This underscores its promising attributes and potential applications in health and wellness. Further research may be guaranteed to fully elucidate its benefits and mechanisms of action.
Collapse
Affiliation(s)
- Mohamed I Fahmy
- Department of Pharmacology and Toxicology, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), Giza, Egypt
| | - Mohamed A Sadek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Kareem Abdou
- College of Pharmacy, Al-Ain University, Abu Dhabi, United Arab Emirates
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ahmed M El-Dessouki
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza, 12566, Egypt
| | - Riham A El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, 11562, Egypt.
| | - Samar S Khalaf
- Biochemistry Department Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| |
Collapse
|
10
|
Harada T, Kondo H, Nakamura K, He Y, Goto S, Takahashi M, Yamasaki H, Matsuda N, Takano M, Abe I, Fukui A, Akioka H, Teshima Y, Yufu K, Shibata H, Takahashi N. Soluble Guanylate Cyclase Stimulator Vericiguat Attenuates Angiotensin II-Induced Oxidative Stress and Cardiac Remodeling. Circ J 2025:CJ-24-0659. [PMID: 40128921 DOI: 10.1253/circj.cj-24-0659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
BACKGROUND Vericiguat, an oral soluble guanylate cyclase stimulator, is a novel therapeutic agent for patients with heart failure with reduced ejection fraction; however, the detailed cardioprotective mechanism remains unclear. We aimed to explore the mechanism of the effect of vericiguat on the myocardium, particularly focusing on oxidative stress, using in vivo and in vitro experiments. METHODS AND RESULTS Male 8-week-old mice were divided into a control group, angiotensin II (AngII) infusion group, and AngII infusion with low- or high-dose vericiguat treatment group. After 14 days of treatment, vericiguat did not affect the systolic or diastolic blood pressure increase caused by AngII infusion. AngII-induced cardiac hypertrophy and fibrosis in the left ventricle (LV) were significantly ameliorated by high-dose vericiguat treatment. AngII-induced O2-overproduction and upregulation of messenger RNA levels of Nppa, Nppb, Myh7, Col1a1, Col3A1, and Tgfb1 in the LV were significantly attenuated by vericiguat in a dose-dependent manner. Incubation of neonatal rat cardiomyocytes using vericiguat and AngII revealed that preceding incubation with vericiguat directly reduced AngII-induced cardiomyocyte O2-production and cardiac hypertrophy-associated gene expression. In addition, AngII-induced phosphorylation of ERK 1/2 or p38 MAPK was significantly attenuated by the incubation with vericiguat. CONCLUSIONS Our study demonstrated that vericiguat suppresses myocardial oxidative stress via the regulation of ERK 1/2 or p38 MAPK signaling, leading to antihypertrophic/fibrotic effects.
Collapse
Affiliation(s)
- Taisuke Harada
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University
| | - Hidekazu Kondo
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University
| | - Kodai Nakamura
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University
| | - Yu He
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University
| | - Shunsuke Goto
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University
| | - Masaki Takahashi
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University
| | - Hirochika Yamasaki
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University
| | - Naoki Matsuda
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University
| | - Masayuki Takano
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University
| | - Ichitaro Abe
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University
| | - Akira Fukui
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University
| | - Hidefumi Akioka
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University
| | - Yasushi Teshima
- Department of Advanced Medical Sciences, Oita University Hospital
| | - Kunio Yufu
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University
| | - Hirotaka Shibata
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University
| | - Naohiko Takahashi
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University
| |
Collapse
|
11
|
van de Bovenkamp AA, Nassiri S, Bakermans AJ, Burchell GL, de Man FS, van Loon RB, Handoko ML. Long-term hemodynamic responses and reverse remodeling after pharmacotherapy in HFpEF versus HFrEF: a systematic review and meta-analysis. Am J Physiol Heart Circ Physiol 2025; 328:H419-H432. [PMID: 39825764 DOI: 10.1152/ajpheart.00544.2024] [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: 08/08/2024] [Revised: 09/06/2024] [Accepted: 12/13/2024] [Indexed: 01/20/2025]
Abstract
The acute response to therapeutic afterload reduction differs between heart failure with preserved (HFpEF) versus reduced ejection fraction (HFrEF), with larger left ventricular (LV) stroke work augmentation in HFrEF compared with HFpEF. This may (partially) explain the neutral effect of HFrEF-medication in HFpEF. It is unclear whether such differences in hemodynamic response persist and/or differentially trigger reverse remodeling in the case of long-term afterload reduction. A systematic search was performed, identifying 21 clinical trials investigating renin-angiotensin-aldosterone system (RAAS) inhibitors, β-blockers, and sodium-glucose cotransport 2 inhibitors that report data on afterload reduction, stroke volume, and reverse remodeling in HFpEF and/or HFrEF. In both HFpEF and HFrEF, meta-analyses revealed limited long-term change in systolic/diastolic blood pressure (-5.6/-3.2 and -4.6/-1.4 mmHg, respectively) and LV afterload reduction (arterial elastance: -0.039 and -0.055 mmHg/mL, respectively). Long-term treatment did not result in an increase in stroke volume, with the exception of β-blockers in HFrEF. Indexed LV mass decreased slightly in both HFpEF and HFrEF (-2.8 and -2.3 g/m2, respectively). In HFrEF, treatment reduced LV end-diastolic and end-systolic volume (-8 and -6 mL, respectively), whereas in HFpEF there was no relevant change. Contrary to acute heart failure studies, long-term afterload reduction had little effect on blood pressure and stroke volume augmentation in both HFpEF and HFrEF. However, reverse remodeling was clearly present in HFrEF but was essentially absent in HFpEF.
Collapse
Affiliation(s)
- Arno A van de Bovenkamp
- Department of Cardiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart failure & Arrhythmias, Amsterdam, The Netherlands
| | - Soufiane Nassiri
- Department of Cardiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart failure & Arrhythmias, Amsterdam, The Netherlands
| | - Adrianus J Bakermans
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - George L Burchell
- Medical Library, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Frances S de Man
- Amsterdam Cardiovascular Sciences, Heart failure & Arrhythmias, Amsterdam, The Netherlands
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ramon B van Loon
- Department of Cardiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart failure & Arrhythmias, Amsterdam, The Netherlands
| | - M Louis Handoko
- Department of Cardiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart failure & Arrhythmias, Amsterdam, The Netherlands
- Department of Cardiology, UMC Utrecht, Utrecht, The Netherlands
| |
Collapse
|
12
|
Sun Y, Xiao L, Chen L, Wang X. Doxorubicin-Induced Cardiac Remodeling: Mechanisms and Mitigation Strategies. Cardiovasc Drugs Ther 2025:10.1007/s10557-025-07673-6. [PMID: 40009315 DOI: 10.1007/s10557-025-07673-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/16/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND The therapeutic prowess of doxorubicin in oncology is marred by its cardiotoxic consequences, manifesting as cardiac remodeling. Pathophysiological alterations triggered by doxorubicin include inflammatory cascades, fibrotic tissue deposition, vascular and valvular changes, and finally cardiomyopathy. These multifarious consequences collectively orchestrate the deterioration of cardiac architecture and function. METHOD By charting the molecular underpinnings and remedial prospects, this review aspires to contribute a novel perspective using latest publications to the ongoing quest for cardioprotection in cancer therapy. RESULTS AND DISCUSSION Experimental analyses demonstrate the pivotal roles of oxidative stress and subsequent necrosis and apoptosis of cardiomyocytes, muscle cells, endothelial cells, and small muscle cells in different parts of the heart. In addition, severe and unusual infiltration of macrophages, mast cells, and neutrophils can amplify oxidative damage and subsequent impacts such as chronic inflammatory responses, vascular and valvular remodeling, and fibrosis. These modifications can render cardiomyopathy, ischemia, heart attack, and other disorders. In an endeavor to counteract these ramifications, a spectrum of emerging adjuvants and strategies are poised to fortify the heart against doxorubicin's deleterious effects. CONCLUSION The compendium of mitigation tactics such as innovative pharmacological agents hold the potential to attenuate the cardiotoxic burden.
Collapse
Affiliation(s)
- Yanna Sun
- Department of Cardiology, The First Affiliated of Zhengzhou University, Zhengzhou City Henan Province, 450052, China
| | - Lili Xiao
- Department of Cardiology, The First Affiliated of Zhengzhou University, Zhengzhou City Henan Province, 450052, China
| | - Linlin Chen
- Department of Cardiology, The First Affiliated of Zhengzhou University, Zhengzhou City Henan Province, 450052, China
| | - Xiaofang Wang
- Department of Cardiology, The First Affiliated of Zhengzhou University, Zhengzhou City Henan Province, 450052, China.
| |
Collapse
|
13
|
da Costa Salomão KC, da Silva MC, Fabiano LC, de Freitas PLZ, Neves CQ, Borges SC, Breithaupt-Faloppa AC, Barbosa CP, Buttow NC. Cardiotoxicity Associated With a Low Doses of 5-FU Promotes Morphoquantitative Changes in the Intrinsic Cardiac Nervous System. Cardiovasc Toxicol 2025; 25:193-204. [PMID: 39864046 DOI: 10.1007/s12012-024-09958-y] [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: 12/11/2023] [Accepted: 12/30/2024] [Indexed: 01/27/2025]
Abstract
5-Fluorouracil (5-FU) is a chemotherapeutic that is used to treat solid tumors. However, 5-FU is associated with several side effects, including cardiotoxicity. Considering the importance of the intrinsic cardiac nervous system (ICNS) for the heart and that little is known about effects of 5-FU on this nervous system plexus, the purpose of the present study was to evaluate effects 5-FU at a low dose on the ICNS and oxidative and inflammatory effects in the heart in Wistar rats. The rats were divided into two groups: treated and 5-FU (n = 6/group). The control group received saline only. The treated group received the following clinical doses of 5-FU: 15 mg/kg for 4 consecutive days, followed by 6 mg/kg for 4 days alternated with non-treatment days, and finally 15 mg/kg as the last dose on day 14. On day 15, the rats were euthanized and underwent thoracotomy. The atria were used for histological analysis, and the ventricles were used for biochemical analysis. The results showed an increase in neuronal density and a decrease in ganglionic and neuronal area in the ICNS. Furthermore, tissue inflammation was observed, indicated by an increase in proinflammatory factors and the enzymatic activity of myeloperoxidase and n-acetyl-glucosaminidase. Oxidative stress was also observed, confirmed by a reduction of endogenous antioxidant defenses and the presence of lipoperoxidation. Treatment with 5-FU also caused cardiac atrophy and fibrosis. These findings indicate that cardiotoxicity is present with 5-FU treatment and affects the morphometric aspects of the ICNS.
Collapse
Affiliation(s)
| | - Mariana Conceição da Silva
- Department of Structural and Functional Biology, State University of Campinas, Campinas, Sao Paulo, Brazil
| | - Lilian Catarim Fabiano
- Department of Morphological Sciences, State University of Maringa, Maringa, Parana, Brazil
| | - Pedro Luiz Zonta de Freitas
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação, Instituto do Coração (Incor), Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | | | | - Ana Cristina Breithaupt-Faloppa
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação, Instituto do Coração (Incor), Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | | - Nilza Cristina Buttow
- Department of Morphological Sciences, State University of Maringa, Maringa, Parana, Brazil.
| |
Collapse
|
14
|
Aniekeme E, De Souza Goncalves B, Sodhi K, Rueda Rios C, Thompson E. Predictors of Mortality in Acute Myocardial Infarction Patients With Systemic Lupus Erythematosus. Cureus 2025; 17:e79578. [PMID: 40008104 PMCID: PMC11856213 DOI: 10.7759/cureus.79578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2025] [Indexed: 02/27/2025] Open
Abstract
Introduction Acute myocardial infarction (AMI) remains a leading cause of mortality globally, with systemic lupus erythematosus (SLE) posing additional risks for adverse outcomes. Methods This retrospective cohort study utilized data from the Healthcare Cost and Utilization Project National Inpatient Sample from 2016 to 2020 to investigate predictors of mortality among hospitalized AMI patients, stratified by SLE status. Results A total of 81,935 patients were included, with variables analyzed using multivariate logistic regression. Among SLE patients, being female, aged ≥65 years, and having a prolonged hospital stay >5 days were associated with higher mortality. Elective admissions were protective while non-Hispanic Black SLE patients showed reduced odds of mortality compared to their non-Hispanic White counterparts. Among non-SLE patients, predictors of increased mortality included being non-Hispanic Black and having prolonged hospital stays. Private insurance coverage was associated with lower mortality in this group. Conclusion These findings highlight critical patient- and hospital-related factors influencing mortality in AMI patients with and without SLE. Targeted strategies focusing on early recognition, effective interventions, and reducing healthcare disparities are essential to improve outcomes in this population.
