1
|
Ding HS, Qu JF, Luo XJ, Luo ZH, Huang ZY, Zhong P, Li X, Liu XX. Maresin 1 attenuates myocardium ischemia/reperfusion injury via SIRT1/HMGB-1/NLRP-3-related mechanisms. Eur J Pharmacol 2025; 998:177456. [PMID: 40054718 DOI: 10.1016/j.ejphar.2025.177456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 03/02/2025] [Accepted: 03/04/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND AND PURPOSE Maresin 1 (MaR 1) has demonstrated potent anti-inflammatory and antioxidant effects in different animal models. However, its impact on heart ischemia/reperfusion injury (IRI) remains uncertain. Pyroptosis, a pro-inflammatory programmed cell death, is associated with IRI. The goal of our research was to assess the role of MaR 1 on pyroptosis during heart IRI mice. METHODS Cardiac IRI was induced in a mouse model, and hypoxia/reoxygenation (H/R) was conducted on neonatal rat ventricle myocytes (NRVMs) to establish an in vitro model. The effects of MaR 1 were assessed using measures such as cardiac infarct area, heart tissue injury, hemodynamic monitoring, apoptotic index, pyroptosis-related proteins, inflammatory reaction and heart enzyme activities. RESULTS MaR 1 injection obviously reduced cardiac infarct area and apoptosis, inhibited myocardial pyroptosis, decreased pro-inflammatory cytokines and suppressed apoptosis via Silent information regulator factor 2-related enzyme 1 (SIRT1). Additionally, MaR 1 injection markedly suppressed the expression of High-mobility group box 1 (HMGB-1)/nuclear factor-κB (NF-κB)/(NOD)-like receptor Pyrin domain-containing 3 (NLRP-3) axis-related proteins by SIRT1. In isolated NRVMs, MaR 1 increased cellular viability, diminished heart enzyme activities and inhibited apoptosis and inflammation. Furthermore, in vitro studies demonstrated that the SIRT1 inhibitor decreased the anti-inflammatory and anti-apoptosis properties of MaR 1 in NRVMs through the HMGB-1/NF-κB/NLRP-3 axis. CONCLUSION Our research suggests that MaR 1 pretreatment may alleviate cardiac IRI and suppress pyroptosis and apoptosis both in vivo and in vitro. MaR 1 inhibits pyroptosis through the SIRT1/HMGB-1/NF-κB/NLRP-3 axis. Therefore, MaR 1 may serve as a promising treatment for cardiac IRI.
Collapse
Affiliation(s)
- Hua-Sheng Ding
- Department of Emergency, Shenzhen Hospital, Southern Medical University, Shenzhen, 518101, PR China.
| | - Ji-Fu Qu
- Department of Emergency, Shenzhen Hospital, Southern Medical University, Shenzhen, 518101, PR China
| | - Xing-Jun Luo
- Department of Anesthesiology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, PR China
| | - Zhi-Hui Luo
- Department of Emergency, Shenzhen Hospital, Southern Medical University, Shenzhen, 518101, PR China
| | - Zhong-Yi Huang
- Department of Emergency, Shenzhen Hospital, Southern Medical University, Shenzhen, 518101, PR China
| | - Peng Zhong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, PR China; Institute of Cardiovascular Diseases, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China
| | - Xin Li
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China.
| | - Xiao-Xiong Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, PR China; Institute of Cardiovascular Diseases, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| |
Collapse
|
2
|
Mylonas N, Siokatas G, Zacharia E, Pol C, Rolland T, Kyriazis ID, Hoffman M, Hildebrand A, Bannister T, Gao E, Goldberg IJ, Yang VW, Bialkowska AB, Elrod JW, Canty JM, Andreadou I, Weil B, Drosatos K. Cardiac ischaemia/reperfusion in pigs and mice increases cardiomyocyte Krüppel-like factor 5 that aggravates tissue injury and remodelling. Cardiovasc Res 2025; 121:900-914. [PMID: 40079359 PMCID: PMC12160836 DOI: 10.1093/cvr/cvaf040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 12/07/2024] [Accepted: 01/22/2025] [Indexed: 03/15/2025] Open
Abstract
AIMS Activation of the transcriptional factor Krüppel-like factor 5 (KLF5) is detrimental to chronic heart failure. We explored the involvement of KLF5 in myocardial ischaemia/reperfusion injury. METHODS AND RESULTS Yorkshire pigs underwent 75' of ischaemia, followed by 3 or 24 h of reperfusion. C57BL/6J mice underwent 30' of ischaemia, followed by 10', 2, 12, 24 h, or 4 weeks of reperfusion. Hearts and isolated cardiomyocytes (CMs) were analysed for gene expression. We assessed cardiac function, infarct size (IS), oxidative stress, and fibrosis in mice subjected to pharmacologic or genetic KLF5 inhibition, as well as pharmacologic inhibition of NADPH oxidases or glucose transporter (GLUT)1 and GLUT4. Bulk RNA sequencing, untargeted 1H-NMR metabolomics, and LC-MS lipidomics were performed. Isolated primary murine CMs were infected with recombinant adenovirus expressing KLF5. During reperfusion, CM KLF5 expression was increased in porcine and murine hearts. Pharmacologic or CM-specific genetic inhibition of KLF5 reduced IS and improved cardiac function in mice. Importantly, acute KLF5 inhibition during early reperfusion suppressed fibrosis and preserved systolic cardiac function 4 weeks post-ischaemia/reperfusion. This improvement was associated with lower NADPH-oxidase 4 (NOX4) expression, less oxidative stress, and suppressed inflammation and cell apoptosis. Pharmacologic inhibition of NOX4 conferred the same benefit. Metabolomic analysis indicated that KLF5 inhibition lowered glucose-derived metabolites (UDP-glucose and lactate) at early reperfusion. Accordingly, cardiac GLUT1 and GLUT4 levels were increased with ischaemia/reperfusion, which was reverted by KLF5 inhibition. Pharmacologic inhibition of both GLUT1 and GLUT4 reduced IS. Finally, myocardial KLF5 overexpression increased GLUT1 mRNA levels and mouse mortality. CONCLUSION Ischaemia/reperfusion increases CM KLF5 expression in pigs and mice. This constitutes a central element of myocardial injury pathophysiology and is associated with stimulation of GLUT1 and GLUT4 expression, activation of NOX4, oxidative stress, inflammation, and apoptosis. Acute KLF5 inhibition during reperfusion constitutes a novel therapeutic approach against myocardial ischaemia/reperfusion injury.
Collapse
Affiliation(s)
- Nikolaos Mylonas
- Metabolic Biology Laboratory, Cardiovascular Center, Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, 231 Albert Sabin Way, CVC-5939, Cincinnati, OH 45267, USA
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou, Athens 15771, Greece
| | - Georgios Siokatas
- Metabolic Biology Laboratory, Cardiovascular Center, Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, 231 Albert Sabin Way, CVC-5939, Cincinnati, OH 45267, USA
| | - Effimia Zacharia
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad St., Philadelphia, PA 19140, USA
| | - Christine Pol
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad St., Philadelphia, PA 19140, USA
| | - Tyler Rolland
- Department of Physiology and Biophysics, The Clinical and Translational Research Center of the University at Buffalo, 875 Ellicott St., Buffalo, NY 14203, USA
| | - Ioannis D Kyriazis
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad St., Philadelphia, PA 19140, USA
- Laboratory of Pharmacology, Faculty of Medicine, University of Thessaly, Larissa 41500, Greece
| | - Matthew Hoffman
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad St., Philadelphia, PA 19140, USA
| | - Alycia Hildebrand
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad St., Philadelphia, PA 19140, USA
| | - Thomas Bannister
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, 120 Scripps Way, Jupiter, FL 33458, USA
| | - Erhe Gao
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad St., Philadelphia, PA 19140, USA
| | - Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, 550 1st Ave., New York, NY 10016, USA
| | - Vincent W Yang
- Department of Medicine, Renaissance School of Medicine, Stony Brook University, 100 Nicolls Rd, Stony Brook, NY 11794, USA
| | - Agnieszka B Bialkowska
- Department of Medicine, Renaissance School of Medicine, Stony Brook University, 100 Nicolls Rd, Stony Brook, NY 11794, USA
| | - John W Elrod
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad St., Philadelphia, PA 19140, USA
| | - John M Canty
- Department of Physiology and Biophysics, The Clinical and Translational Research Center of the University at Buffalo, 875 Ellicott St., Buffalo, NY 14203, USA
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou, Athens 15771, Greece
| | - Brian Weil
- Department of Physiology and Biophysics, The Clinical and Translational Research Center of the University at Buffalo, 875 Ellicott St., Buffalo, NY 14203, USA
| | - Konstantinos Drosatos
- Metabolic Biology Laboratory, Cardiovascular Center, Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, 231 Albert Sabin Way, CVC-5939, Cincinnati, OH 45267, USA
| |
Collapse
|
3
|
Alcover S, López S, Ramos-Regalado L, Muñoz-García N, Gallinat A, Suades R, Badimon L, Vilahur G. Cardioprotection During Myocardial Infarction in Diabetic Cardiomyopathy. Diabetes 2025; 74:1021-1032. [PMID: 40080393 PMCID: PMC12097457 DOI: 10.2337/db24-0510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 03/07/2025] [Indexed: 03/15/2025]
Abstract
Patients with diabetes are at an increased risk of diabetic cardiomyopathy (DCM) and acute myocardial infarction (AMI). Protecting the heart against AMI is more challenging in DCM than in nondiabetic hearts. We investigated whether intravenous (i.v.) atorvastatin administration during AMI exerts cardioprotection in DCM as seen in nondiabetic hearts. Sprague-Dawley rats were divided into streptozotocin-induced DCM and normoglycemic control groups. Our model of DCM rats exhibited interstitial fibrosis and cardiac dysfunction at 5 weeks. At this time point, all animals underwent AMI induction (coronary ligation for 45 min), receiving i.v. atorvastatin or vehicle during ischemia. Animals were reperfused and sacrificed 24 h later for myocardial infarct size analysis and cardiac tissue sampling. Echocardiography was performed. DCM vehicle rats had larger infarcts than normoglycemic vehicle-treated animals at a comparable area-at-risk. Intravenous atorvastatin reduced infarct size and preserved systolic function in both groups. Compared with vehicle animals, i.v. atorvastatin inhibited RhoA membrane translocation, induced AMPK phosphorylation, prevented apoptosis execution, and improved cardiac remodelling in the infarcted heart of both groups, whereas innate immune cell infiltration was further reduced in i.v. atorvastatin-treated DCM animals. The proven cardioprotective effectiveness of this i.v. statin formulation in the presence of DCM warrants its further development into a clinically therapeutic option. ARTICLE HIGHLIGHTS Diabetic cardiomyopathy (DCM) significantly increases the risk of acute myocardial infarction and attenuates or abolishes the cardioprotective effects of several therapeutic approaches. Whether intravenous atorvastatin administration during ongoing acute myocardial infarction retains its cardioprotective potential in the presence of DCM was investigated. Intravenous atorvastatin during ischemia reduces infarct size and preserves cardiac function in DCM rats. The efficacy of this intravenous statin formulation in DCM supports its development as a viable therapeutic option for clinical use.
Collapse
Affiliation(s)
| | - Sergi López
- Sant Pau Research Institute (IR SANT PAU), Barcelona, Spain
| | | | | | - Alex Gallinat
- Sant Pau Research Institute (IR SANT PAU), Barcelona, Spain
| | - Rosa Suades
- Sant Pau Research Institute (IR SANT PAU), Barcelona, Spain
| | - Lina Badimon
- Sant Pau Research Institute (IR SANT PAU), Barcelona, Spain
- CiberCV, Institute of Health Carlos III, Madrid, Spain
| | - Gemma Vilahur
- Sant Pau Research Institute (IR SANT PAU), Barcelona, Spain
- CiberCV, Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
4
|
Al Rifai N, Stone K, Bo B, Zhang B, Kasiviswanathan D, Redington AN, Haworth KJ. Assessing the oxygen scavenging capacity and myocardial gas embolization risk of ultrasonically activated phase shift perfluorobutane droplets. J Mater Chem B 2025. [PMID: 40405824 PMCID: PMC12099491 DOI: 10.1039/d4tb02700k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 04/29/2025] [Indexed: 05/24/2025]
Abstract
This study investigated oxygen scavenging efficiency and the risk of embolization of the cardiac vasculature using ultrasound-triggered phase-shift perfluorobutane (PFB) droplets in vitro and ex vivo. The emulsion comprised lipid-shelled perfluorobutane core droplets with a modal diameter of 0.98 ± 0.03 μm. The droplets were prepared using a high-pressure microfluidizer. The embolization risk was assessed using a modified ex vivo rat Langendorff preparation to accommodate an EkoSonic™ Endovascular Device. The EkoSonic™ Device was composed of an infusion catheter and an ultrasonic core to generate ultrasound at 2.35 MHz and nucleate acoustic droplet vaporization of the droplets. The oxygen scavenging efficiency was studied in an isolated beating heart and an in vitro flow phantom setup with target concentrations ranging from 0.05 × 10-4 to 5.0 × 10-4 mL mL-1. Gas embolization from the acoustic droplet vaporization (ADV)-nucleated microbubbles was assessed based on cardiac perfusion and cardiac functional parameters. No change in cardiac perfusion was observed when using droplets with target concentrations below 1.5 × 10-4 mL mL-1, either with or without ultrasound insonation of the droplets. Oxygen scavenging increased with increasing droplet target concentration. The ADV transition efficiency increased with increasing droplet concentration between 0.05 × 10-4 and 0.5 × 10-4 mL mL-1 and decreased for higher concentrations. The conclusion of this study was that ultrasound-triggered phase-shift perfluorobutane droplets effectively scavenge oxygen without causing significant embolization at concentrations below 1.5 × 10-4 mL mL-1. Oxygen scavenging increased with higher droplet concentrations, whereas the transition efficiency of ADV reached the largest value at 0.5 × 10-4 mL mL-1, indicating an optimal performance balancing safety and efficacy exists.
Collapse
Affiliation(s)
- Nour Al Rifai
- Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267-0586, USA.
| | - Kateryna Stone
- Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267-0586, USA.
| | - Bin Bo
- Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267-0586, USA.
| | - Bin Zhang
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45229, USA
| | | | - Andrew N Redington
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Kevin J Haworth
- Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267-0586, USA.
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45229, USA
- Medical Sciences Baccalaureate Program, University of Cincinnati, Cincinnati, OH 45267-0586, USA
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45267-0586, USA
| |
Collapse
|
5
|
Davidson SM, Andreadou I, Antoniades C, Bartunek J, Basso C, Brundel BJJM, Byrne RA, Chiva-Blanch G, da Costa Martins P, Evans PC, Girão H, Giricz Z, Gollmann-Tepeköylü C, Guzik T, Gyöngyösi M, Hübner N, Joner M, Kleinbongard P, Krieg T, Liehn E, Madonna R, Maguy A, Paillard M, Pesce M, Petersen SE, Schiattarella GG, Sluijter JPG, Steffens S, Streckfuss-Bömeke K, Thielmann M, Tucker A, Van Linthout S, Wijns W, Wojta J, Wu JC, Perrino C. Opportunities and challenges for the use of human samples in translational cardiovascular research: a scientific statement of the ESC Working Group on Cellular Biology of the Heart, the ESC Working Group on Cardiovascular Surgery, the ESC Council on Basic Cardiovascular Science, the ESC Scientists of Tomorrow, the European Association of Percutaneous Cardiovascular Interventions of the ESC, and the Heart Failure Association of the ESC. Cardiovasc Res 2025; 121:702-729. [PMID: 40084813 PMCID: PMC12101359 DOI: 10.1093/cvr/cvaf023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/23/2024] [Accepted: 10/21/2024] [Indexed: 03/16/2025] Open
Abstract
Animal models offer invaluable insights into disease mechanisms but cannot entirely mimic the variability and heterogeneity of human populations, nor the increasing prevalence of multi-morbidity. Consequently, employing human samples-such as whole blood or fractions, valvular and vascular tissues, myocardium, pericardium, or human-derived cells-is essential for enhancing the translational relevance of cardiovascular research. For instance, myocardial tissue slices, which preserve crucial structural and functional characteristics of the human heart, can be used in vitro to examine drug responses. Human blood serves as a rich source of biomarkers, including extracellular vesicles, various types of RNA (miRNA, lncRNA, and circRNAs), circulating inflammatory cells, and endothelial colony-forming cells, facilitating detailed studies of cardiovascular diseases. Primary cardiomyocytes and vascular cells isolated from human tissues are invaluable for mechanistic investigations in vitro. In cases where these are unavailable, human induced pluripotent stem cells serve as effective substitutes, albeit with specific limitations. However, the use of human samples presents challenges such as ethical approvals, tissue procurement and storage, variability in patient genetics and treatment regimens, and the selection of appropriate control samples. Biobanks are central to the efficient use of these scarce and valuable resources. This scientific statement discusses opportunities to implement the use of human samples for cardiovascular research within specific clinical contexts, offers a practical framework for acquiring and utilizing different human materials, and presents examples of human sample applications for specific cardiovascular diseases, providing a valuable resource for clinicians, translational and basic scientists engaged in cardiovascular research.