Collapse
Affiliation(s)
- Etuk Aniekeme
- Department of Cardiology, Marshall University Joan C. Edwards School of Medicine, Huntington, USA
| | - Bruno De Souza Goncalves
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, USA
- Department of Biomedical Sciences, Marshall University Joan C. Edwards School of Medicine, Huntington, USA
| | - Komal Sodhi
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, USA
- Department of Biomedical Sciences, Marshall University Joan C. Edwards School of Medicine, Huntington, USA
- Department of Cardiology, Marshall University Joan C. Edwards School of Medicine, Huntington, USA
| | - Carlos Rueda Rios
- Department of Cardiology, Marshall University Joan C. Edwards School of Medicine, Huntington, USA
| | - Ellen Thompson
- Department of Cardiology, Marshall University Joan C. Edwards School of Medicine, Huntington, USA
| |
Collapse
|
15
|
Laska M, Vitous J, Jirik R, Hendrych M, Drazanova E, Kratka L, Nadenicek J, Novakova M, Stracina T. Heart remodelling affects ECG in rat DOCA/salt model. Physiol Res 2024; 73:S727-S753. [PMID: 39808174 PMCID: PMC11827063 DOI: 10.33549/physiolres.935512] [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: 07/09/2024] [Accepted: 10/24/2024] [Indexed: 01/18/2025] Open
Abstract
Myocardial remodelling involves structural and functional changes in the heart, potentially leading to heart failure. The deoxycorticosterone acetate (DOCA)/salt model is a widely used experimental approach to study hypertension-induced cardiac remodelling. It allows to investigate the mechanisms underlying myocardial fibrosis and hypertrophy, which are key contributors to impaired cardiac function. In this study, myocardial remodelling in rat deoxycorticosterone acetate/salt model was examined over a three-week period. The experiment involved 11 male Sprague-Dawley rats, divided into two groups: fibrosis (n=6) and control (n=5). Myocardial remodelling was induced in the fibrosis group through unilateral nephrectomy, deoxyco-rticosterone acetate administration, and increased salt intake. The results revealed significant structural changes, including increased left ventricular wall thickness, myocardial fractional volume, and development of myocardial fibrosis. Despite these changes, left ventricular ejection fraction was preserved and even increased. ECG analysis showed significant prolongation of the PR interval and widening of the QRS complex in the fibrosis group, indicating disrupted atrioventricular and ventricular conduction, likely due to fibrosis and hypertrophy. Correlation analysis suggested a potential relationship between QRS duration and myocardial hypertrophy, although no significant correlations were found among other ECG parameters and structural changes detected by MRI. The study highlights the advantage of the DOCA/salt model in exploring the impact of myocardial remodelling on electrophysiological properties. Notably, this study is among the first to show that early myocardial remodelling in this model is accompanied by distinct electrophysiological changes, suggesting that advanced methods combined with established animal models can open new opportunities for research in this field. Key words Myocardial fibrosis, Remodelling, Animal model, DOCA-salt, Magnetic resonance imaging.
Collapse
Affiliation(s)
- M Laska
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Alonazi AS, Almodawah S, Aldigi R, Bin Dayel A, Alamin M, Almotairi AR, El-Tohamy MF, Alharbi H, Ali R, Alshammari TK, Alrasheed NM. Potential cardioprotective effect of paroxetine against ventricular remodeling in an animal model of myocardial infarction: a comparative study. BMC Pharmacol Toxicol 2024; 25:99. [PMID: 39696491 DOI: 10.1186/s40360-024-00824-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Post-myocardial infarction (MI) remodeling involves various structural and functional changes, such as inflammation and fibrosis. Upregulation of G protein-coupled receptor kinase 2 (GRK2) is linked to the progression of cardiovascular diseases, including myocardial infarction. The inhibitory effects of paroxetine on GRK2 are recognized, yet its protective effect on post-MI remodeling has not been elucidated. Here, we investigated the cardioprotective effect of paroxetine in an animal model of MI, focusing on post-MI cardiac remodeling and comparing its effect to a β-blocker and an angiotensin receptor antagonist. METHODS Albino Wistar rats were divided into five groups (control; untreated MI; and MI pre-treated with either paroxetine, metoprolol, or irbesartan). MI was induced using isoproterenol (100 mg.kg-1) on days 16 and 17. Cardioprotective effects were determined by assessing markers of cardiac injury, histopathology, inflammation, oxidative stress, and fibrosis. Statistical analysis performed using a one-way analysis of variance, followed by an appropriate post hoc test, the differences between the groups were considered significant when the (P < 0.05). RESULTS Paroxetine significantly attenuated cardiac injury biomarkers including serum Tn-I and CK-MB levels. In terms of cardiac remodeling, paroxetine significantly reduced the relative HW/BW index and the plasms FGF23 level. Furthermore, it modulated markers of fibrosis, inflammation, and oxidative stress. CONCLUSION The current findings suggest that pre-treatment with paroxetine may exert a beneficial effect that protects against post-MI remodeling, including modulating fibrotic, inflammatory, and angiogenesis-related factors. Therefore, the current findings show the promising role of paroxetine as a cardioprotective that attenuates post-MI remodeling processes.
Collapse
Affiliation(s)
- Asma S Alonazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| | - Sara Almodawah
- College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Rana Aldigi
- College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Anfal Bin Dayel
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Maha Alamin
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ahmad R Almotairi
- Department of Pathology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Maha F El-Tohamy
- Department of Chemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Hana Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Rehab Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Tahani K Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Nouf M Alrasheed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
17
|
Shahsavarnajand Bonab H, Tolouei Azar J, Soraya H, Nouri Habashi A. Aerobic interval training preconditioning protocols inhibit isoproterenol-induced pathological cardiac remodeling in rats: Implications on oxidative balance, autophagy, and apoptosis. SPORTS MEDICINE AND HEALTH SCIENCE 2024; 6:344-357. [PMID: 39309465 PMCID: PMC11411311 DOI: 10.1016/j.smhs.2024.01.010] [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: 08/02/2023] [Revised: 01/23/2024] [Accepted: 01/31/2024] [Indexed: 09/25/2024] Open
Abstract
This study aimed to investigate the potential cardioprotective effects of moderate and high-intensity aerobic interval training (MIIT and HIIT) preconditioning. The focus was on histological changes, pro-oxidant-antioxidant balance, autophagy initiation, and apoptosis in myocardial tissue incited by isoproterenol-induced pathological cardiac remodeling (ISO-induced PCR). Male Wistar rats were randomly divided into control (n = 6), ISO (n = 8), MIIT (n = 4), HIIT (n = 4), MIIT + ISO (n = 8), and HIIT + ISO (n = 8) groups. The MIIT and HIIT protocols were administered for 10 weeks, followed by the induction of cardiac remodeling using subcutaneous injection of ISO (100 mg/kg for two consecutive days). Alterations in heart rate (HR), mean arterial pressure (MAP), rate pressure product (RPP), myocardial oxygen consumption (MV ˙ O2), cardiac hypertrophy, histopathological changes, pro-oxidant-antioxidant balance, autophagy biomarkers (Beclin-1, Atg7, p62, LC3 I/II), and apoptotic cell distribution were measured. The findings revealed that the MIIT + ISO and HIIT + ISO groups demonstrated diminished myocardial damage, hemorrhage, immune cell infiltration, edema, necrosis, and apoptosis compared to ISO-induced rats. MIIT and HIIT preconditioning mitigated HR, enhanced MAP, and preserved MV ˙ O2 and RPP. The pro-oxidant-antioxidant balance was sustained in both MIIT + ISO and HIIT + ISO groups, with MIIT primarily inhibiting pro-apoptotic autophagy progression through maintaining pro-oxidant-antioxidant balance, and HIIT promoting pro-survival autophagy. The results demonstrated the beneficial effects of both MIIT and HIIT as AITs preconditioning in ameliorating ISO-induced PCR by improving exercise capacity, hemodynamic parameters, and histopathological changes. Some of these protective effects can be attributed to the modulation of cardiac apoptosis, autophagy, and oxidative stress.
Collapse
Affiliation(s)
- Hakimeh Shahsavarnajand Bonab
- Department of Exercise Physiology and Corrective Exercises, Faculty of Sport Sciences, Urmia University, Urmia, Iran
| | - Javad Tolouei Azar
- Department of Exercise Physiology and Corrective Exercises, Faculty of Sport Sciences, Urmia University, Urmia, Iran
| | - Hamid Soraya
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Akbar Nouri Habashi
- Department of Exercise Physiology and Corrective Exercises, Faculty of Sport Sciences, Urmia University, Urmia, Iran
| |
Collapse
|
18
|
Yerra VG, Connelly KA. Extrarenal Benefits of SGLT2 Inhibitors in the Treatment of Cardiomyopathies. Physiology (Bethesda) 2024; 39:0. [PMID: 38888433 DOI: 10.1152/physiol.00008.2024] [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: 02/01/2024] [Revised: 06/05/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors have emerged as pivotal medications for heart failure, demonstrating remarkable cardiovascular benefits extending beyond their glucose-lowering effects. The unexpected cardiovascular advantages have intrigued and prompted the scientific community to delve into the mechanistic underpinnings of these novel actions. Preclinical studies have generated many mechanistic theories, ranging from their renal and extrarenal effects to potential direct actions on cardiac muscle cells, to elucidate the mechanisms linking these drugs to clinical cardiovascular outcomes. Despite the strengths and limitations of each theory, many await validation in human studies. Furthermore, whether SGLT2 inhibitors confer therapeutic benefits in specific subsets of cardiomyopathies akin to their efficacy in other heart failure populations remains unclear. By examining the shared pathological features between heart failure resulting from vascular diseases and other causes of cardiomyopathy, certain specific molecular actions of SGLT2 inhibitors (particularly those targeting cardiomyocytes) would support the concept that these medications will yield therapeutic benefits across a broad range of cardiomyopathies. This article aims to discuss the important mechanisms of SGLT2 inhibitors and their implications in hypertrophic and dilated cardiomyopathies. Furthermore, we offer insights into future research directions for SGLT2 inhibitor studies, which hold the potential to further elucidate the proposed biological mechanisms in greater detail.
Collapse
Affiliation(s)
- Veera Ganesh Yerra
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada
| | - Kim A Connelly
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada
| |
Collapse
|
19
|
El Khayari A, Hakam SM, Malka G, Rochette L, El Fatimy R. New insights into the cardio-renal benefits of SGLT2 inhibitors and the coordinated role of miR-30 family. Genes Dis 2024; 11:101174. [PMID: 39224109 PMCID: PMC11367061 DOI: 10.1016/j.gendis.2023.101174] [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: 02/01/2023] [Revised: 09/15/2023] [Accepted: 10/10/2023] [Indexed: 09/04/2024] Open
Abstract
Sodium-glucose co-transporter inhibitors (SGLTis) are the latest class of anti-hyperglycemic agents. In addition to inhibiting the absorption of glucose by the kidney causing glycosuria, these drugs also demonstrate cardio-renal benefits in diabetic subjects. miR-30 family, one of the most abundant microRNAs in the heart, has recently been linked to a setting of cardiovascular diseases and has been proposed as novel biomarkers in kidney dysfunctions as well; their expression is consistently dysregulated in a variety of cardio-renal dysfunctions. The mechanistic involvement and the potential interplay between miR-30 and SGLT2i effects have yet to be thoroughly elucidated. Recent research has stressed the relevance of this cluster of microRNAs as modulators of several pathological processes in the heart and kidneys, raising the possibility of these small ncRNAs playing a central role in various cardiovascular complications, notably, endothelial dysfunction and pathological remodeling. Here, we review current evidence supporting the pleiotropic effects of SGLT2is in cardiovascular and renal outcomes and investigate the link and the coordinated implication of the miR-30 family in endothelial dysfunction and cardiac remodeling. We also discuss the emerging role of circulating miR-30 as non-invasive biomarkers and attractive therapeutic targets for cardiovascular diseases and kidney diseases. Clinical evidence, as well as metabolic, cellular, and molecular aspects, are comprehensively covered.
Collapse
Affiliation(s)
- Abdellatif El Khayari
- Institute of Biological Sciences (ISSB-P), UM6P Faculty of Medical Sciences, Mohammed VI Polytechnic University (UM6P), Ben-Guerir 43150, Morocco
| | - Soukaina Miya Hakam
- Institute of Biological Sciences (ISSB-P), UM6P Faculty of Medical Sciences, Mohammed VI Polytechnic University (UM6P), Ben-Guerir 43150, Morocco
| | - Gabriel Malka
- Institute of Biological Sciences (ISSB-P), UM6P Faculty of Medical Sciences, Mohammed VI Polytechnic University (UM6P), Ben-Guerir 43150, Morocco
| | - Luc Rochette
- Equipe d'Accueil (EA 7460): Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Université de Bourgogne – Franche Comté, Faculté des Sciences de Santé, 7 Bd Jeanne d'Arc, Dijon 21000, France
| | - Rachid El Fatimy
- Institute of Biological Sciences (ISSB-P), UM6P Faculty of Medical Sciences, Mohammed VI Polytechnic University (UM6P), Ben-Guerir 43150, Morocco
| |
Collapse
|
20
|
Salles ACP, Alexandre-Santos B, de Souza Carvalho T, Proença AB, Sepúlveda-Fragoso V, Fernandes T, Oliveira EM, da Nóbrega ACL, Frantz EDC, Magliano DC. ER stress improvement by aerobic training or enalapril differently ameliorates pathological cardiac remodeling in obese mice. Mol Cell Biochem 2024; 479:3167-3179. [PMID: 38308790 DOI: 10.1007/s11010-024-04925-1] [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: 07/19/2023] [Accepted: 01/05/2024] [Indexed: 02/05/2024]
Abstract
Overactivation of the classic arm of the renin-angiotensin system (RAS) is one of the main mechanisms involved in obesity-related cardiac remodeling, and a possible relationship between RAS and ER stress in the cardiovascular system have been described. Thus, the aim of this study is to evaluate if activating the protective arm of the RAS by ACE inhibition or aerobic exercise training could overturn diet-induced pathological cardiac hypertrophy by attenuating ER stress. Male C57BL/6 mice were fed a control (SC) or a high-fat diet (HF) for 16 weeks. In the 8th week, HF-fed animals were randomly divided into HF, enalapril treatment (HF-En), and aerobic exercise training (HF-Ex) groups. Body mass (BM), food and energy intake, plasma analyzes, systolic blood pressure (SBP), physical conditioning, and plasma ACE and ACE2 activity were evaluated. Cardiac morphology, and protein expression of hypertrophy, cardiac metabolism, RAS, and ER stress markers were assessed. Data presented as mean ± standard deviation and analyzed by one-way ANOVA with Holm-Sidak post-hoc. HF group had increased BM and SBP, and developed pathological concentric cardiac hypertrophy, with overactivation of the classic arm of the RAS, and higher ER stress. Both interventions reverted the increase in BM, and SBP, and favored the protective arm of the RAS. Enalapril treatment improved pathological cardiac hypertrophy with partial reversal of the concentric pattern, and slightly attenuated cardiac ER stress. In contrast, aerobic exercise training induced physiological eccentric cardiac hypertrophy, and fully diminished ER stress.
Collapse
Affiliation(s)
- Amanda Conceição Pimenta Salles
- Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
- Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Beatriz Alexandre-Santos
- Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
- Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Thais de Souza Carvalho
- Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Ana Beatriz Proença
- Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Vinicius Sepúlveda-Fragoso
- Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Tiago Fernandes
- Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Edilamar Menezes Oliveira
- Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, SP, Brazil
- National Institute for Science & Technology - INCT (In)activity & Exercise, CNPq, Niteroi, RJ, Brazil
| | - Antonio Claudio Lucas da Nóbrega
- Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
- National Institute for Science & Technology - INCT (In)activity & Exercise, CNPq, Niteroi, RJ, Brazil
| | - Eliete Dalla Corte Frantz
- Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
- Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
- National Institute for Science & Technology - INCT (In)activity & Exercise, CNPq, Niteroi, RJ, Brazil
| | - D'Angelo Carlo Magliano
- Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil.
- Department of Morphology, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil.
| |
Collapse
|
21
|
Leem AY, Yu HT, Sung M, Chung KS, Kim Y, Woo A, Kim SY, Park MS, Kim YS, Yang YH, Kim HE, Lee JG, Kim K, Kim KB, Joung B, Park J, Lee SH. Clinical implication of electrocardiogram change in patients experiencing lung transplantation with end stage lung disease. Front Physiol 2024; 15:1440307. [PMID: 39539957 PMCID: PMC11558550 DOI: 10.3389/fphys.2024.1440307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction End-stage lung disease causes cardiac remodeling and induces electrocardiogram (ECG) changes. On the other way, whether lung transplantation (LTx) in end-stage lung disease patients are associated with ECG change is unknown. The object of this study was to investigate ECG changes before and after LTx in end-stage lung disease patients and whether these changes had clinical significance. Method This was a single-center retrospective cohort study of 280 end-stage lung disease patients who consecutively underwent LTx at a tertiary referral hospital. ECG findings before LTx and within 1 week and 1, 3, and 6 months after LTx were obtained and analyzed. To find clinical meaning, the ECG at 1 month after LTx was analyzed according to 1-year survival (survivor vs non-survivor groups). Survival data were estimated using the Kaplan-Meier method. Results Significant differences were observed in the PR interval, QRS duration, QT interval, QTc interval, and heart rate before LTx and 1 month after LTx; the PR interval, QRS duration, QTc interval, and heart rate were decreased. Particularly, the QTc interval was significantly decreased 1 month after LTx, whereas there was no significant change in the QTc interval from 1 to 6 months thereafter. The PR interval, QT interval, QTc interval, and heart rate were significantly different between the survivor and non-survivor groups. The serial changes in QTc interval before LTx and 1 and 3 months after LTx were also significantly different between the survivor and non-survivor groups (p = 0.040 after adjusting for age and body mass index). Upon dividing the patients based on the range of QTc interval change ≤ -8 ms, >-8-10 ms, >10-35 ms, >35 ms), the survival rate was significantly lower in the group whose QTc interval at 1 month after LTx decreased by > 35 m (p = 0.019). Conclusion LTx in patients with end-stage lung disease may induce ECG changes. Patients whose QTc interval at 1 month after LTx decreased by > 35 ms have a significantly higher 1-year mortality rate. Hence, these ECG changes may have clinical and prognostic significance.
Collapse
Affiliation(s)
- Ah Young Leem
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hee Tae Yu
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - MinDong Sung
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyung Soo Chung
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yeonkyeong Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ala Woo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Song Yee Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Moo Suk Park
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Sam Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Ho Yang
- Department of Thoracic and Cardiovascular Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ha Eun Kim
- Department of Thoracic and Cardiovascular Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin Gu Lee
- Department of Thoracic and Cardiovascular Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyuseok Kim
- Department of Biomedical Engineering, Eulji University, Seoul, Republic of Korea
| | - Kyu Bom Kim
- Department of Radiation Convergence Engineering, College of Health Science, Yonsei University, Wonju, Gangwon, Republic of Korea
| | - Boyoung Joung
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Junbeom Park
- Division of Cardiology, Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
- Department of Biomedical Engineering, Emory University School of Medicine, Atlanta, GA, United States
| | - Su Hwan Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
22
|
Zia-Ul-Sabah, Alqahtani SAM, Alghamdi BH, Wani JI, Aziz S, Durrani HK, Patel AA, Rangraze I, Wani SJ. Association of type-D personality and left-ventricular remodelling in patients treated with primary percutaneous intervention after ST-segment elevation myocardial infarction. BMC Cardiovasc Disord 2024; 24:600. [PMID: 39468433 PMCID: PMC11520066 DOI: 10.1186/s12872-024-04254-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 10/14/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Type-D personality is an established predisposing factor for various diseases. Type-D traits have been shown to pose a 26% increased risk of coronary artery disease after controlling for other confounding factors. Significant associations have been reported between type-D personality traits and dyslipidaemia, impaired endothelial function, coronary heart disease (CAD), acute myocardial infarction, and other adverse cardiovascular events. OBJECTIVE To assess the association between type-D personality and left-ventricular adverse remodelling in patients treated with percutaneous coronary intervention following index ST-segment elevation myocardial infarction. METHODS All patients hospitalized and treated with percutaneous coronary intervention (PCI) after their index ST-segment elevation myocardial infarction (STEMI) between 1 January 2022 to 31 December 2023 were prospectively enrolled. Type-D personality traits in the study population were determined at baseline using type-D Scale-14 (DS14) instrument, whereas any positive change in left ventricular end diastolic volume (LVEDV) ≥ 20% at follow up period of 12-months from baseline was defined as left-ventricular adverse remodelling (LVAR). Univariate and multivariate analysis was done to establish the independent predictors of LVAR. The area under receiver-operating characteristic curve (AUROC) was employed to assess the sensitivity and specificity of the identified independent predictors. RESULTS A total of 124 patients were enrolled in the study. The mean age of the study population was 67 ± 10 years and the overall incidence of LVAR was found to be 25%. Multivariate regression analysis revealed that type-D personality is a significant independent predictor of LVAR [Formula: see text] apart from the already established independent predictors Killip Class[Formula: see text], baseline Global Longitudinal strain (GLS)[Formula: see text], and 3-vessel CAD[Formula: see text]. In ROC curve analysis type-D personality as an independent predictor of LVAR achieved a sensitivity of 41.4% and a specificity of 87.1%, p < 0.02. CONCLUSION Type-D personality trait is a significant independent predictor of LVAR in patients treated with PCI after their index-STEMI.
Collapse
Affiliation(s)
- Zia-Ul-Sabah
- Department of Medicine, College of Medicine, King Khalid University, Abha, Saudi Arabia.
| | | | - Bandar Hezam Alghamdi
- Prince Faisal bin Khalid Cardiac Center, King Faisal Medical City, Abha, Saudi Arabia
| | - Javed Iqbal Wani
- Department of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Shahid Aziz
- Department of Medicine, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | | | - Ayyub Ali Patel
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Imran Rangraze
- Department of Internal Medicine, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, UAE
| | | |
Collapse
|
23
|
Fatima M, Andleeb H, Rehman T, Gul O, Azeezz S, Rehman H, Nawaz H. Effect of alantolactones on cardiac parameters of animals under artificially induced oxidative stress. Biomedicine (Taipei) 2024; 14:12-22. [PMID: 39386184 PMCID: PMC11460572 DOI: 10.37796/2211-8039.1457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 10/12/2024] Open
Abstract
Purpose Phytochemicals have been found effective in reducing the oxidative stress and damage to cardiovascular and other tissues. In this study, the effects of alantolactone (AL) on cardiac parameters in rabbits exposed to artificially-induced oxidative stress were investigated. Method The oxidative stress was induced in a group of White New Zealand rabbits by injecting 40% hydrogen peroxide solution (1 ml/kg body weight) thrice with an interval of 72 h. The hydrogen peroxide-treated animals were orally treated with AL extracted from the roots of Inula helenium (1 ml/kg repeated thrice after 72 h). Blood samples were taken before and after the hydrogen peroxide and AL treatments, and the sera were subjected to analysis of oxidative damage in terms of malondialdehyde content (MDA), total antioxidant activity (TAOA), linoleic acid reduction capacity (LARC), hydroxyl radical scavenging capacity (HRSC), 2,2-diphenyl-1-picrylhydrazyl radical scavenging capacity (DPPH RSC), superoxide dismutase activity (SOD) and catalase activity, and cardiac parameters including troponin-I content (Trop-I), creatine kinase-MB (CKMB), aspartate transaminase (AST). Results The hydrogen peroxide treatment substantially enhanced MDA content and SOD activity and decreased LARC, HRSC, DPPH, and catalase activity. The AL treatment significantly decreased MDA content, TAOA, Trop-I, CK-MB, and AST levels and increased LARC, DPPH RSC, HRSC, and catalase activity. Conclusion The observed effect of AL treatment on the animals' oxidative stress, antioxidant status, and cardiac biomarkers emphasizes that AL may effectively manage oxidative stress and cardiac damage.
Collapse
Affiliation(s)
- Mishal Fatima
- Department of Biochemistry, Bahauddin Zakariya University, Multan, 60800,
Pakistan
| | - Hina Andleeb
- Department of Biochemistry, Bahauddin Zakariya University, Multan, 60800,
Pakistan
| | - Tanzila Rehman
- Department of Chemistry, The Women University Multan, Multan,
Pakistan
| | - Ouz Gul
- Department of Biochemistry, Bahauddin Zakariya University, Multan, 60800,
Pakistan
| | - Shanza Azeezz
- Department of Biochemistry, Bahauddin Zakariya University, Multan, 60800,
Pakistan
| | - Huzaifa Rehman
- Department of Biochemistry, Bahauddin Zakariya University, Multan, 60800,
Pakistan
| | - Haq Nawaz
- Department of Biochemistry, Bahauddin Zakariya University, Multan, 60800,
Pakistan
| |
Collapse
|
24
|
Sobhy MH, Ismail A, Abdel-Hamid MS, Wagih M, Kamel M. 2-Methoxyestradiol ameliorates doxorubicin-induced cardiotoxicity by regulating the expression of GLUT4 and CPT-1B in female rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7129-7139. [PMID: 38652282 PMCID: PMC11422279 DOI: 10.1007/s00210-024-03073-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 03/27/2024] [Indexed: 04/25/2024]
Abstract
The clinical usage of doxorubicin (DOX) is hampered due to cardiomyopathy. Studies reveal that estrogen (E2) modulates DOX-induced cardiotoxicity. Yet, the exact mechanism is unclear. The objective of the current study is to evaluate the influence of E2 and more specifically its metabolite 2-methoxyestradiol (2ME) on cardiac remodeling and the reprogramming of cardiac metabolism in rats subjected to DOX cardiotoxicity. Seventy-two female rats were divided into groups. Cardiotoxicity was induced by administering DOX (2.5 mg/kg three times weekly for 2 weeks). In some groups, the effect of endogenous E2 was abolished by ovariectomy (OVX) or by using the estrogen receptor (ER) blocker Fulvestrant (FULV). The effect of administering exogenous E2 or 2ME in the OVX group was studied. Furthermore, the influence of entacapone (COMT inhibitor) on induced cardiotoxicity was investigated. The evaluated cardiac parameters included ECG, histopathology, cardiac-related enzymes (creatine kinase isoenzyme-MB (CK-MB) and lactate dehydrogenase (LDH)), and lipid profile markers (total cholesterol (TC), triglyceride (TG), and high-density lipoprotein (HDL)). The expression levels of key metabolic enzymes (glucose transporter-4 (GLUT4) and carnitine palmitoyltransferase-1B (CPT-1B)) were assessed. Our results displayed that co-treatment of E2 and/or 2ME with DOX significantly reduced DOX-induced cardiomyopathy and enhanced the metabolism of the heart through the maintenance of GLUT4 and CPT-1B enzymes. On the other hand, co-treatment of DOX with OVX, entacapone, or FULV increased the toxic effect of DOX by further reducing these important metabolic enzymes. E2 and 2ME abrogate DOX-induced cardiomyopathy partly through modulation of GLUT 4 and CPT-1B enzymes.
Collapse
Affiliation(s)
- Mohamed H Sobhy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
- Nanomedicine Research Labs, Center for Materials Science, Zewail City of Science and Technology, 6th of October City, Giza, Egypt
| | - Ahmed Ismail
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Mohammed S Abdel-Hamid
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| | - Mohamed Wagih
- Department of Pathology, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Marwa Kamel
- Department of Cancer Biology, Unit of Pharmacology and Experimental Therapeutics, National Cancer Institute, Cairo University, Cairo, Egypt.
| |
Collapse
|
25
|
Guo Q, Wang J, Ni C, Pan J, Zou J, Shi Y, Sun J, Zhang X, Wang D, Luan F. Research progress on the natural products in the intervention of myocardial infarction. Front Pharmacol 2024; 15:1445349. [PMID: 39239656 PMCID: PMC11374734 DOI: 10.3389/fphar.2024.1445349] [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: 06/07/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024] Open
Abstract
Coronary heart disease is a prevalent cardiovascular ailment globally, with myocardial infarction (MI) being one of its most severe manifestations. The morbidity and mortality of MI are escalating, showing an increasing trend among younger, highly educated individuals, thereby posing a serious threat to public health. Currently, thrombolysis, percutaneous coronary intervention, and coronary artery bypass grafting are the primary clinical treatments for MI. Although these methods significantly reduce patient mortality, complications often result in poor prognoses. Due to limitations in chemical synthetic drug research, the focus has shifted towards developing herbs based on natural substances. Natural medicines represent a novel approach for safer and more effective MI management and treatment. They can control multiple pathogenic variables by targeting various pathways and systems. This paper investigates the molecular mechanisms of MI and evaluates the application of natural products and medicinal plants in MI treatment over the past 5 years, demonstrating their specific good therapeutic potential and superior tolerance. These natural therapies have been shown to mitigate myocardial cell damage caused by MI through mechanisms such as oxidative stress, inflammation, apoptosis, angiogenesis, myocardial fibrosis, autophagy, endoplasmic reticulum stress, mitophagy, and pyroptosis. This review offers the latest insights into the application of natural products and medicinal plants in MI treatment, elucidating their mechanisms of action and serving as an important reference for MI prevention.