Collapse
Affiliation(s)
- Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Ioanna Andreadou
- School of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Charalambos Antoniades
- RDM Division of Cardiovascular Medicine, Acute Multidisciplinary Imaging and Interventional Centre, University of Oxford, Headley Way, Headington, Oxford OX3 9DU, UK
| | - Jozef Bartunek
- Cardiovascular Center Aalst, OLV Hospital, Aalst, Belgium
| | - Cristina Basso
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, Cardiovascular Pathology, University of Padua, Padua, Italy
| | - Bianca J J M Brundel
- Physiology, Amsterdam UMC Location Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, The Netherlands
| | - Robert A Byrne
- Cardiovascular Research Institute Dublin, Mater Private Network, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Gemma Chiva-Blanch
- Faculty of Health Sciences, Universitat Oberta de Catalunya, Barcelona, Spain
- Department of Endocrinology and Nutrition, August Pi i Sunyer Biomedical Research Institute, Hospital Clínic of Barcelona, Barcelona, Spain
- Biomedical Network Research Centre on Obesity and Nutrition Physiopathology, Instituto de Salud Carlos III, Madrid, Spain
| | - Paula da Costa Martins
- Department of Molecular Genetics, Faculty of Sciences and Engineering, Maastricht, The Netherlands
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Paul C Evans
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Henrique Girão
- Center for Innovative Biomedicine and Biotechnology, Clinical Academic Centre of Coimbra, Faculty of Medicine, University of Coimbra, Coimbra Institute for Clinical and Biomedical Research, Coimbra, Portugal
| | - Zoltan Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Can Gollmann-Tepeköylü
- Department for Cardiac Surgery, Cardiac Regeneration Research, Medical University of Innsbruck, Anichstraße 35 A, 6020 Innsbruck, Austria
| | - Tomasz Guzik
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Mariann Gyöngyösi
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Norbert Hübner
- Max Delbrück Center in the Helmholtz Association, Berlin, Germany
- Charite-Universitätsmedizin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Michael Joner
- Department of Cardiology, German Heart Center Munich, Technical University of Munich, Lazarettstrasse 36, 80636 Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Petra Kleinbongard
- Faculty of Medicine University of Duisburg-Essen, Institute of Pathophysiology, Duisburg-Essen, Germany
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Elisa Liehn
- Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Rosalinda Madonna
- Cardiology Division, Department of Pathology, University of Pisa, Pisa, Italy
| | - Ange Maguy
- Department of Physiology, University of Bern, Bern, Switzerland
| | - Melanie Paillard
- Laboratoire CarMeN—IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, Univ-Lyon, 69500 Bron, France
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, Milan, Italy
- Department of Aerospace and Mechanical Engineering, Politecnico di Torino, Italy
- Department of Cell Biology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Steffen E Petersen
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University London, Charterhouse Square, London, UK
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, West Smithfield, London, UK
- Health Data Research UK, London, UK
- Alan Turing Institute, London, UK
| | - Gabriele G Schiattarella
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Department of Advanced Biomedical Sciences, Federico II University, Via Pansini 5, 80131 Naples, Italy
- Deutsches Herzzentrum der Charité (DHZC), Charité-Universitätsmedizin Berlin, Berlin, Germany
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Joost P G Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Sabine Steffens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Katrin Streckfuss-Bömeke
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
- Clinic for Cardiology and Pneumology, University Medicine Göttingen, Germany and German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| | - Matthias Thielmann
- West-German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Art Tucker
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University London, Charterhouse Square, London, UK
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, West Smithfield, London, UK
| | - Sophie Van Linthout
- Berlin Institute of Health at Charité, BIH Center for Regenerative Therapies, Universitätmedizin Berlin, Berlin, Germany
- Max Delbrück Center in the Helmholtz Association, Berlin, Germany
| | - William Wijns
- The Lambe Institute for Translational Research and Curam, University of Galway, Galway, Ireland
| | - Johann Wojta
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
- Core Facilities, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford, CA, USA
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
6
|
Homilius C, Seefeldt JM, Hansen J, Nielsen R, de Paoli FV, Boedtkjer E. Lactate orchestrates metabolic hemodynamic adaptations through a unique combination of venocontraction, artery relaxation, and positive inotropy. Acta Physiol (Oxf) 2025; 241:e70037. [PMID: 40167405 PMCID: PMC11960580 DOI: 10.1111/apha.70037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/14/2025] [Accepted: 03/14/2025] [Indexed: 04/02/2025]
Abstract
AIM H+ facilitates metabolic blood flow regulation while negatively impacting cardiac contractility. Cardiovascular consequences of conjugate bases accumulating alongside H+ remain unclear. Here, we evaluate the cardiovascular effects of nine prominent carboxylates-particularly lactate, 3-hydroxybutyrate, and butyrate-linked to metabolic and microbial activity. METHODS Comparing the actions of pH-adjusted Na-carboxylates to equiosmolar NaCl, we study arteries and veins isolated from healthy rats and humans with ischaemic heart disease, isolated perfused rat hearts, and rat cardiovascular function in vivo. RESULTS The tested carboxylates generally relax arteries and veins. L-lactate relaxes human and rat arteries up to 70% (EC50 = 10.1 mM) and rat brachial and mesenteric veins up to 30% of pre-contractions, yet stands out by augmenting contractions of rat femoral, saphenous, and lateral marginal veins and human internal thoracic and great saphenous veins up to 50%. D-lactate shows only minor actions. In isolated perfused hearts, 10 mM L-lactate increases coronary flow (17.1 ± 7.7%) and left ventricular developed pressure (10.1 ± 3.0%) without affecting heart rate. L-lactate infusion in rats-reaching 3.7 ± 0.3 mM in the circulation-increases left ventricular end-diastolic volume (11.3 ± 2.8%), stroke volume (22.6 ± 3.0%), cardiac output (23.4 ± 3.5%), and ejection fraction (10.6 ± 2.0%), and lowers systemic vascular resistance (34.1 ± 3.7%) without influencing blood pressure or heart rate. The ketone body 3-hydroxybutyrate causes lactate accumulation and elevates left ventricular end-diastolic volume in vivo. CONCLUSION Carboxylate metabolites generally relax arteries and veins. L-lactate relaxes arteries, lowering systemic vascular resistance, causes preferential venocontraction with increased ventricular diastolic filling, and elevates cardiac contractility and cardiac output. We propose that L-lactate optimizes cardiovascular function during metabolic disturbances.