Collapse
Affiliation(s)
- Qiuting Guo
- College of Pharmacy, Xianyang Polytechnic Institute, Xianyang, China
| | - Jinhui Wang
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Caixia Ni
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Jiaojiao Pan
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Junbo Zou
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Yajun Shi
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Jing Sun
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Xiaofei Zhang
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Deng Wang
- Department of Pharmacy, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Fei Luan
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| |
Collapse
|
26
|
Wang M, Mo D, Zhang N, Yu H. Ferroptosis in diabetic cardiomyopathy: Advances in cardiac fibroblast-cardiomyocyte interactions. Heliyon 2024; 10:e35219. [PMID: 39165946 PMCID: PMC11334834 DOI: 10.1016/j.heliyon.2024.e35219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/19/2024] [Accepted: 07/24/2024] [Indexed: 08/22/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is a common complication of diabetes, and its pathogenesis remains elusive. Ferroptosis, a process dependent on iron-mediated cell death, plays a crucial role in DCM via disrupted iron metabolism, lipid peroxidation, and weakened antioxidant defenses. Hyperglycemia, oxidative stress, and inflammation may exacerbate ferroptosis in diabetes. This review emphasizes the interaction between cardiac fibroblasts and cardiomyocytes in DCM, influencing ferroptosis occurrence. By exploring ferroptosis modulation for potential therapeutic targets, this article offers a fresh perspective on DCM treatment. The study systematically covers the interplay, mechanisms, and targeted drugs linked to ferroptosis in DCM development.
Collapse
Affiliation(s)
| | | | - Ning Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Haichu Yu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| |
Collapse
|
27
|
Ibrahim Z, Khan NA, Siddiqui R, Qaisar R, Marzook H, Soares NC, Elmoselhi AB. Gut matters in microgravity: potential link of gut microbiota and its metabolites to cardiovascular and musculoskeletal well-being. Nutr Metab (Lond) 2024; 21:66. [PMID: 39123239 PMCID: PMC11316329 DOI: 10.1186/s12986-024-00836-6] [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: 02/08/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
The gut microbiota and its secreted metabolites play a significant role in cardiovascular and musculoskeletal health and diseases. The dysregulation of the intestinal microbiota poses a significant threat to cardiovascular and skeletal muscle well-being. Nonetheless, the precise molecular mechanisms underlying these changes remain unclear. Furthermore, microgravity presents several challenges to cardiovascular and musculoskeletal health compromising muscle strength, endothelial dysfunction, and metabolic changes. The purpose of this review is to critically examine the role of gut microbiota metabolites on cardiovascular and skeletal muscle functions and dysfunctions. It also explores the molecular mechanisms that drive microgravity-induced deconditioning in both cardiovascular and skeletal muscle. Key findings in this review highlight that several alterations in gut microbiota and secreted metabolites in microgravity mirror characteristics seen in cardiovascular and skeletal muscle diseases. Those alterations include increased levels of Firmicutes/Bacteroidetes (F/B) ratio, elevated lipopolysaccharide levels (LPS), increased in para-cresol (p-cresol) and secondary metabolites, along with reduction in bile acids and Akkermansia muciniphila bacteria. Highlighting the potential, modulating gut microbiota in microgravity conditions could play a significant role in mitigating cardiovascular and skeletal muscle diseases not only during space flight but also in prolonged bed rest scenarios here on Earth.
Collapse
Affiliation(s)
- Zeinab Ibrahim
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, 27272, UAE
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Naveed A Khan
- Microbiota Research Center, Istinye University, Istanbul, 34010, Turkey
| | - Ruqaiyyah Siddiqui
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh, EH14 4AS,, UK
- Microbiota Research Center, Istinye University, Istanbul, 34010, Turkey
| | - Rizwan Qaisar
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, 27272, UAE
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Hezlin Marzook
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, 27272, UAE
| | - Nelson C Soares
- Center for Applied and Translational Genomics (CATG), Mohammed Bin Rashid university of Medicine and Health Sciences, Dubai, 0000, United Arab Emirates
- Laboratory of Proteomics, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA), Av Padre Cruz, Lisbon, 1649-016, Portugal
| | - Adel B Elmoselhi
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, 27272, UAE.
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates.
| |
Collapse
|
28
|
Tanisha, Amudha C, Raake M, Samuel D, Aggarwal S, Bashir ZMD, Marole KK, Maryam I, Nazir Z. Diagnostic Modalities in Heart Failure: A Narrative Review. Cureus 2024; 16:e67432. [PMID: 39314559 PMCID: PMC11417415 DOI: 10.7759/cureus.67432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2024] [Indexed: 09/25/2024] Open
Abstract
Heart failure (HF) can present acutely or progress over time. It can lead to morbidity and mortality affecting 6.5 million Americans over the age of 20. The HF type is described according to the ejection fraction classification, defined as the percentage of blood volume that exits the left ventricle after myocardial contraction, undergoing ejection into the circulation, also called stroke volume, and is proportional to the ejection fraction. Cardiac catheterization is an invasive procedure to evaluate coronary artery disease leading to HF. Several biomarkers are being studied that could lead to early detection of HF and better symptom management. Testing for various biomarkers in the patient's blood is instrumental in confirming the diagnosis and elucidating the etiology of HF. There are various biomarkers elevated in response to increased myocardial stress and volume overload, including B-type natriuretic peptide (BNP) and its N-terminal prohormone BNP. We explored online libraries such as PubMed, Google Scholar, and Cochrane to find relevant articles. Our narrative review aims to extensively shed light on diagnostic modalities and novel techniques for diagnosing HF.
Collapse
Affiliation(s)
- Tanisha
- Department of Internal Medicine No. 4, O.O. Bogomolets National Medical University, Kyiv, UKR
| | - Chaithanya Amudha
- Department of Medicine and Surgery, Saveetha Medical College and Hospital, Chennai, IND
| | - Mohammed Raake
- Department of Surgery, Annamalai University, Chennai, IND
| | - Dany Samuel
- Department of Radiology, Medical University of Varna, Varna, BGR
| | | | - Zainab M Din Bashir
- Department of Medicine and Surgery, Combined Military Hospital (CMH) Lahore Medical College and Institute of Dentistry, Lahore, PAK
| | - Karabo K Marole
- Department of Medicine and Surgery, St. George's University School of Medicine, St. George's, GRD
| | - Iqra Maryam
- Department of Radiology, Allama Iqbal Medical College, Lahore, PAK
| | - Zahra Nazir
- Department of Internal Medicine, Combined Military Hospital, Quetta, PAK
| |
Collapse
|
29
|
Oguntade AS, Taylor H, Lacey B, Lewington S. Adiposity, fat-free mass and incident heart failure in 500 000 individuals. Open Heart 2024; 11:e002711. [PMID: 38964877 PMCID: PMC11227841 DOI: 10.1136/openhrt-2024-002711] [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: 04/14/2024] [Accepted: 06/20/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND AND AIMS The independent role of body fat distribution and fat-free mass in heart failure (HF) risk is unclear. We investigated the role of different body composition compartments in risk of HF. METHODS Present analyses include 428 087 participants (mean age 55.9 years, 44% male) from the UK Biobank. Associations of long-term average levels of body composition measures with incident HF were determined using adjusted Cox proportional hazards regression models. RESULTS Over a median follow-up of 13.8 years, there were 10 455 first-ever incident HF events. Overall, HF risk was more strongly associated with central adiposity (waist circumference (WC) adjusted for body mass index (BMI); HR 1.38, 95% CI 1.32 to 1.45) than general adiposity (BMI adjusted for WC; HR 1.22, 95% CI 1.16 to 1.27). Although dual X-ray absorptiometry-derived body fat remained positively related to HF after adjustment for fat-free mass (HR 1.37, 95% CI 1.18 to 1.59), the association of fat-free mass with HF was substantially attenuated by fat mass (HR 1.12, 95% CI 1.01 to 1.26) while visceral fat (VAT) remained associated with HF independent of subcutaneous fat (HR 1.20, 95% CI 1.09 to 1.33). In analyses of HF subtypes, HF with preserved ejection fraction was independently associated with all fat measures (eg, VAT: HR 1.23, 95% CI 1.12 to 1.35; body fat: HR 1.36, 95% CI 1.17 to 1.57) while HF with reduced ejection fraction was not independently associated with fat measures (eg, VAT: HR 1.29, 95% CI 0.98 to 1.68; body fat: HR 1.29, 95% CI 0.80 to 2.07). CONCLUSIONS This large-scale study shows that excess adiposity and fat mass are associated with higher HF risk while the association of fat-free mass with HF could be explained largely by its correlation with fat mass. The study also describes the independent relevance of body fat distribution to HF subtypes, suggesting different mechanisms may be driving their aetiopathogenesis.
Collapse
Affiliation(s)
- Ayodipupo S Oguntade
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Department of Cardiology, Hull University Teaching Hospitals NHS Trust, Hull, UK
| | - Hannah Taylor
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Ben Lacey
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- UK Biobank, Stockport, UK
| | - Sarah Lewington
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| |
Collapse
|
30
|
von Bibra C, Hinkel R. Non-human primate studies for cardiomyocyte transplantation-ready for translation? Front Pharmacol 2024; 15:1408679. [PMID: 38962314 PMCID: PMC11221829 DOI: 10.3389/fphar.2024.1408679] [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: 03/28/2024] [Accepted: 05/21/2024] [Indexed: 07/05/2024] Open
Abstract
Non-human primates (NHP) are valuable models for late translational pre-clinical studies, often seen as a last step before clinical application. The unique similarity between NHPs and humans is often the subject of ethical concerns. However, it is precisely this analogy in anatomy, physiology, and the immune system that narrows the translational gap to other animal models in the cardiovascular field. Cell and gene therapy approaches are two dominant strategies investigated in the research field of cardiac regeneration. Focusing on the cell therapy approach, several xeno- and allogeneic cell transplantation studies with a translational motivation have been realized in macaque species. This is based on the pressing need for novel therapeutic options for heart failure patients. Stem cell-based remuscularization of the injured heart can be achieved via direct injection of cardiomyocytes (CMs) or patch application. Both CM delivery approaches are in the late preclinical stage, and the first clinical trials have started. However, are we already ready for the clinical area? The present review concentrates on CM transplantation studies conducted in NHPs, discusses the main sources and discoveries, and provides a perspective about human translation.
Collapse
Affiliation(s)
- Constantin von Bibra
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behavior, Stiftung Tieraerztliche Hochschule Hannover, University of Veterinary Medicine, Hanover, Germany
- Laboratory Animal Science Unit, German Primate Center, Leibniz Institute for Primate Research, Goettingen, Germany
- DZHK (German Centre of Cardiovascular Research), Partner Site Lower Saxony, Goettingen, Germany
| | - Rabea Hinkel
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behavior, Stiftung Tieraerztliche Hochschule Hannover, University of Veterinary Medicine, Hanover, Germany
- Laboratory Animal Science Unit, German Primate Center, Leibniz Institute for Primate Research, Goettingen, Germany
- DZHK (German Centre of Cardiovascular Research), Partner Site Lower Saxony, Goettingen, Germany
| |
Collapse
|
31
|
Wang X, Zhao X, Wang X, Cao L, Lu B, Wang Z, Zhang W, Ti Y, Zhong M. Effect of levosimendan on ventricular remodelling in patients with left ventricular systolic dysfunction: a meta-analysis. ESC Heart Fail 2024; 11:1352-1376. [PMID: 38419326 PMCID: PMC11098670 DOI: 10.1002/ehf2.14714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/10/2024] [Accepted: 01/18/2024] [Indexed: 03/02/2024] Open
Abstract
Heart failure is the final stage of several cardiovascular diseases, and the key to effectively treating heart failure is to reverse or delay ventricular remodelling. Levosimendan is a novel inotropic and vasodilator agent used in heart failure, whereas the impact of levosimendan on ventricular remodelling is still unclear. This study aims to investigate the impact of levosimendan on ventricular remodelling in patients with left ventricular systolic dysfunction. Electronic databases were searched to identify eligible studies. A total of 66 randomized controlled trials involving 7968 patients were included. Meta-analysis results showed that levosimendan increased left ventricular ejection fraction [mean difference (MD) = 3.62, 95% confidence interval (CI) (2.88, 4.35), P < 0.00001] and stroke volume [MD = 6.59, 95% CI (3.22, 9.96), P = 0.0001] and significantly reduced left ventricular end-systolic volume [standard mean difference (SMD) = -0.52, 95% CI (-0.67, -0.37), P < 0.00001], left ventricular end-diastolic volume index [SMD = -1.24, 95% CI (-1.61, -0.86), P < 0.00001], and left ventricular end-systolic volume index [SMD = -1.06, 95% CI (-1.43, -0.70), P < 0.00001]. In terms of biomarkers, levosimendan significantly reduced the level of brain natriuretic peptide [SMD = -1.08, 95% CI (-1.60, -0.56), P < 0.0001], N-terminal pro-brain natriuretic peptide [SMD = -0.99, 95% CI (-1.41, -0.56), P < 0.00001], and interleukin-6 [SMD = -0.61, 95% CI (-0.86, -0.35), P < 0.00001]. Meanwhile, levosimendan may increase the incidence of hypotension [risk ratio (RR) = 1.24, 95% CI (1.12, 1.39), P < 0.0001], hypokalaemia [RR = 1.57, 95% CI (1.08, 2.28), P = 0.02], headache [RR = 1.89, 95% CI (1.50, 2.39), P < 0.00001], atrial fibrillation [RR = 1.31, 95% CI (1.12, 1.52), P = 0.0005], and premature ventricular complexes [RR = 1.86, 95% CI (1.27, 2.72), P = 0.001]. In addition, levosimendan reduced all-cause mortality [RR = 0.83, 95% CI (0.74, 0.94), P = 0.002]. In conclusion, our study found that levosimendan might reverse ventricular remodelling when applied in patients with left ventricular systolic dysfunction, especially in patients undergoing cardiac surgery, decompensated heart failure, and septic shock.
Collapse
Affiliation(s)
- Xia Wang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of CardiologyQilu Hospital of Shandong UniversityChina
| | - Xiu‐Zhi Zhao
- Department of CardiologyPeople's Hospital of Lixia District of JinanJinanShandongChina
| | - Xi‐Wen Wang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of CardiologyQilu Hospital of Shandong UniversityChina
| | - Lu‐Ying Cao
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of CardiologyQilu Hospital of Shandong UniversityChina
| | - Bin Lu
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of CardiologyQilu Hospital of Shandong UniversityChina
| | - Zhi‐Hao Wang
- Department of Geriatric MedicineShandong Key Laboratory of Cardiovascular Proteomics, Qilu Hospital, Cheeloo College of Medicine, Shandong UniversityJinanChina
| | - Wei Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of CardiologyQilu Hospital of Shandong UniversityChina
| | - Yun Ti
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of CardiologyQilu Hospital of Shandong UniversityChina
| | - Ming Zhong
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of CardiologyQilu Hospital of Shandong UniversityChina
| |
Collapse
|
32
|
Wu H, Jiang W, Pang P, Si W, Kong X, Zhang X, Xiong Y, Wang C, Zhang F, Song J, Yang Y, Zeng L, Liu K, Jia Y, Wang Z, Ju J, Diao H, Bian Y, Yang B. m 6A reader YTHDF1 promotes cardiac fibrosis by enhancing AXL translation. Front Med 2024; 18:499-515. [PMID: 38806989 DOI: 10.1007/s11684-023-1052-4] [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] [Received: 08/07/2023] [Accepted: 12/04/2023] [Indexed: 05/30/2024]
Abstract
Cardiac fibrosis caused by ventricular remodeling and dysfunction such as post-myocardial infarction (MI) can lead to heart failure. RNA N6-methyladenosine (m6A) methylation has been shown to play a pivotal role in the occurrence and development of many illnesses. In investigating the biological function of the m6A reader YTHDF1 in cardiac fibrosis, adeno-associated virus 9 was used to knock down or overexpress the YTHDF1 gene in mouse hearts, and MI surgery in vivo and transforming growth factor-β (TGF-β)-activated cardiac fibroblasts in vitro were performed to establish fibrosis models. Our results demonstrated that silencing YTHDF1 in mouse hearts can significantly restore impaired cardiac function and attenuate myocardial fibrosis, whereas YTHDF1 overexpression could further enhance cardiac dysfunction and aggravate the occurrence of ventricular pathological remodeling and fibrotic development. Mechanistically, zinc finger BED-type containing 6 mediated the transcriptional function of the YTHDF1 gene promoter. YTHDF1 augmented AXL translation and activated the TGF-β-Smad2/3 signaling pathway, thereby aggravating the occurrence and development of cardiac dysfunction and myocardial fibrosis. Consistently, our data indicated that YTHDF1 was involved in activation, proliferation, and migration to participate in cardiac fibrosis in vitro. Our results revealed that YTHDF1 could serve as a potential therapeutic target for myocardial fibrosis.
Collapse
Affiliation(s)
- Han Wu
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Weitao Jiang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Ping Pang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Wei Si
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xue Kong
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xinyue Zhang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yuting Xiong
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Chunlei Wang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Feng Zhang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Jinglun Song
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yang Yang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Linghua Zeng
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Kuiwu Liu
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yingqiong Jia
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Zhuo Wang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Jiaming Ju
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Hongtao Diao
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
| | - Yu Bian
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
| | - Baofeng Yang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
33
|
Aslan A, Ari Yuka S. Therapeutic peptides for coronary artery diseases: in silico methods and current perspectives. Amino Acids 2024; 56:37. [PMID: 38822212 PMCID: PMC11143054 DOI: 10.1007/s00726-024-03397-3] [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: 01/25/2024] [Accepted: 05/06/2024] [Indexed: 06/02/2024]
Abstract
Many drug formulations containing small active molecules are used for the treatment of coronary artery disease, which affects a significant part of the world's population. However, the inadequate profile of these molecules in terms of therapeutic efficacy has led to the therapeutic use of protein and peptide-based biomolecules with superior properties, such as target-specific affinity and low immunogenicity, in critical diseases. Protein‒protein interactions, as a consequence of advances in molecular techniques with strategies involving the combined use of in silico methods, have enabled the design of therapeutic peptides to reach an advanced dimension. In particular, with the advantages provided by protein/peptide structural modeling, molecular docking for the study of their interactions, molecular dynamics simulations for their interactions under physiological conditions and machine learning techniques that can work in combination with all these, significant progress has been made in approaches to developing therapeutic peptides that can modulate the development and progression of coronary artery diseases. In this scope, this review discusses in silico methods for the development of peptide therapeutics for the treatment of coronary artery disease and strategies for identifying the molecular mechanisms that can be modulated by these designs and provides a comprehensive perspective for future studies.
Collapse
Affiliation(s)
- Ayca Aslan
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, Esenler, Istanbul, Turkey
- Health Biotechnology Joint Research and Application Center of Excellence, Esenler, Istanbul, Turkey
| | - Selcen Ari Yuka
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, Esenler, Istanbul, Turkey.
- Health Biotechnology Joint Research and Application Center of Excellence, Esenler, Istanbul, Turkey.
| |
Collapse
|
34
|
Mohammadi K, Shafie D, Ghomashi N, Abdolizadeh A, Sadeghpour M. Kinin-kallikrein system: New perspectives in heart failure. Heart Fail Rev 2024; 29:729-737. [PMID: 38381277 DOI: 10.1007/s10741-024-10393-y] [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] [Accepted: 02/13/2024] [Indexed: 02/22/2024]
Abstract
Heart failure (HF) is a pervasive clinical challenge characterized by compromised cardiac function and reduced quality of life. The kinin-kallikrein system (KSS), a multifaceted peptide cascade, has garnered substantial attention due to its potential role in HF. Through activation of B1 and/or B2 receptors and downstream signaling, kinins modulate various physiological processes, including inflammation, coagulation, pain, blood pressure control, and vascular permeability. Notably, aberrations in KKS components have been linked to HF risk. The elevation of vasodilatory bradykinin (BK) due to kallikrein activity reduces preload and afterload, while concurrently fostering sodium reabsorption inhibition. However, kallikrein's conversion of prorenin to renin leads to angiotensinsII upregulation, resulting in vasoconstriction and fluid retention, alongside increased immune cell activity that fuels inflammation and cardiac remodeling. Importantly, prolonged KKS activation resulting from volume overload and tissue stretch contributes to cardiac collagen loss. The conventional renin-angiotensin-aldosterone system (RAAS) inhibitors used in HF management may inadvertently intensify KKS activity, exacerbating collagen depletion and cardiac remodeling. It is crucial to balance the KKS's role in acute cardiac damage, which may temporarily enhance function and metabolic parameters against its detrimental long-term effects. Thus, KKS blockade emerges as a promising strategy to impede HF progression. By attenuating the link between immune system function and tissue damage, KKS inhibition can potentially reduce cardiac remodeling and alleviate HF symptoms. However, the nuanced roles of BK in various acute conditions necessitate further investigation into the sustained benefits of kallikrein inhibitors in patients with chronic HF.
Collapse
Affiliation(s)
- Keivan Mohammadi
- Shahid Chamran Heart Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Davood Shafie
- Heart Failure Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Newsha Ghomashi
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Abdolizadeh
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Majid Sadeghpour
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
35
|
Lin K, Shi D, Wang A, Ge J, Cheng D, Yan Y. Noninvasive Monitoring of Early Cardiac Fibrosis in Diabetic Mice by [ 68Ga]Ga-DOTA-FAPI-04 PET/CT Imaging. ACS OMEGA 2024; 9:17195-17203. [PMID: 38645332 PMCID: PMC11024947 DOI: 10.1021/acsomega.3c10061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 04/23/2024]
Abstract
Cardiac fibrosis represents one of the representative pathological characteristics in the diabetic heart. Active fibroblasts play an essential role in the progression of cardiac fibrosis. The technologies for noninvasive monitoring of activated fibroblasts still have to be investigated. The purpose of this study was to evaluate the feasibility of targeted fibroblast activation protein (FAP) molecular imaging in the early evaluation of diabetic cardiac fibrosis using [68Ga]Ga-DOTA-FAPI-04 PET/CT. PET/CT imaging was conducted in db/db mice and db/m mice at weeks 12 and 24. Diabetic heart injury was determined using echocardiography and serum biomarkers. Additionally, the levels of cardiac fibrosis were also assessed. In our study, conventional diagnostic modalities, including echocardiography and serum biomarkers, failed to monitor early-stage cardiac dysfunction and fibrosis in diabetic mice. Conversely, the results of [68Ga]Ga-DOTA-FAPI-04 PET/CT imaging demonstrated that diabetic mice had increased myocardial uptake of radioactive tracers in both early-stage and late-stage diabetes, consistent with the elevated FAP expression and increased cardiac fibrosis level. Notably, cardiac PET signals exhibited significant correlations with left ventricular ejection fractions, the E/A ratio, and the level of serum TGF-β1, PIIINP, and sST2. The results demonstrated the potential of [68Ga]Ga-DOTA-FAPI-04 PET/CT imaging for visualizing activated fibroblasts and detecting early-stage diabetic heart injury and fibrosis noninvasively. They also demonstrated the clinical superiority of [68Ga]Ga-DOTA-FAPI-04 PET/CT imaging over echocardiography and serum biomarkers in the early monitoring of diabetes-related cardiac dysfunction and fibrosis.
Collapse
Affiliation(s)
- Kaibin Lin
- Department
of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Dai Shi
- Department
of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai
Institute of Medical Imaging, Shanghai 200032, China
- Institute
of Nuclear Medicine, Fudan University, Shanghai 200032, China
- Cancer
Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ai Wang
- Department
of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Junbo Ge
- Department
of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Dengfeng Cheng
- Department
of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai
Institute of Medical Imaging, Shanghai 200032, China
- Institute
of Nuclear Medicine, Fudan University, Shanghai 200032, China
- Cancer
Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yan Yan
- Department
of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
36
|
Menezes CADS, de Oliveira ALG, Barbosa ICM, de Jesus ACP, Chaves AT, Rocha MODC. Galectin-3 (Gal-3) and the tissue inhibitor of matrix metalloproteinase (TIMP-2) as potential biomarkers for the clinical evolution of chronic Chagas cardiomyopathy. Acta Trop 2024; 252:107153. [PMID: 38373528 DOI: 10.1016/j.actatropica.2024.107153] [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: 12/23/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 02/21/2024]
Abstract
BACKGROUND Chronic Chagas cardiomyopathy (CCC) is responsible for the highest morbidity and worst prognosis in Chagas disease patients. However, predicting factors that correlate with disease progression, morbidity, and mortality is challenging. It is necessary to have simple, quantitative, and economical risk biomarkers that add value to conventional methods and assist in the diagnosis and prognosis of patients with CCC or in evolution. OBJECTIVES We evaluated molecules related to cardiac remodeling and fibrosis, such as MMP-2, MMP-9, TIMP-2, TIMP-1, PICP, CTXI, and Gal-3, and correlated these biomarkers with echocardiographic variables (LVDD, LVEF, and E/e' ratio). METHODS Blood samples from Chagasic patients without apparent cardiopathy (WAC), CCC patients, and healthy individuals were used to perform plasma molecule dosages using Luminex or ELISA. RESULTS MMP-2 and TIMP-2 presented higher levels in CCC; in these patients, the inhibitory role of TIMP-2 over MMP-2 was reinforced. The ratio of MMP-2/TIMP-2 in WAC patients showed a bias in favor of the gelatinase pathway. MMP-9 and TIMP-1 showed higher levels in Chagas patients compared to healthy subjects. PICP and CTXI are not associated with cardiac deterioration in Chagas disease. Increased levels of Gal-3 are associated with worse cardiac function in CCC. Receiver operating characteristic (ROC) curve analysis identified Gal-3 and TIMP-2 as putative biomarkers to discriminate WAC from cardiac patients. CONCLUSIONS Among the molecules evaluated, Gal-3 and TIMP-2 have the potential to be used as biomarkers of cardiac remodeling and progressive myocardial fibrosis in Chagas disease.