Collapse
Affiliation(s)
| | - Jacob M. Seefeldt
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of CardiologyAarhus University HospitalAarhusDenmark
| | - Jakob Hansen
- Department of Forensic MedicineAarhus UniversityAarhusDenmark
| | - Roni Nielsen
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of CardiologyAarhus University HospitalAarhusDenmark
| | - Frank V. de Paoli
- Department of BiomedicineAarhus UniversityAarhusDenmark
- Department of Cardiothoracic and Vascular SurgeryAarhus University HospitalAarhusDenmark
| | | |
Collapse
|
7
|
Zhi Y, Yang B, Huo J, Wang H, Yang B, Zhou YF, Xiao F, Yang HQ. Tyrosine phosphorylation of Kir6.2 subunit negatively regulates cardiac K ATP channel activity. Basic Res Cardiol 2025:10.1007/s00395-025-01108-x. [PMID: 40251281 DOI: 10.1007/s00395-025-01108-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 04/06/2025] [Accepted: 04/08/2025] [Indexed: 04/20/2025]
Abstract
The plasma membrane ATP-sensitive potassium (KATP) channel in cardiac myocytes plays a critical role in protecting the heart against ischemic injury. Post-translational modifications regulate KATP channel activity and play a role in cardioprotection. However, the role of tyrosine phosphorylation in KATP channel regulation remains unclear. In this study, we investigated the cardiac KATP channel subtype Kir6.2/SUR2A and demonstrated that a protein tyrosine kinase inhibitor significantly increased the current amplitude through blunting the ATP sensitivity of KATP channels without altering the single-channel current or the channel surface expression. Mutation screening identified Y258 in the Kir6.2 subunit as the tyrosine phosphorylation site of the KATP channel. In cardiomyocytes, KATP channel currents can be reversibly enhanced or weakened by inhibiting the tyrosine kinase epidermal growth factor receptor or the protein tyrosine phosphatase 1B. Furthermore, in a perfused mouse heart model, the inhibitor of epidermal growth factor receptor exhibited a significant cardioprotective effect in a KATP channel dependent manner, indicating the pharmacological potential for treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Yating Zhi
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Cyrus Tang Medical Institute, Medical College of Soochow University, Suzhou, 215028, China
| | - Bin Yang
- Department of Bioinformatics and Computational Biology, School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Jianyi Huo
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Cyrus Tang Medical Institute, Medical College of Soochow University, Suzhou, 215028, China
| | - Haojie Wang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Cyrus Tang Medical Institute, Medical College of Soochow University, Suzhou, 215028, China
| | - Bo Yang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Cyrus Tang Medical Institute, Medical College of Soochow University, Suzhou, 215028, China
| | - Ya-Feng Zhou
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Cyrus Tang Medical Institute, Medical College of Soochow University, Suzhou, 215028, China.
| | - Fei Xiao
- Department of Bioinformatics and Computational Biology, School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, China.
| | - Hua-Qian Yang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Cyrus Tang Medical Institute, Medical College of Soochow University, Suzhou, 215028, China.
| |
Collapse
|
8
|
Gyöngyösi M, Guthrie J, Hasimbegovic E, Han E, Riesenhuber M, Hamzaraj K, Bergler-Klein J, Traxler D, Emmert MY, Hackl M, Derdak S, Lukovic D. Critical analysis of descriptive microRNA data in the translational research on cardioprotection and cardiac repair: lost in the complexity of bioinformatics. Basic Res Cardiol 2025:10.1007/s00395-025-01104-1. [PMID: 40205177 DOI: 10.1007/s00395-025-01104-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/11/2025]
Abstract
The unsuccessful translation of cardiac regeneration and cardioprotection from animal experiments to clinical applications in humans has raised the question of whether microRNA bioinformatics can narrow the gap between animal and human research outputs. We reviewed the literature for the period between 2000 and 2024 and found 178 microRNAs involved in cardioprotection and cardiac regeneration. On analyzing the orthologs and annotations, as well as downstream regulation, we observed species-specific differences in the diverse regulation of the microRNAs and related genes and transcriptomes, the influence of the experimental setting on the microRNA-guided biological responses, and database-specific bioinformatics results. We concluded that, in addition to reducing the number of in vivo experiments, following the 3R animal experiment rules, the bioinformatics approach allows the prediction of several currently unknown interactions between pathways, coding and non-coding genes, proteins, and downstream regulatory elements. However, a comprehensive analysis of the miRNA-mRNA-protein networks needs a profound bioinformatics and mathematical education and training to appropriately design an experimental study, select the right bioinformatics tool with programming language skills and understand and display the bioinformatics output of the results to translate the research data into clinical practice. In addition, using in-silico approaches, a risk of deviating from the in vivo processes exists, with adverse consequences on the translational research.
Collapse
Affiliation(s)
- Mariann Gyöngyösi
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria.
| | - Julia Guthrie
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Zimmermannplatz 10, 1090, Vienna, Austria
| | - Ena Hasimbegovic
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Emilie Han
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Martin Riesenhuber
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Kevin Hamzaraj
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Jutta Bergler-Klein
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Denise Traxler
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Maximilian Y Emmert
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charite (DHZC), Berlin, Germany
| | | | - Sophia Derdak
- Core Facilities, Medical University of Vienna, Vienna, Austria
| | - Dominika Lukovic
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
9
|
Kakaei Y, Hussain S, Elmahdy A, Berger E, Shekka Espinosa A, Sevastianova V, Sheybani Z, Al-Awar A, Kalani M, Jha S, Zulfaj E, Nejat A, Jha A, Pylova T, Krasnikova M, Andersson EA, Silva VRR, Omerovic E, Redfors B. Comparison of the proteomic landscape in experimental ischemia reperfusion with versus without ischemic preconditioning. Sci Rep 2025; 15:11836. [PMID: 40195349 PMCID: PMC11976975 DOI: 10.1038/s41598-025-90735-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 02/14/2025] [Indexed: 04/09/2025] Open
Abstract
Myocardial ischemic preconditioning (IPC) enhances myocardial resilience to ischemic injury. Myocardial stunning is a transient, reversible dysfunction, while necrosis involves irreversible cell death. The relationship between IPC, stunning, and necrosis is not well understood, requiring further molecular investigation. This study aimed to investigate the proteomic changes associated with IPC, focusing on its relationship with myocardial stunning and necrosis. A novel 13.5-minute ischemia-reperfusion (I/R) rat model was specifically chosen to induce myocardial stunning, providing a unique approach to assess IPC effects in this context. Rats underwent either IPC with two 5-minute ischemia/reperfusion cycles followed by a 13.5-minute ischemic period or the procedure without IPC (no ischemic preconditioning, NIPC). Myocardial samples were collected at early (T1) and 4-hour post-reperfusion (T2) time points for proteomic analysis. Protein levels were quantified by differential labeling using TMTpro reagents, and subsequent liquid chromatography-mass spectrometry. IPC induced upregulation of proteins involved in endocytosis and Fc gamma R-mediated phagocytosis pathways at T1, while downregulating proteins related to tissue remodeling, immune response, and coagulation at T2. Conversely, NIPC exhibited upregulation of proteins associated with tissue damage and inflammation. IPC rats demonstrated enhanced leukocyte migration, complement activation, and immune response between T1 and T2. Consistent proteomic changes were observed between T1 and T2 in IPC vs. NIPC groups, and common alterations between IPC T2 vs. T1 and NIPC T2 vs. T1 comparisons underline shared pathways in cardiac complement and coagulation cascades. Our study reveals distinct proteomic changes induced by IPC in the context of myocardial stunning and necrosis. IPC activates early protective pathways, attenuates tissue damage and inflammation, and preserves myocardial function. These findings underscore IPC's reparative potential and identify myocardial stunning as an important, transient adaptation, which may have implications for supportive clinical management in I/R.