Collapse
Affiliation(s)
- Cristiane Alves da Silva Menezes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte, Minas Gerais CEP 31270-901, Brazil.
| | - Ana Laura Grossi de Oliveira
- Faculdade de Medicina, Programa de Pós-graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Isabela Cristina Magalhães Barbosa
- Faculdade de Medicina, Programa de Pós-graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Augusto César Parreiras de Jesus
- Faculdade de Medicina, Programa de Pós-graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Ana Thereza Chaves
- Faculdade de Medicina, Programa de Pós-graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Manoel Otávio da Costa Rocha
- Faculdade de Medicina, Programa de Pós-graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
37
|
Johri N, Matreja PS, Agarwal S, Nagar P, Kumar D, Maurya A. Unraveling the Molecular Mechanisms of Activated Protein C (APC) in Mitigating Reperfusion Injury and Cardiac Ischemia: a Promising Avenue for Novel Therapeutic Interventions. J Cardiovasc Transl Res 2024; 17:345-355. [PMID: 37851312 DOI: 10.1007/s12265-023-10445-y] [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: 06/04/2023] [Accepted: 10/02/2023] [Indexed: 10/19/2023]
Abstract
Ischemic heart disease, which results from plaque formation in the coronary arteries, hinders the flow of oxygenated blood to the heart, leading to ischemia. Reperfusion injury remains a significant challenge for researchers, and the mechanisms underlying myocardial ischemia-reperfusion injury (MIRI) are not entirely understood. The review directs future research into potential targets in clinical treatment based on our present understanding of the pathophysiological mechanisms of MIRI. The study provides insights into the mechanisms underlying MIRI and offers direction for future research in this area. The use of targeted therapies may hold promise in improving cardiac function in the elderly and minimizing the adverse effects of revascularization therapies. The purpose of this review is to analyze the role of activated protein C (APC) in the pathogenesis of ischemic heart disease, heart failure, and myocardial ischemia-reperfusion injury, and discuss the potential of APC-based therapeutics.
Collapse
Affiliation(s)
- Nishant Johri
- Department of Pharmacy Practice & Pharmacology, Teerthanker Mahaveer College of Pharmacy, Moradabad, Uttar Pradesh, India.
- School of Health & Psychological Sciences, City, University of London, London, United Kingdom.
| | - Prithpal S Matreja
- Department of Pharmacology, Teerthanker Mahaveer Medical College and Research Centre, Moradabad, Uttar Pradesh, India
| | - Shalabh Agarwal
- Department of Cardiology, Teerthanker Mahaveer Hospital & Research Centre, Moradabad, Uttar Pradesh, India
| | - Priya Nagar
- Department of Pharmacy Practice & Pharmacology, Teerthanker Mahaveer College of Pharmacy, Moradabad, Uttar Pradesh, India
| | - Deepanshu Kumar
- Department of Pharmacy Practice & Pharmacology, Teerthanker Mahaveer College of Pharmacy, Moradabad, Uttar Pradesh, India
| | - Aditya Maurya
- Department of Pharmacy Practice & Pharmacology, Teerthanker Mahaveer College of Pharmacy, Moradabad, Uttar Pradesh, India
| |
Collapse
|
38
|
Ricciotti E, Haines PG, Chai W, FitzGerald GA. Prostanoids in Cardiac and Vascular Remodeling. Arterioscler Thromb Vasc Biol 2024; 44:558-583. [PMID: 38269585 PMCID: PMC10922399 DOI: 10.1161/atvbaha.123.320045] [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: 08/22/2023] [Accepted: 01/09/2024] [Indexed: 01/26/2024]
Abstract
Prostanoids are biologically active lipids generated from arachidonic acid by the action of the COX (cyclooxygenase) isozymes. NSAIDs, which reduce the biosynthesis of prostanoids by inhibiting COX activity, are effective anti-inflammatory, antipyretic, and analgesic drugs. However, their use is limited by cardiovascular adverse effects, including myocardial infarction, stroke, hypertension, and heart failure. While it is well established that NSAIDs increase the risk of atherothrombotic events and hypertension by suppressing vasoprotective prostanoids, less is known about the link between NSAIDs and heart failure risk. Current evidence indicates that NSAIDs may increase the risk for heart failure by promoting adverse myocardial and vascular remodeling. Indeed, prostanoids play an important role in modulating structural and functional changes occurring in the myocardium and in the vasculature in response to physiological and pathological stimuli. This review will summarize current knowledge of the role of the different prostanoids in myocardial and vascular remodeling and explore how maladaptive remodeling can be counteracted by targeting specific prostanoids.
Collapse
Affiliation(s)
- Emanuela Ricciotti
- Department of Systems Pharmacology and Translational Therapeutics (E.R., G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
- Institute for Translational Medicine and Therapeutics (E.R., G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Philip G Haines
- Rhode Island Hospital, Department of Medicine, Warren Alpert Medical School of Brown University, Providence (P.G.H.)
| | - William Chai
- Health and Human Biology, Division of Biology and Medicine, Brown University, Providence, RI (W.C.)
| | - Garret A FitzGerald
- Department of Systems Pharmacology and Translational Therapeutics (E.R., G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
- Institute for Translational Medicine and Therapeutics (E.R., G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
- Department of Medicine (G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
| |
Collapse
|
39
|
Dias LG, Reis CHO, Dos Santos L, Krause Neto W, Lima-Leopoldo AP, Baker JS, Leopoldo AS, Bocalini DS. Strength training improves heart function, collagen and strength in rats with heart failure. J Physiol Sci 2024; 74:10. [PMID: 38365576 PMCID: PMC10873996 DOI: 10.1186/s12576-024-00899-3] [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: 10/28/2023] [Accepted: 01/20/2024] [Indexed: 02/18/2024]
Abstract
BACKGROUND/OBJECTIVES Myocardial infarction (MI) frequently leads to cardiac remodeling and failure with impaired life quality, playing an important role in cardiovascular deaths. Although physical exercise is a well-recognized effective non-pharmacological therapy for cardiovascular diseases, the effects of strength training (ST) on the structural and functional aspects of cardiac remodeling need to be further documented. In this study, we aimed to investigate the role of a linear block ST protocol in the rat model of MI. METHODS AND RESULTS After 6 weeks of MI induction or sham surgery, male adult rats performed ST for the following 12 weeks. The ladder-based ST program was organized in three mesocycles of 4 weeks, with one load increment for each block according to the maximal carrying load test. After 12 weeks, the infarcted-trained rats exhibited an increase in performance, associated with reduced cardiac hypertrophy and pulmonary congestion compared with the untrained group. Despite not changing MI size, the ST program partially prevented cardiac dilatation and ventricular dysfunction assessed by echocardiography and hemodynamics, and interstitial fibrosis evaluated by histology. In addition, isolated cardiac muscles from infarcted-trained rats had improved contractility parameters in a steady state, and in response to calcium or stimuli pauses. CONCLUSIONS The ST in infarcted rats increased the capacity to carry mass, associated with attenuation of cardiac remodeling and pulmonary congestion with improving cardiac function that could be attributed, at least in part, to the improvement of myocardial contractility.
Collapse
Affiliation(s)
- Leisiane G Dias
- Experimental Physiology and Biochemistry Laboratory. Physical Education and Sport Center, Federal University of Espirito Santo, Vitoria, Brazil
| | - Carlos H O Reis
- Experimental Physiology and Biochemistry Laboratory. Physical Education and Sport Center, Federal University of Espirito Santo, Vitoria, Brazil
| | - Leonardo Dos Santos
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espirito Santo, Vitoria, Brazil
| | - Walter Krause Neto
- Department of Morphology and Genetics, Federal University of São Paulo, São Paulo, Brazil
| | - Ana Paula Lima-Leopoldo
- Experimental Physiology and Biochemistry Laboratory. Physical Education and Sport Center, Federal University of Espirito Santo, Vitoria, Brazil
| | - Julien S Baker
- Department of Sport, Physical Education and Health, Centre for Health and Exercise Science Research, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - André S Leopoldo
- Experimental Physiology and Biochemistry Laboratory. Physical Education and Sport Center, Federal University of Espirito Santo, Vitoria, Brazil
| | - Danilo S Bocalini
- Experimental Physiology and Biochemistry Laboratory. Physical Education and Sport Center, Federal University of Espirito Santo, Vitoria, Brazil.
| |
Collapse
|
40
|
Guimarães LC, Fidale TM, Pereira TCR, Lopes PR, Ferreira-Junior MD, Deconte SR, Ferreira-Neto ML, Brito WS, Gomes RM, de Souza FR, Cavalcante KVN, Herrera GC, de Moura FBR, Resende ES. Cardioprotective Effects of Leucine Supplementation against Doxorubicin-Induced Cardiotoxicit. Cardiovasc Toxicol 2024; 24:122-132. [PMID: 38165500 DOI: 10.1007/s12012-023-09817-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/18/2023] [Indexed: 01/03/2024]
Abstract
Doxorubicin is one of the most important antitumor drugs used in oncology; however, its cardiotoxic effect limits the therapeutic use and raises concerns regarding patient prognosis. Leucine is a branched-chain amino acid used in dietary supplementation and has been studied to attenuate the toxic effects of doxorubicin in animals, which increases oxidative stress. Oxidative stress in different organs can be estimated using several methods, including catalase expression analysis. This study aimed to analyze the effect of leucine on catalase levels in rat hearts after doxorubicin administration. Adult male Wistar rats were separated into two groups: Standard diet (SD) and 5% Leucine-Enriched Diet (LED). The animals had free access to diet from D0 to D28. At D14, the groups were subdivided in animals injected with Doxorubicin and animals injected with vehicle, until D28, and the groups were SD, SD + Dox, LED and LED + Dox. At D28, the animals were submitted do Transthoracic Echocardiography and euthanized. Despite Dox groups had impaired body weight gain, raw heart weight was not different between the groups. No substantial alterations were observed in macroscopic evaluation of the heart. Although, Doxorubicin treatment increased total interstitial collagen in the heart, which in addition to Type I collagen, is lower in LED groups. Western blot analysis showed that catalase expression in the heart of LED groups was lower than that in SD groups. In conclusion, leucine supplementation reduced both the precocious Dox-induced cardiac remodeling and catalase levels in the heart.
Collapse
Affiliation(s)
- Lucas C Guimarães
- Laboratory of Experimental Medicine, Department of Health Sciences - PGCS, Faculty of Medicine, Federal University of Uberlândia, Uberlândia, Brazil.
| | - Thiago M Fidale
- Biotechnology Institute. Department of Medicine, Federal University of Catalão, Catalão, Goiás, Brazil
| | - Talita C R Pereira
- Institute of Biomedical Sciences, Department of Physiology, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Paulo R Lopes
- School of Dentistry-FOAr, Department of Physiology and Pathology, Universidade Estadual Paulista "Júlio de Mesquita Filho"-UNESP, Araraquara, SP, Brazil
| | - Marcos D Ferreira-Junior
- Laboratory of Endocrine Physiology and Metabolism, ICB, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Simone R Deconte
- UFU-ICBIM. Department of Physiology and Biophysics, Federal University of Uberlândia, Uberlândia, Brazil
| | - Marcos L Ferreira-Neto
- UFU-ICBIM. Department of Physiology and Biophysics, Federal University of Uberlândia, Uberlândia, Brazil
| | | | - Rodrigo M Gomes
- Laboratory of Endocrine Physiology and Metabolism, ICB, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Fernanda R de Souza
- Laboratory of Experimental Medicine, Department of Medicine, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Keilah V N Cavalcante
- Laboratory of Endocrine Physiology and Metabolism, ICB, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Gustavo C Herrera
- The Veterinary Hospital, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | | | - Elmiro S Resende
- Laboratory of Experimental Medicine, Department of Health Sciences - PGCS, Faculty of Medicine, Federal University of Uberlândia, Uberlândia, Brazil
- Postgraduate Program in Health Sciences-PGCS, Faculty of Medicine, Federal University of Uberlândia, Uberlândia, MG, Brazil
| |
Collapse
|
41
|
Yang D, Wan X, Schwieterman N, Cavus O, Kacira E, Xu X, Laurita KR, Wold LE, Hund TJ, Mohler PJ, Deschênes I, Fu JD. MicroRNA-1 Deficiency Is a Primary Etiological Factor Disrupting Cardiac Contractility and Electrophysiological Homeostasis. Circ Arrhythm Electrophysiol 2024; 17:e012150. [PMID: 38126205 PMCID: PMC10842700 DOI: 10.1161/circep.123.012150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND MicroRNA-1 (miR1), encoded by the genes miR1-1 and miR1-2, is the most abundant microRNA in the heart and plays a critical role in heart development and physiology. Dysregulation of miR1 has been associated with various heart diseases, where a significant reduction (>75%) in miR1 expression has been observed in patient hearts with atrial fibrillation or acute myocardial infarction. However, it remains uncertain whether miR1-deficiency acts as a primary etiological factor of cardiac remodeling. METHODS miR1-1 or miR1-2 knockout mice were crossbred to produce 75%-miR1-knockdown (75%KD; miR1-1+/-:miR1-2-/- or miR1-1-/-:miR1-2+/-) mice. Cardiac pathology of 75%KD cardiomyocytes/hearts was investigated by ECG, patch clamping, optical mapping, transcriptomic, and proteomic assays. RESULTS In adult 75%KD hearts, the overall miR1 expression was reduced to ≈25% of the normal wild-type level. These adult 75%KD hearts displayed decreased ejection fraction and fractional shortening, prolonged QRS and QT intervals, and high susceptibility to arrhythmias. Adult 75%KD cardiomyocytes exhibited prolonged action potentials with impaired repolarization and excitation-contraction coupling. Comparatively, 75%KD cardiomyocytes showcased reduced Na+ current and transient outward potassium current, coupled with elevated L-type Ca2+ current, as opposed to wild-type cells. RNA sequencing and proteomics assays indicated negative regulation of cardiac muscle contraction and ion channel activities, along with a positive enrichment of smooth muscle contraction genes in 75%KD cardiomyocytes/hearts. miR1 deficiency led to dysregulation of a wide gene network, with miR1's RNA interference-direct targets influencing many indirectly regulated genes. Furthermore, after 6 weeks of bi-weekly intravenous tail-vein injection of miR1 mimics, the ejection fraction and fractional shortening of 75%KD hearts showed significant improvement but remained susceptible to arrhythmias. CONCLUSIONS miR1 deficiency acts as a primary etiological factor in inducing cardiac remodeling via disrupting heart regulatory homeostasis. Achieving stable and appropriate microRNA expression levels in the heart is critical for effective microRNA-based therapy in cardiovascular diseases.
Collapse
Affiliation(s)
- Dandan Yang
- The Dorothy M. Davis Heart and Lung Research Institute, Frick Center for Heart Failure and Arrhythmia, Dept of Physiology and Cell Biology, The Ohio State University, Columbus
| | - Xiaoping Wan
- The Dorothy M. Davis Heart and Lung Research Institute, Frick Center for Heart Failure and Arrhythmia, Dept of Physiology and Cell Biology, The Ohio State University, Columbus
| | - Neill Schwieterman
- The Dorothy M. Davis Heart and Lung Research Institute, Dept of Surgery, Division of Cardiac Surgery, The Ohio State University, Columbus
| | - Omer Cavus
- The Dorothy M. Davis Heart and Lung Research Institute, Frick Center for Heart Failure and Arrhythmia, Dept of Physiology and Cell Biology, The Ohio State University, Columbus
- Pennsylvania State University, Heart and Vascular Institute, Hershey, PA
| | - Ege Kacira
- The Dorothy M. Davis Heart and Lung Research Institute, Frick Center for Heart Failure and Arrhythmia, Dept of Physiology and Cell Biology, The Ohio State University, Columbus
| | - Xianyao Xu
- The Dorothy M. Davis Heart and Lung Research Institute, Frick Center for Heart Failure and Arrhythmia, Depts of Internal Medicine & Biomedical Engineering, The Ohio State University, Columbus
| | - Kenneth R. Laurita
- Dept of Medicine, Heart and Vascular Research Center, The MetroHealth System, Case Western Reserve University, Cleveland, OH
| | - Loren E. Wold
- The Dorothy M. Davis Heart and Lung Research Institute, Dept of Surgery, Division of Cardiac Surgery, The Ohio State University, Columbus
| | - Thomas J. Hund
- The Dorothy M. Davis Heart and Lung Research Institute, Frick Center for Heart Failure and Arrhythmia, Depts of Internal Medicine & Biomedical Engineering, The Ohio State University, Columbus
| | - Peter J. Mohler
- The Dorothy M. Davis Heart and Lung Research Institute, Frick Center for Heart Failure and Arrhythmia, Dept of Physiology and Cell Biology, The Ohio State University, Columbus
| | - Isabelle Deschênes
- The Dorothy M. Davis Heart and Lung Research Institute, Frick Center for Heart Failure and Arrhythmia, Dept of Physiology and Cell Biology, The Ohio State University, Columbus
| | - Ji-Dong Fu
- The Dorothy M. Davis Heart and Lung Research Institute, Frick Center for Heart Failure and Arrhythmia, Dept of Physiology and Cell Biology, The Ohio State University, Columbus
| |
Collapse
|
42
|
Momeni Z, Danesh S, Ahmadpour M, Eshraghi R, Farkhondeh T, Pourhanifeh MH, Samarghandian S. Protective Roles and Therapeutic Effects of Gallic Acid in the Treatment of Cardiovascular Diseases: Current Trends and Future Directions. Curr Med Chem 2024; 31:3733-3751. [PMID: 37815180 DOI: 10.2174/0109298673259299230921150030] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/19/2023] [Accepted: 08/18/2023] [Indexed: 10/11/2023]
Abstract
Cardiovascular diseases (CVDs) are serious life-threatening illnesses and significant problematic issues for public health having a heavy economic burden on all society worldwide. The high incidence of these diseases as well as high mortality rates make them the leading causes of death and disability. Therefore, finding novel and more effective therapeutic methods is urgently required. Gallic acid, an herbal medicine with numerous biological properties, has been utilized in the treatment of various diseases for thousands of years. It has been demonstrated that gallic acid possesses pharmacological potential in regulating several molecular and cellular processes such as apoptosis and autophagy. Moreover, gallic acid has been investigated in the treatment of CVDs both in vivo and in vitro. Herein, we aimed to review the available evidence on the therapeutic application of gallic acid for CVDs including myocardial ischemia-reperfusion injury and infarction, drug-induced cardiotoxicity, hypertension, cardiac fibrosis, and heart failure, with a focus on underlying mechanisms.
Collapse
Affiliation(s)
- Zahra Momeni
- Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Sepideh Danesh
- Research Hub Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Ahmadpour
- Research Hub Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Eshraghi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Tahereh Farkhondeh
- Department of Toxicology and Pharmacology, School of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Hub Institute, Tehran University of Medical Sciences, Tehran, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Saeed Samarghandian
- University of Neyshabur Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
43
|
Nanda D, Pant P, Machha P, Sowpati DT, Kumarswamy R. Transcriptional changes during isoproterenol-induced cardiac fibrosis in mice. Front Mol Biosci 2023; 10:1263913. [PMID: 38178867 PMCID: PMC10765171 DOI: 10.3389/fmolb.2023.1263913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 10/23/2023] [Indexed: 01/06/2024] Open
Abstract
Introduction: β-adrenergic stimulation using β-agonists such as isoproterenol has been routinely used to induce cardiac fibrosis in experimental animal models. Although transcriptome changes in surgical models of cardiac fibrosis such as transverse aortic constriction (TAC) and coronary artery ligation (CAL) are well-studied, transcriptional changes during isoproterenol-induced cardiac fibrosis are not well-explored. Methods: Cardiac fibrosis was induced in male C57BL6 mice by administration of isoproterenol for 4, 8, or 11 days at 50 mg/kg/day dose. Temporal changes in gene expression were studied by RNA sequencing. Results and discussion: We observed a significant alteration in the transcriptome profile across the different experimental groups compared to the saline group. Isoproterenol treatment caused upregulation of genes associated with ECM organization, cell-cell contact, three-dimensional structure, and cell growth, while genes associated with fatty acid oxidation, sarcoplasmic reticulum calcium ion transport, and cardiac muscle contraction are downregulated. A number of known long non-coding RNAs (lncRNAs) and putative novel lncRNAs exhibited differential regulation. In conclusion, our study shows that isoproterenol administration leads to the dysregulation of genes relevant to ECM deposition and cardiac contraction, and serves as an excellent alternate model to the surgical models of heart failure.
Collapse
Affiliation(s)
- Disha Nanda
- Council of Scientific and Industrial Research (CSIR)–Centre for Cellular and Molecular Biology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Priyanka Pant
- Council of Scientific and Industrial Research (CSIR)–Centre for Cellular and Molecular Biology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Pratheusa Machha
- Council of Scientific and Industrial Research (CSIR)–Centre for Cellular and Molecular Biology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Divya Tej Sowpati
- Council of Scientific and Industrial Research (CSIR)–Centre for Cellular and Molecular Biology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Regalla Kumarswamy
- Council of Scientific and Industrial Research (CSIR)–Centre for Cellular and Molecular Biology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
44
|
Liang B, Zhang XX, Gu N. Guanxin V Relieves Ventricular Remodeling by Inhibiting Inflammation: Implication from Virtual Screening, Systematic Pharmacology, Molecular Docking, and Experimental Validation. Chin J Integr Med 2023; 29:1077-1086. [PMID: 37528325 DOI: 10.1007/s11655-023-3642-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2023] [Indexed: 08/03/2023]
Abstract
OBJECTIVE To reveal the anti-inflammatory mechanism of Guanxin V, which is prescribed for ventricular remodeling in clinical practice. METHODS Guanxin V-, ventricular remodeling-, and inflammation-related targets were obtained through an integrated strategy of virtual screening and systematic pharmacology, and then the shared targets were visualised with a Venn diagram. Guanxin V network and the protein-protein interaction network were drawn, and enrichment analysis was conducted. Finally, the main results obtained from the integrated strategy were validated by molecular docking and in vivo experiments. RESULTS A total of 251, 11,425, and 15,246 Guanxin V-, ventricular remodeling-, and inflammation-related targets were acquired, respectively. Then, 211 shared targets were considered to contribute to the mechanism of ventricular remodeling treated by Guanxin V. Guanxin network and the protein-protein interaction network were drawn, and enrichment analysis showed some cardiovascular-related biological processes and signaling pathways. Molecular docking revealed that the Guanxin V-derived compounds could align with key targets. Final in vivo experiments proved that Guanxin V reverses ventricular remodeling by inhibiting inflammation. CONCLUSION Guanxin V relieves ventricular remodeling by regulating inflammation, which provides new ideas for the anti-ventricular remodeling mechanism of Guanxin V.
Collapse
Affiliation(s)
- Bo Liang
- Department of Cardiology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, China
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Xiao-Xiao Zhang
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, China
| | - Ning Gu
- Department of Cardiology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, China.
| |
Collapse
|
45
|
Batista ANR, Garcia T, Prudente R, Barbosa MF, Modesto P, Franco E, de Godoy I, Paiva S, Azevedo P, Tanni SE. Cardiac function, myocardial fat deposition, and lipid profile in young smokers: a cross-sectional study. Front Cardiovasc Med 2023; 10:1225621. [PMID: 38034384 PMCID: PMC10682099 DOI: 10.3389/fcvm.2023.1225621] [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: 06/12/2023] [Accepted: 10/26/2023] [Indexed: 12/02/2023] Open
Abstract
Background There is a possibility that cardiac morphometric characteristics are associated with the lipid profile, that is, the composition and concentration of triglycerides, total cholesterol, HDL, LDL, and others lipoproteins in young smokers without comorbidities. Thus, this study aimed to evaluate the association of cardiac morphometric characteristics, myocardial fat deposition, and smoking cessation with the lipid profile of young smokers. Methods A clinical and laboratory evaluation of lipids and the smoking status was performed on 57 individuals, including both a smoker group and a control group. Cardiac magnetic resonance imaging (MRI) with proton spectroscopy was performed to identify cardiac changes and triglyceride (TG) deposition in myocardial tissue. Results No differences were observed between the groups (control vs. smokers) in relation to the amount of myocardial TG deposition (p = 0.47); however, when TG deposition was correlated with cardiac MRI variables, a positive correlation was identified between smoking history and myocardial TG deposition [hazard ratio (95% CI), 0.07 (0.03-0.12); p = 0.002]. Furthermore, it was observed that the smoking group had lower high-density lipoprotein cholesterol [51 (45.5-59.5) mg/dl vs. 43 (36-49.5) mg/dl, p = 0.003] and higher TG [73 (58-110) mg/dl vs. 122 (73.5-133) mg/dl, p = 0.01] and very-low-density lipoprotein cholesterol [14.6 (11.6-22.2) mg/dl vs. 24.4 (14.7-26.6) mg/dl, p = 0.01] values. In the control and smoking groups, a negative correlation between TGs and the diameter of the aortic root lumen and positive correlation with the thickness of the interventricular septum and end-diastolic volume (EDV) of both the right ventricle (RV) and left ventricle (LV) were noted. Moreover, in the RV, positive correlations with the end-systolic volume (ESV) index (ESVI), stroke volume (SV), ESV, and EDV were observed. Regarding serum free fatty acids, we found a negative correlation between their values and the diameter of the lumen of the ascending aortic vessel. Lipoprotein lipase showed a positive correlation with the SV index of the RV and negative correlation with the diameter of the lumen of the ascending aortic vessel. Conclusion Several associations were observed regarding cardiac morphometric characteristics, myocardial fat deposition, and smoking cessation with the lipid profile of young smokers.
Collapse
Affiliation(s)
- Ana Natália Ribeiro Batista
- Pneumology Area, Department of Internal Medicine, Botucatu School of Medicine, São Paulo State University (UNESP), Botucatu, Brazil
| | - Thaís Garcia
- Pneumology Area, Department of Internal Medicine, Botucatu School of Medicine, São Paulo State University (UNESP), Botucatu, Brazil
| | - Robson Prudente
- Pulmonary Function Laboratory, Clinical Hospital of Botucatu Medical School, São Paulo State University (UNESP), Botucatu, Brazil
| | - Maurício F. Barbosa
- Department of Tropical Diseases and Diagnostic Imaging, Botucatu School of Medicine, São Paulo State University (UNESP), Botucatu, Brazil
| | - Pamela Modesto
- Pneumology Area, Department of Internal Medicine, Botucatu School of Medicine, São Paulo State University (UNESP), Botucatu, Brazil
| | - Estefânia Franco
- Pulmonary Function Laboratory, Clinical Hospital of Botucatu Medical School, São Paulo State University (UNESP), Botucatu, Brazil
| | - Irma de Godoy
- Pneumology Area, Department of Internal Medicine, Botucatu School of Medicine, São Paulo State University (UNESP), Botucatu, Brazil
| | - Sergio Paiva
- Pneumology Area, Department of Internal Medicine, Botucatu School of Medicine, São Paulo State University (UNESP), Botucatu, Brazil
| | - Paula Azevedo
- Pneumology Area, Department of Internal Medicine, Botucatu School of Medicine, São Paulo State University (UNESP), Botucatu, Brazil
| | - Suzana Erico Tanni
- Pneumology Area, Department of Internal Medicine, Botucatu School of Medicine, São Paulo State University (UNESP), Botucatu, Brazil
| |
Collapse
|
46
|
Pipilas DC, Hanley A, Singh JP, Mela T. Cardiac Contractility Modulation for Heart Failure: Current and Future Directions. JOURNAL OF THE SOCIETY FOR CARDIOVASCULAR ANGIOGRAPHY & INTERVENTIONS 2023; 2:101176. [PMID: 39131075 PMCID: PMC11307863 DOI: 10.1016/j.jscai.2023.101176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/30/2023] [Accepted: 09/06/2023] [Indexed: 08/13/2024]
Abstract
Cardiac contractility modulation (CCM) is a Food and Drug Administration-approved device-based therapy for patients with heart failure. The system delivers biphasic electric stimulation to the ventricular myocardium during the absolute refractory period to augment left ventricular contraction. CCM therapy promotes acute and chronic changes at the cellular level, leading to favorable remodeling throughout the myocardium. CCM improves quality of life, New York Heart Association class, left ventricular ejection fraction, peak oxygen uptake, and the composite end point of cardiovascular death and heart failure hospitalizations. This review will focus on the biological basis, indications, and evidence for CCM, as well as the future applications of this technology.