Collapse
Affiliation(s)
- Yalda Kakaei
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Shafaat Hussain
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Ahmed Elmahdy
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Evelin Berger
- Proteomics Core Facility, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Aaron Shekka Espinosa
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Valentyna Sevastianova
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Zahra Sheybani
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Amin Al-Awar
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Mana Kalani
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Sandeep Jha
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital/S, Gothenburg, Sweden
| | - Ermir Zulfaj
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
| | - Amirali Nejat
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
| | - Abhishek Jha
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Tetiana Pylova
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Maryna Krasnikova
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Erik Axel Andersson
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Vagner Ramon Rodrigues Silva
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Elmir Omerovic
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital/S, Gothenburg, Sweden
| | - Björn Redfors
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Bruna stråket 16, SU/S, 41345, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital/S, Gothenburg, Sweden
| |
Collapse
|
10
|
Hernandez-Resendiz S, Vilskersts R, Aluja D, Andreadou I, Bencsik P, Dambrova M, Efentakis P, Gao F, Giricz Z, Inserte J, Kelly-Laubscher R, Kiss A, Krieg T, Kwak BR, Lecour S, Lopaschuk G, Mączewski M, Waszkiewicz M, Oknińska M, Pagliaro P, Podesser B, Prag HA, Ruiz-Meana M, Szabados T, Zuurbier CJ, Ferdinandy P, Hausenloy DJ. IMproving Preclinical Assessment of Cardioprotective Therapies (IMPACT): a small animal acute myocardial infarction randomized-controlled multicenter study on the effect of ischemic preconditioning. Basic Res Cardiol 2025; 120:335-346. [PMID: 40072549 PMCID: PMC11976871 DOI: 10.1007/s00395-025-01102-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/17/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025]
Abstract
Although many cardioprotective interventions have been shown to limit infarct size (IS), in preclinical animal studies of acute myocardial ischemia/reperfusion injury (IRI), their clinical translation to patient benefit has been largely disappointing. A major factor is the lack of rigor and reproducibility in the preclinical studies. To address this, we have established the IMproving Preclinical Assessment of Cardioprotective Therapies (IMPACT) small animal multisite acute myocardial infarction (AMI) network, with centralized randomization and blinded core laboratory IS analysis, and have validated the network using ischemic preconditioning (IPC). Eight sites from the COST Innovators Grant (IG16225) network participated in the IMPACT AMI study. Mice and rats were randomly allocated into Sham, Control, or IPC groups. The IRI group underwent 45 min (mice) or 30 min (rats) of left coronary artery occlusion followed by 24 h reperfusion. IPC comprised three cycles of 5 min occlusion/reperfusion before IRI. IS was determined by a blinded core lab. The majority of site showed significant cardioprotection with IPC. In pooled mouse data, IPC (N = 42) reduced IS/AAR by 35% compared to control (N = 48) (30 ± 16% versus 46 ± 13%; p < 0.005), and in rat data, IPC (N = 36) reduced IS/AAR by 29% when compared to control (N = 39) (32 ± 19% versus 45 ± 14%; p < 0.01). The IMPACT multisite mouse and rat AMI networks, with centralized randomization and blinded core IS analysis, were established to improve the reproducibility of cardioprotective interventions in preclinical studies and to facilitate the translation of these therapies for patient benefit.
Collapse
Affiliation(s)
- Sauri Hernandez-Resendiz
- Cardiovascular and Metabolic Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Reinis Vilskersts
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
- Department of Pharmaceutical Chemistry, Riga Stradins University, Riga, Latvia
| | - David Aluja
- Vall d'hebron Institut de Recerca (VHIR), Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Péter Bencsik
- Department of Pharmacology and Pharmacotherapy, Albert-Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Maija Dambrova
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
- Department of Pharmaceutical Chemistry, Riga Stradins University, Riga, Latvia
| | - Panagiotis Efentakis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Fei Gao
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
- Centre for Quantitative Medicine (CQM), Duke-NUS Medical School, Singapore, Singapore
| | - Zoltán Giricz
- Pharmahungary Group, Szeged, Hungary
- MTA-se System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad Tér 4, Budapest, 1089, Hungary
| | - Javier Inserte
- Vall d'hebron Institut de Recerca (VHIR), Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - Roisin Kelly-Laubscher
- Department of Pharmacology and Therapeutics, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Attila Kiss
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Wien, Austria
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Brenda R Kwak
- Department of Pathology and Immunology, and Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Sandrine Lecour
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Gary Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
| | - Michał Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Michał Waszkiewicz
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Marta Oknińska
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Pasquale Pagliaro
- Clinical and Biological Science Department, University of Turin, Turin, Italy
| | - Bruno Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Wien, Austria
| | - Hiran A Prag
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Marisol Ruiz-Meana
- Vall d'hebron Institut de Recerca (VHIR), Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - Tamara Szabados
- Department of Pharmacology and Pharmacotherapy, Albert-Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Coert J Zuurbier
- Amsterdam UMC, Location AMC, Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A.), Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Péter Ferdinandy
- MTA-se System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad Tér 4, Budapest, 1089, Hungary.
| | - Derek J Hausenloy
- Cardiovascular and Metabolic Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore, Singapore.
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore.
- Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore.
- The Hatter Cardiovascular Institute, University College London, London, UK.
| |
Collapse
|
11
|
Flethøj M, Debes KP, Larsen C, de Blanck C, Ludvigsen TP, Kirchhoff J, Møller JE, Larsen S, Gøtze JP, Jespersen T, Olsen LH. Impact of obesity on infarct size, circulating biomarkers, mitochondrial function and mortality in a Göttingen minipig myocardial infarct model. Lab Anim (NY) 2025; 54:103-111. [PMID: 40164843 PMCID: PMC11957994 DOI: 10.1038/s41684-025-01533-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 02/25/2025] [Indexed: 04/02/2025]
Abstract
Obesity is a risk factor for the development of coronary artery disease and myocardial infarction (MI). However, most large animal studies of MI are performed in lean animals. Here we assessed the impact of obesity on echocardiographic findings, infarct size, circulating biomarkers, mitochondrial respiratory capacity and mortality in a closed-chest minipig model of MI. The initial study population consisted of 20 obese (median 60.0 kg [interquartile range 55.9-64.6 kg]) and 18 lean (25.0 kg [23.4-36.5 kg]) female Göttingen minipigs. The duration of obesity induction using a western-style diet was up to approximately 6 months (156 days [24-162 days]) before the induction of MI. The induction of MI by 120-min balloon occlusion of the left anterior descending coronary artery was feasible in 17 lean and 17 obese animals. Mortality was higher in obese compared with lean animals (53% versus 12%), driven primarily by refractory ventricular fibrillation during occlusion. Electrocardiographic findings showed longer QRS and QT intervals and more extensive ST-segment elevation in obese animals compared with lean animals during occlusion. Plasma concentrations of pro-atrial natriuretic peptide, pro-C-type natriuretic peptide and cardiac troponin T were significantly lower in obese compared with lean animals. Infarct size estimated 8 weeks after MI was significantly smaller in obese (10% [9-11%]) compared with lean animals (13% [13-15%]). Finally, mitochondrial-complex-I-linked respiratory capacity was overall significantly higher in obese animals; however, no group difference was found in intrinsic mitochondrial respiratory capacity.
Collapse
Affiliation(s)
- Mette Flethøj
- Research and Early Development, Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - Karina Poulsdóttir Debes
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Cecilie Larsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Caroline de Blanck
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Trine Pagh Ludvigsen
- Research and Early Development, Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - Jeppe Kirchhoff
- Research and Early Development, Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - Jacob Eifer Møller
- Department of Cardiology, Copenhagen University Hospital Denmark, University of Southern Denmark, Odense, Denmark
| | - Steen Larsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Jens P Gøtze
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital Denmark, Copenhagen, Denmark
| | - Thomas Jespersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lisbeth Høier Olsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark.
| |
Collapse
|
12
|
Pylova T, Elmahdy A, Krasnikova M, Jha A, Andersson EA, Kakaei Y, Shekka Espinosa A, Al-Awar A, Zulfaj E, Nejat A, Sevastianova V, Kalani M, Ryberg H, Tivesten Å, Omerovic E, Redfors B. Associations between female sex hormones, estrous cycle, ischemic preconditioning and myocardial infarct size after ischemia-reperfusion injury. Basic Res Cardiol 2025; 120:321-333. [PMID: 39945823 PMCID: PMC11976772 DOI: 10.1007/s00395-025-01099-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 04/08/2025]
Abstract
Studies on sex differences in myocardial infarction (MI) typically focus on males versus females, the exploration of hormonal physiologic variations and their impact on the infarct size remains limited. The objective of this study was to examine whether infarct size after myocardial ischemia/reperfusion injury in female rats differs in different phases of the estrous cycle, and according to the levels of sex hormones; and to assess whether the effect of ischemic preconditioning on infarct size varies in different phases of the estrous cycle and between sexes. Female rats were divided into three groups based on the estrous cycle: proestrus, estrus, and diestrus. A fourth group consisted of ovariectomized female rats. Male rats were included as a fifth group, and orchiectomized males as a sixth group. Each group underwent ischemia/reperfusion injury, with or without prior ischemic preconditioning (IPC). Plasma sex hormone levels were measured with gas chromatography-tandem mass spectrometry. Females in the proestrus showed significantly smaller infarct size compared to all other groups. Multivariable analyses identified proestrus, IPC, and estradiol as independent predictors of smaller infarct size while male sex and gonadectomy as independent predictors of larger infarct size. There was a statistical interaction between IPC and both sex and hormonal status, with a greater protective effect of IPC on infarct size in males and gonadectomized rats. After ischemia-reperfusion, proestrus female rats developed the smallest while male and gonadectomized rats the largest infarct size. Conversely, IPC conferred greater cardioprotection in male and gonadectomized rats than females in proestrus.