Collapse
Affiliation(s)
- Daniel C. Pipilas
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Alan Hanley
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, Massachusetts
| | - Jagmeet P. Singh
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, Massachusetts
| | - Theofanie Mela
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
47
|
Gaytan SL, Beaven E, Gadad SS, Nurunnabi M. Progress and prospect of nanotechnology for cardiac fibrosis treatment. INTERDISCIPLINARY MEDICINE 2023; 1:e20230018. [PMID: 38089921 PMCID: PMC10712437 DOI: 10.1002/inmd.20230018] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/17/2023] [Accepted: 08/03/2023] [Indexed: 02/01/2024]
Abstract
Cardiac fibrosis is the excessive accumulation of extracellular matrix components in the heart, leading to reduced cardiac functionality and heart failure. This review provides an overview of the therapeutic applications of nanotechnology for the treatment of cardiac fibrosis. We first delve into the fundamental pathophysiology of cardiac fibrosis, highlighting the key molecular players, including Matrix Metalloproteinases, Transforming Growth Factor-beta, and several growth factors, cytokines, and signaling molecules. Each target presents a unique opportunity to develop targeted nano-therapies. We then focus on recent advancements in nanotechnology and how nanoparticles can be engineered to deliver drugs or therapeutic genes. These advanced delivery approaches have shown significant potential to inhibit fibrosis-promoting factors, thereby mitigating the fibrotic response and potentially reversing disease progression. In addition, we discuss the challenges associated with developing and translating nanotechnology-based drug delivery systems, including ensuring biocompatibility, safety, and regulatory compliance. This review highlights how nanotechnology can bridge the gap between lab research and clinical practice for treating cardiac fibrosis.
Collapse
Affiliation(s)
- Samantha L. Gaytan
- Department of Pharmaceutical SciencesSchool of PharmacyThe University of Texas El PasoEl PasoTexasUSA
- Department of Interdisciplinary Health SciencesCollege of Health SciencesThe University of Texas El PasoEl PasoTexasUSA
| | - Elfa Beaven
- Department of Pharmaceutical SciencesSchool of PharmacyThe University of Texas El PasoEl PasoTexasUSA
- Department of Biomedical EngineeringCollege of EngineeringThe University of Texas El PasoEl PasoTexasUSA
| | - Shrikanth S. Gadad
- Center of Emphasis in CancerDepartment of Molecular and Translational MedicinePaul L. Foster School of MedicineTexas Tech University Health Sciences Center El PasoEl PasoTexasUSA
| | - Md Nurunnabi
- Department of Pharmaceutical SciencesSchool of PharmacyThe University of Texas El PasoEl PasoTexasUSA
- Department of Interdisciplinary Health SciencesCollege of Health SciencesThe University of Texas El PasoEl PasoTexasUSA
- Department of Biomedical EngineeringCollege of EngineeringThe University of Texas El PasoEl PasoTexasUSA
- Border Biomedical Research CenterThe University of Texas El PasoEl PasoTexasUSA
| |
Collapse
|
48
|
Wu L, Wu M, Zhao D, Chen S, Wang G, Xu L, Wang Y, An L, Wu S, Miao C, Hong J. Elevated high-sensitivity C-reactive protein levels increase the risk of new-onset cardiac conduction disorders. Cardiovasc Diabetol 2023; 22:268. [PMID: 37777746 PMCID: PMC10543876 DOI: 10.1186/s12933-023-01987-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/07/2023] [Indexed: 10/02/2023] Open
Abstract
BACKGROUND Previous studies have reported that inflammatory responses can promote the onset of cardiovascular diseases; however, its association with cardiac conduction disorders remains unclear. The present community-based cohort study aimed to elucidate the effects of inflammatory responses on the risk of developing cardiac conduction disorders. METHODS After the exclusion of participants failing to meet the inclusion criteria, 86,234 eligible participants (mean age: 50.57 ± 11.88 years) were included. The participants were divided into high-sensitivity C-reactive protein (hsCRP) ≤ 3 mg/L, and hsCRP > 3 mg/L groups based on hsCRP values. Multivariate Cox proportional hazard model was used to analyze the relationship between inflammatory responses and various cardiac conduction disorders. RESULTS After adjusting for confounding factors, we observed that compared with the hsCRP ≤ 3 mg/L group, the hsCRP > 3 mg/L group exhibited increased risks of atrioventricular block (hazard ratio [HR]:1.64, 95%confidence interval [CI] 1.44-1.87) and left (HR:1.25, 95% CI 1.07-1.45) and right bundle branch block (HR:1.31, 95% CI 1.17-1.47). Moreover, the risk of various cardiac conduction disorders increased for every 1 standard deviation increase in log (hsCRP). The restricted cubic spline function confirmed a linear relationship between log (hsCRP) and the risk of developing cardiac conduction disorders (All nonlinearity P > 0.05). CONCLUSIONS High hsCRP levels are an independent risk factor for cardiac conduction disorders, and hsCRP levels are dose-dependently associated with the risk of conduction disorders. Our study results may provide new strategies for preventing cardiac conduction disorders.
Collapse
Affiliation(s)
- Lili Wu
- Department of Cardiology, Shanghai Songjiang District Central Hospital, Shanghai, China
- Division of Cardiovascular Diseases, Department of Emergency and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.100 Haining Road, Hongkou District, Shanghai, 200080, China
| | - Meimei Wu
- Department of Emergency and Critical Care Medicine, Shanghai Songjiang District Central Hospital, Shanghai, China
| | - Dandan Zhao
- Division of Cardiovascular Diseases, Department of Emergency and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.100 Haining Road, Hongkou District, Shanghai, 200080, China
| | - Shuohua Chen
- Department of Cardiology, Kailuan Hospital, North China University of Science and Technology, No. 57 Xinhua East Road, Tangshan, 063001, China
| | - Guodong Wang
- Department of Cardiology, Kailuan Hospital, North China University of Science and Technology, No. 57 Xinhua East Road, Tangshan, 063001, China
| | - Lina Xu
- Division of Cardiovascular Diseases, Department of Emergency and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.100 Haining Road, Hongkou District, Shanghai, 200080, China
| | - Yujing Wang
- Division of Cardiovascular Diseases, Department of Emergency and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.100 Haining Road, Hongkou District, Shanghai, 200080, China
| | - Lina An
- Division of Cardiovascular Diseases, Department of Emergency and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.100 Haining Road, Hongkou District, Shanghai, 200080, China
| | - Shouling Wu
- Department of Cardiology, Kailuan Hospital, North China University of Science and Technology, No. 57 Xinhua East Road, Tangshan, 063001, China.
| | - Congliang Miao
- Division of Cardiovascular Diseases, Department of Emergency and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.100 Haining Road, Hongkou District, Shanghai, 200080, China.
| | - Jiang Hong
- Division of Cardiovascular Diseases, Department of Emergency and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.100 Haining Road, Hongkou District, Shanghai, 200080, China.
| |
Collapse
|
49
|
Izadifar M, Berecz T, Li B, Tang JKKY, Foldes G, Apati A, Nagy A. Speckle-Tracking Strain Analysis for Mapping Spatiotemporal Contractility of Induced Pluripotent Stem Cell (iPSC)-Derived Cardiomyocytes. Curr Protoc 2023; 3:e889. [PMID: 37747346 DOI: 10.1002/cpz1.889] [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] [Indexed: 09/26/2023]
Abstract
Human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (hiPSC-CMs) hold tremendous potential for cardiovascular disease modeling, drug screening, personalized medicine, and pathophysiology studies. The availability of a robust protocol and functional assay for studying phenotypic behavior of hiPSC-CMs is essential for establishing an in vitro disease model. Many heart diseases manifest due to changes in the mechanical strain of cardiac tissue. Therefore, non-invasive evaluation of the contractility properties of hiPSC-CMs remains crucial to gain an insight into the pathogenesis of cardiac diseases. Speckle tracking-based strain analysis is an efficient non-invasive method that uses video microscopy and image analysis of beating hiPSC-CMs for quantitative evaluation of mechanical contractility properties. This article presents step-by-step protocols for extracting quantitative contractility properties of an hiPSC-CM system obtained from five members of a family, of whom three were affected by DiGeorge syndrome, using speckle tracking-based strain analysis. The hiPSCs from the family members were differentiated and purified into hiPSC-CMs using metabolic selection. Time-lapse images of hiPSC-CMs were acquired using high-spatial-resolution and high-time-resolution phase-contrast video microscopy. Speckled images were characterized by evaluating the cross-correlation coefficient, speckle size, speckle contrast, and speckle quality of the images. The optimum parameters of the speckle tracking algorithm were determined by performing sensitivity analysis concerning computation time, effective mapping area, average contraction velocity, and strain. Furthermore, the hiPSC-CM response to adrenaline was evaluated to validate the sensitivity of the strain analysis algorithm. Then, we applied speckle tracking-based strain analysis to characterize the dynamic behavior of patient-specific hiPSC-CMs from the family members affected/unaffected by DiGeorge syndrome. Here, we report an efficient and manipulation-free method to analyze the contraction displacement vector and velocity field, contraction-relaxation strain rate, and contractile cycles. Implementation of this method allows for quantitative analysis of the contractile phenotype characteristics of hiPSC-CMs to distinguish possible cardiac manifestation of DiGeorge syndrome. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Differentiation of iPSCs into iPSC-derived cardiomyocytes (iPSC-CMs) and metabolic selection of differentiated iPSC-CMs Support Protocol 1: Culture, maintenance, and expansion of human iPSCs Support Protocol 2: Immunohistochemistry of iPSC-CMs Basic Protocol 2: Time-lapse speckle imaging of iPSC-CMs and speckle quality characterization Support Protocol 3: Enhancement of local contrast of videos by applying contrast limited adaptive histogram equalization (CLAHE) to all frames Support Protocol 4: Evaluation of average speckle size Support Protocol 5: Evaluation of average speckle contrast Support Protocol 6: Determination of relative peak height, Pc(x), of consecutive images acquired from video microscopy of iPSC-CMs Basic Protocol 3: Speckle tracking-based analysis of beating iPSC-CMs Support Protocol 7: Validation of sensitivity of the speckle tracking analysis for mapping the contractility of iPSC-CMs Basic Protocol 4: Data extraction, visualization, and mapping of contractile cycles of iPSC-CMs.
Collapse
Affiliation(s)
- Mohammad Izadifar
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Tunde Berecz
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Biao Li
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | | | - Gabor Foldes
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Agota Apati
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Andras Nagy
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
50
|
Liu C, Zhou D, Zhang Q, Wei H, Lu Y, Li B, Zhan H, Cheng J, Wang C, Yang Y, Li S, Hu C, Liao X. Transcription factor EB (TFEB) improves ventricular remodeling after myocardial infarction by inhibiting Wnt/ β-catenin signaling pathway. PeerJ 2023; 11:e15841. [PMID: 37609444 PMCID: PMC10441526 DOI: 10.7717/peerj.15841] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/12/2023] [Indexed: 08/24/2023] Open
Abstract
Background Adverse left ventricular remodeling after myocardial infarction (MI) compromises cardiac function and increases heart failure risk. Until now, comprehension of the role transcription factor EB (TFEB) plays after MI is limited. Objectives The purpose of this study was to describe the effects of TFEB on fibroblasts differentiation and extracellular matrix expression after MI. Methods AAV9 (adeno-associated virus) mediated up- and down-regulated TFEB expressions were generated in C57BL/6 mice two weeks before the MI modeling. Echocardiography, Masson, Sirius red staining immunofluorescence, and wheat germ agglutinin staining were performed at 3 days, and 1, 2, and 4 weeks after MI modeling. Fibroblasts collected from SD neonatal rats were transfected by adenovirus and siRNA, and cell counting kit-8 (CCK8), immunofluorescence, wound healing and Transwell assay were conducted. Myocardial fibrosis-related proteins were identified by Western blot. PNU-74654 (100 ng/mL) was used for 12 hours to inhibit β-catenin-TCF/LEF1 complex. Results The up-regulation of TFEB resulted in reduced fibroblasts proliferation and its differentiation into myofibroblasts in vitro studies. A significant up-regulation of EF and down-regulation of myocyte area was shown in the AAV9-TFEB group. Meanwhile, decreased protein level of α-SMA and collagen I were observed in vitro study. TFEB didn't affect the concentration of β-catenin. Inhibition of TFEB, which promoted cell migration, proliferation and collagen I expression, was counteracted by PNU-74654. Conclusions TFEB demonstrated potential in restraining fibrosis after MI by inhibiting the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Cong Liu
- Department of Emergency Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Dawang Zhou
- Department of Emergency Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Qiang Zhang
- Department of Emergency Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Hongyan Wei
- Department of Emergency Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuanzheng Lu
- Department of Emergency Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Bo Li
- Department of Emergency Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Haohong Zhan
- Department of Emergency Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jingge Cheng
- Department of Emergency Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Chuyue Wang
- Department of Emergency Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yilin Yang
- Department of Emergency Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuhao Li
- Department of Emergency Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chunlin Hu
- Department of Emergency Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoxing Liao
- Department of Emergency Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|