Collapse
Affiliation(s)
- Tetiana Pylova
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Bla Straket 5 B Wallenberglab/SU, 413 45, Gothenburg, Sweden.
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Ahmed Elmahdy
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Bla Straket 5 B Wallenberglab/SU, 413 45, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maryna Krasnikova
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Bla Straket 5 B Wallenberglab/SU, 413 45, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Abhishek Jha
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Bla Straket 5 B Wallenberglab/SU, 413 45, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Erik Axel Andersson
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Bla Straket 5 B Wallenberglab/SU, 413 45, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Yalda Kakaei
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Bla Straket 5 B Wallenberglab/SU, 413 45, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Aaron Shekka Espinosa
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Bla Straket 5 B Wallenberglab/SU, 413 45, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Amin Al-Awar
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Bla Straket 5 B Wallenberglab/SU, 413 45, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ermir Zulfaj
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Bla Straket 5 B Wallenberglab/SU, 413 45, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Amirali Nejat
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Bla Straket 5 B Wallenberglab/SU, 413 45, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Valentyna Sevastianova
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Bla Straket 5 B Wallenberglab/SU, 413 45, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Mana Kalani
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Bla Straket 5 B Wallenberglab/SU, 413 45, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Ryberg
- Department of Clinical Chemistry, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Åsa Tivesten
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Bla Straket 5 B Wallenberglab/SU, 413 45, Gothenburg, Sweden
- Department of Endocrinology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Elmir Omerovic
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Bla Straket 5 B Wallenberglab/SU, 413 45, Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Björn Redfors
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Bla Straket 5 B Wallenberglab/SU, 413 45, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| |
Collapse
|
13
|
Giordano M, Femminò S, Blua F, Boccato F, Rubeo C, Mantuano B, Cioffi F, Comità S, Brovero A, Ciullo R, Bertinaria M, Penna C, Pagliaro P. Macrophage and cardiomyocyte roles in cardioprotection: Exploiting the NLRP3 Inflammasome inhibitor INF150. Vascul Pharmacol 2025; 159:107487. [PMID: 40097083 DOI: 10.1016/j.vph.2025.107487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/03/2025] [Accepted: 03/13/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Cardiovascular diseases remain the leading cause of disability and death in the Western world. Effective cardioprotection involves limiting ischemia/reperfusion injury (IRI), including cell death (pyroptosis) driven by the NLRP3 inflammasome. While various cardiac resident cellular populations contribute to cardioprotection, it remains unclear whether targeting resident macrophages is inherently cardioprotective. Given that INF150, an NLRP3 inhibitor, exhibits varying abilities to penetrate cardiomyocytes and macrophages, we sought to address this question. METHODS We studied the cardioprotective potential of INF150, the potent metabolite of the NLRP3 inhibitor INF195, in isolated hearts or cells. In isolated hearts, we measured infarct size, caspase-1 cleavage, and interleukins (IL) release, while in macrophages, naïve H9c2 and differentiated H9c2 cells, we analyzed cell viability, and pyroptosis markers, including IL-1β release and Gasdermin D cleavage, following hypoxia/reoxygenation (H/R). RESULTS AND CONCLUSION While INF150 effectively shielded macrophages from LPS/ATP challenges, it failed to penetrate H9c2 and differentiated H9c2, even at high concentrations (no changes in pyroptosis markers induced by H/R). In the isolated mice heart model, INF150 did not demonstrate cardioprotective effects: infarct size, IL-1β, cleaved caspase-1 levels did not change significantly across tested concentrations of INF150. These findings suggest that while INF150 shows promise in macrophage/phagocytic models, its inability to penetrate cardiomyocytes limits its effectiveness in the whole cardiac tissue. Our results underscore the importance of cardiomyocyte uptake for effective cardioprotection, highlighting the need for NLRP3 inhibitors capable of targeting these cells directly. Future research should focus on enhancing the delivery and cardiomyocyte uptake of NLRP3 inhibitors to achieve cardioprotection. Unlike its precursor, INF195, which penetrates H9c2 cells, INF150 does not appear to offer cardioprotection in the whole organ.
Collapse
Affiliation(s)
- Magalì Giordano
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, TO 10043, Italy
| | - Saveria Femminò
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, TO 10043, Italy
| | - Federica Blua
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Torino, Italy
| | - Francesca Boccato
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Torino, Italy
| | - Chiara Rubeo
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, TO 10043, Italy
| | - Beatrice Mantuano
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, TO 10043, Italy
| | - Francesca Cioffi
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, TO 10043, Italy
| | - Stefano Comità
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, TO 10043, Italy
| | - Arianna Brovero
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, TO 10043, Italy
| | - Rosa Ciullo
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, TO 10043, Italy
| | - Massimo Bertinaria
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Torino, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, TO 10043, Italy; National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, TO 10043, Italy; National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy.
| |
Collapse
|
14
|
Pan QM, Bi FF, Jing ZH, Cao M, Cui C, Liu F, Jin L, Yi-Jie H, Tian H, Yu T, Yun W, Shan HL, Zhou YH. A New target of ischemic ventricular arrhythmias-ITFG2. Eur J Pharmacol 2025; 991:177301. [PMID: 39864577 DOI: 10.1016/j.ejphar.2025.177301] [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: 10/26/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
ITFG2 is an intracellular protein known to modulate the immune response of T-cells. Our previous investigation revealed that ITFG2 specifically targets ATP5b to regulate ATP energy metabolism and maintain mitochondrial function, thereby protecting the heart from ischemic injury. However, the role of ITFG2 in ischemic ventricular arrhythmias and its underlying mechanisms have not been previously reported. In this study, we found ITFG2 overexpression, induced by an adeno-associated virus serotype 9 vector, partially reduced the incidence of ischemic ventricular arrhythmias and shortened the duration of ventricular arrhythmias in mice after myocardial infarction. Conversely, shRNA-mediated knockdown of endogenous ITFG2 aggravated ischemic ventricular arrhythmias. ITFG2 overexpression also shortened the prolonged QRS complex and increased the epicardial conduction velocity in MI mice. Additionally, the hearts from ITFG2 overexpression mice exhibited a higher maximal upstroke velocity at phase 0 of transmembrane action potential compared to MI mice. Patch-clamp analyses demonstrated a 50% increase in the peak current of voltage-dependent Na+ channel by ITFG2 overexpression in isolated ventricular cardiomyocytes post MI. In cultured neonatal mouse cardiomyocytes under hypoxic conditions, ITFG2 up-regulated Nav1.5 protein expression by inhibiting its ubiquitination. Co-immunoprecipitation experiments showed that ITFG2 reduces the binding affinity between NEDD4-2 and Nav1.5, thereby inhibiting Nav1.5 ubiquitination. Taken together, our data highlight the critical role of ITFG2 in reducing susceptibility to ischemic ventricular arrhythmias by down-regulating Nav1.5 ubiquitination. These findings suggest that ITFG2 may serve as a novel target for treating ischemic ventricular arrhythmias.
Collapse
Affiliation(s)
- Qing-Ming Pan
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, 150081, PR China; Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Fang-Fang Bi
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, 150081, PR China; Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Ze-Hong Jing
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, 150081, PR China; Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Miao Cao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, 150081, PR China; Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Chen Cui
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, 150081, PR China; Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Fu Liu
- Department of Basic Medicine, Institute of Respiratory Diseases Xiamen Medical College, Xiamen Medical College, Xiamen, Fujian, 361023, PR China
| | - Li Jin
- Department of Basic Medicine, Institute of Respiratory Diseases Xiamen Medical College, Xiamen Medical College, Xiamen, Fujian, 361023, PR China; Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - He Yi-Jie
- Department of Basic Medicine, Institute of Respiratory Diseases Xiamen Medical College, Xiamen Medical College, Xiamen, Fujian, 361023, PR China; Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Hua Tian
- Department of Basic Medicine, Institute of Respiratory Diseases Xiamen Medical College, Xiamen Medical College, Xiamen, Fujian, 361023, PR China
| | - Tong Yu
- Hanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, 201620, PR China
| | - Wu Yun
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Hong-Li Shan
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, 150081, PR China; Hanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, 201620, PR China.
| | - Yu-Hong Zhou
- Department of Basic Medicine, Institute of Respiratory Diseases Xiamen Medical College, Xiamen Medical College, Xiamen, Fujian, 361023, PR China; State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, 150081, PR China.
| |
Collapse
|
15
|
Fudim M, Konecny F, Heuring JJ, Durst CA, Fain ES, Patel MR. Left Ventricular Unloading Using Intra-aortic Entrainment Pumping Before Reperfusion Reduces Post-AMI Infarct Size. J Card Fail 2025; 31:538-548. [PMID: 39147311 DOI: 10.1016/j.cardfail.2024.07.022] [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: 12/09/2023] [Revised: 07/10/2024] [Accepted: 07/18/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Anterior myocardial infarction standard of care prioritizes swift coronary reperfusion. Recent studies show left ventricular (LV) unloading with transvalvular axial flow pumps for 30 minutes before reperfusion (vs immediate reperfusion) decreases 28-day infarct size. Intra-aortic entrainment pumping, using hardware located away from the heart to provide support throughout the cardiac cycle, decreases effective systemic vascular resistance and augments visceral blood flow and pressure, and may reproduce this benefit with a decreased risk. This study characterized the hemodynamic effects of unloading before and during reperfusion using intra-aortic entrainment pumping and investigated whether unloading decreased anterior myocardial infarction scar size. METHODS AND RESULTS Yorkshire swine were subjected to 90 minutes of left anterior descending artery balloon occlusion and randomly assigned to immediate reperfusion (n = 6) vs 30 minutes unloading before reperfusion followed by 120 minutes of further unloading (n = 7). Unloading was achieved using percutaneous entrainment pumping in the descending aorta. The anterior myocardial infarction model matches that used in recent transvalvular pumping studies. Mortality before randomization was 22%. After randomization, mortality was 36% for immediate reperfusion and 0% for unloading. Unloading showed immediate hemodynamic benefit that increased through reperfusion and continued support, leading to distinct differences in cardiac function between groups after 30 minutes of reperfusion. Unloading increased stroke volume and cardiac efficiency at this timepoint relative to preocclusion baseline and reduced 28-day LV scar size by 37%-45%. CONCLUSIONS We present the first preclinical data showing extracardiac LV unloading before coronary reperfusion using intra-aortic entrainment pumping decreases 28-day infarct size. Extracardiac unloading to decrease LV scar size may provide an alternative to transvalvular pumping with potential advantages, including reduced risk.
Collapse
Affiliation(s)
- Marat Fudim
- Duke University Medical Center, Durham, North Carolina
| | - Filip Konecny
- Schulich School of Medicine & Dentistry, London, Ontario, Canada
| | | | | | | | | |
Collapse
|
16
|
Tong C, Zhou B. Cardioprotective strategies in myocardial ischemia-reperfusion injury: Implications for improving clinical translation. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2025; 11:100278. [PMID: 40182153 PMCID: PMC11967023 DOI: 10.1016/j.jmccpl.2024.100278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/11/2024] [Accepted: 12/13/2024] [Indexed: 04/05/2025]
Abstract
Ischemic heart disease is the most common cause of death and disability globally which is caused by reduced or complete cessation of blood flow to a portion of the myocardium. One of its clinical manifestations is myocardial infarction, which is commonly treated by restoring of blood flow through reperfusion therapies. However, serious ischemia-reperfusion injury (IRI) can occur, significantly undermining clinical outcomes, for which there is currently no effective therapy. This review revisits several potential pharmacological IRI intervention strategies that have entered preclinical or clinical research phases. Here, we discuss what we have learned through translational failures over the years, and propose possible ways to enhance translation efficiency.
Collapse
Affiliation(s)
- Chao Tong
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Bingying Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen 518063, China
| |
Collapse
|
17
|
Moreno-Arciniegas A, Cádiz L, Galán-Arriola C, Clemente-Moragón A, Ibáñez B. Cardioprotection strategies for anthracycline cardiotoxicity. Basic Res Cardiol 2025; 120:71-90. [PMID: 39249555 PMCID: PMC11790697 DOI: 10.1007/s00395-024-01078-6] [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: 06/11/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/10/2024]
Abstract
Thanks to the fantastic progress in cancer therapy options, there is a growing population of cancer survivors. This success has resulted in a need to focus much effort into improving the quality of life of this population. Cancer and cardiovascular disease share many common risk factors and have an interplay between them, with one condition mechanistically affecting the other and vice versa. Furthermore, widely prescribed cancer therapies have known toxic effects in the cardiovascular system. Anthracyclines are the paradigm of efficacious cancer therapy widely prescribed with a strong cardiotoxic potential. While some cancer therapies cardiovascular toxicities are transient, others are irreversible. There is a growing need to develop cardioprotective therapies that, when used in conjunction with cancer therapies, can prevent cardiovascular toxicity and thus improve long-term quality of life in survivors. The field has three main challenges: (i) identification of the ultimate mechanisms leading to cardiotoxicity to (ii) identify specific therapeutic targets, and (iii) more sensible diagnostic tools to early identify these conditions. In this review we will focus on the cardioprotective strategies tested and under investigation. We will focus this article into anthracycline cardiotoxicity since it is still the agent most widely prescribed, the one with higher toxic effects on the heart, and the most widely studied.
Collapse
Affiliation(s)
| | - Laura Cádiz
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Carlos Galán-Arriola
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Agustín Clemente-Moragón
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Borja Ibáñez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
- Cardiology Department, IIS-Fundación Jiménez Díaz University Hospital, Madrid, Spain.
| |
Collapse
|
18
|
Basalay MV, Korsak A, He Z, Gourine AV, Davidson SM, Yellon DM. SGLT2 Inhibition Induces Cardioprotection by Increasing Parasympathetic Activity. Circ Res 2025; 136:229-231. [PMID: 39690446 PMCID: PMC11741129 DOI: 10.1161/circresaha.124.324708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Affiliation(s)
- Maryna V Basalay
- The Hatter Cardiovascular Institute, University College London, United Kingdom (M.V.B., Z.H., S.M.D., D.M.Y.)
| | - Alla Korsak
- Department of Neuroscience, Physiology and Pharmacology, Centre for Cardiovascular and Metabolic Neuroscience, University College London, United Kingdom (A.K., A.V.G.)
| | - Zhenhe He
- The Hatter Cardiovascular Institute, University College London, United Kingdom (M.V.B., Z.H., S.M.D., D.M.Y.)
| | - Alexander V Gourine
- Department of Neuroscience, Physiology and Pharmacology, Centre for Cardiovascular and Metabolic Neuroscience, University College London, United Kingdom (A.K., A.V.G.)
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, United Kingdom (M.V.B., Z.H., S.M.D., D.M.Y.)
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, United Kingdom (M.V.B., Z.H., S.M.D., D.M.Y.)
| |
Collapse
|
19
|
Yusof NLM, Yellon DM, Davidson SM. Novel Selective Cardiac Myosin-Targeted Inhibitors Alleviate Myocardial Ischaemia-Reperfusion Injury. Cardiovasc Drugs Ther 2025:10.1007/s10557-024-07663-0. [PMID: 39754660 DOI: 10.1007/s10557-024-07663-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 01/06/2025]
Abstract
PURPOSE Reperfusion of the ischaemic heart is essential to limit myocardial infarction. However, reperfusion can cause cardiomyocyte hypercontracture. Recently, cardiac myosin-targeted inhibitors (CMIs), such as Mavacamten (MYK-461) and Aficamten (CK-274), have been developed to treat patients with cardiac hypercontractility. These CMIs are well tolerated and safe in clinical trials. We hypothesised that, by limiting hypercontraction, CMIs may reduce hypercontracture and protect hearts in the setting of ischaemia and reperfusion (IR). METHODS We investigated the ability of MYK-461 and CK-274 to inhibit hypercontracture of adult rat cardiomyocytes (ARVC) in vitro following ATP depletion. A suitable dose of CMIs for subsequent in vivo IR studies was identified using cardiac echocardiography of healthy male Sprague Dawley rats. Rats were anaesthetized and subject to coronary artery ligation for 30 min followed by 2 h of reperfusion. Prior to reperfusion, CMI or vehicle was administered intraperitoneally. Ischaemic preconditioning (IPC) was used as a positive control group. Infarct size was assessed by tetrazolium chloride staining and extent of hypercontracture was assessed by histological staining. RESULTS Treatment with CMIs inhibited ARVC hypercontracture in vitro. MYK-461 (2 mg/kg) and CK-274 (0.5 mg/kg to 2 mg/kg) significantly reduced infarct size vs. vehicle. IR caused extensive contraction band necrosis, which was reduced significantly by IPC but not by CMIs, likely due to assay limitations. GDC-0326, an inhibitor of PI3Kα, abrogated CK-274-mediated protection following IR injury. GDC-0326 reduced phosphorylation of AKT when administered together with CK-274. CONCLUSION This study identifies CMIs as novel cardioprotective agents in the setting of IR injury.
Collapse
Affiliation(s)
- Nur Liyana Mohammed Yusof
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000, Cheras, Kuala Lumpur, Malaysia
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK.
| |
Collapse
|
20
|
Nagy RN, Makkos A, Baranyai T, Giricz Z, Szabó M, Kravcsenko-Kiss B, Bereczki Z, Ágg B, Puskás LG, Faragó N, Schulz R, Gyöngyösi M, Lukovic D, Varga ZV, Görbe A, Ferdinandy P. Cardioprotective microRNAs (protectomiRs) in a pig model of acute myocardial infarction and cardioprotection by ischaemic conditioning: MiR-450a. Br J Pharmacol 2025; 182:396-416. [PMID: 39294819 DOI: 10.1111/bph.17313] [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: 09/27/2023] [Revised: 06/05/2024] [Accepted: 07/04/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND AND PURPOSE Cardioprotective miRNAs (protectomiRs) are promising therapeutic tools. Here, we aimed to identify protectomiRs in a translational porcine model of acute myocardial infarction (AMI) and to validate their cardiocytoprotective effect. EXPERIMENTAL APPROACH ProtectomiR candidates were selected after systematic analysis of miRNA expression changes in cardiac tissue samples from a closed-chest AMI model in pigs subjected to sham operation, AMI and ischaemic preconditioning, postconditioning or remote preconditioning, respectively. Cross-species orthologue protectomiR candidates were validated in simulated ischaemia-reperfusion injury (sI/R) model of isolated rat ocardiomyocytes and in human AC16 cells as well. For miR-450a, we performed target prediction and analysed the potential mechanisms of action by GO enrichment and KEGG pathway analysis. KEY RESULTS Out of the 220 detected miRNAs, four were up-regulated and 10 were down-regulated due to all three conditionings versus AMI. MiR-450a and miR-451 mimics at 25 nM were protective in rat cardiomyocytes, and miR-450a showed protection in human cardiomyocytes as well. MiR-450a has 3987 predicted mRNA targets in pigs, 4279 in rats and 8328 in humans. Of these, 607 genes are expressed in all three species. A total of 421 common enriched GO terms were identified in all three species, whereas KEGG pathway analysis revealed 13 common pathways. CONCLUSION AND IMPLICATIONS This is the first demonstration that miR-450a is associated with cardioprotection by ischaemic conditioning in a clinically relevant porcine model and shows cardiocytoprotective effect in human cardiomyocytes, making it a promising drug candidate. The mechanism of action of miR-450a involves multiple cardioprotective pathways. LINKED ARTICLES This article is part of a themed issue Non-coding RNA Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.2/issuetoc.
Collapse
Grants
- OTKA ANN 107803 Hungarian Scientific Research Fund
- OTKA K-105555 Hungarian Scientific Research Fund
- 2018-1.3.1-VKE-2018-00024 National Research, Development and Innovation Office
- NVKP-16-1-2016-0017 National Heart Program National Research, Development and Innovation Office
- OTKA-FK 134751 National Research, Development and Innovation Office
- TKP/ITM/NFKIH National Research, Development and Innovation Office
- OTKAK21-139105 National Research, Development and Innovation Office
- RRF-2.3.1-21-2022-00003 European Union
- EU COST Action CardioRNA.eu, Cardioprotection.eu
- 88öu1 Austrian-Hungarian Action Scholarship
- 739593 European Union's Horizon 2020
- 2019-1.1.1-PIACI-KFI-2019-00367 National Research, Development and Innovation Fund
- 2020-1.1.5-GYORSÍTÓSÁV-2021-00011 National Research, Development and Innovation Fund
- ÚNKP-20-5 National Research, Development and Innovation Fund
- ÚNKP-23-4-II-SE-34 National Research, Development and Innovation Fund
- János Bolyai Research Scholarship of Hungarian Academy of Sciences
Collapse
Affiliation(s)
- Regina N Nagy
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - András Makkos
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Tamás Baranyai
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zoltán Giricz
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Márta Szabó
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Bernadett Kravcsenko-Kiss
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zoltán Bereczki
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Bence Ágg
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - László G Puskás
- Laboratory of Functional Genomics, Biological Research Centre, Szeged, Hungary
| | - Nóra Faragó
- Laboratory of Functional Genomics, Biological Research Centre, Szeged, Hungary
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University of Giessen, Giessen, Germany
| | - Mariann Gyöngyösi
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Dominika Lukovic
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Zoltán V Varga
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary
| | - Anikó Görbe
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Péter Ferdinandy
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| |
Collapse
|
21
|
Szabados T, Makkos A, Ágg B, Benczik B, Brenner GG, Szabó M, Váradi B, Vörös I, Gömöri K, Varga ZV, Görbe A, Bencsik P, Ferdinandy P. Pharmacokinetics and cardioprotective efficacy of intravenous miR-125b* microRNA mimic in a mouse model of acute myocardial infarction. Br J Pharmacol 2025; 182:432-450. [PMID: 39472767 DOI: 10.1111/bph.17345] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 07/16/2024] [Accepted: 08/01/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND AND PURPOSE MicroRNA (miRNA) therapy is a promising approach to induce cardioprotection. We have previously identified cardiac microRNA-125b* (microRNA-125b-2-3p; miR-125b*) as a potential cardioprotective miRNA, termed ProtectomiR. We aimed to characterize the pharmacokinetics and pharmacodynamics, and the effect of miR-125b* mimic on infarct size using an in vivo mouse model. EXPERIMENTAL APPROACH To characterize the pharmacokinetics properties of miR-125b* mimic, a single injection of 10-μg miR-125b* mimic or its scramble miRNA control, or vehicle i.v. was given to C57BL/6 mice. MiR-125b* expression was measured from plasma, heart, kidney and liver samples. Effect of miR-125b* on area at risk and infarct size was assessed after 45-min coronary occlusion, followed by 24-h reperfusion; 10-μg miR-125b* mimic or 10-μg non-targeting miRNA mimic control or vehicle were administered via the right jugular vein at 10th mins of coronary occlusion. To assess molecular mechanism involved in cardioprotection, expression of mRNA targets of miR-125b* were measured from ventricular myocardium at 1, 2, 4, 8 or 24 h post-treatment using quantitative real time polymerase chain reaction. KEY RESULTS MiR-125b* expression was markedly increased in plasma and myocardium 1 h, and in the liver 2h after treatment. Infarct size was significantly reduced after miR-125b* mimic treatment when compared to the vehicle. The expression of Ccna2, Eef2k and Cacnb2 target mRNAs was significantly reduced 8 h after injection of miR-125b* mimic. CONCLUSION AND IMPLICATIONS This is the first demonstration of pharmacokinetic and molecular pharmacodynamic properties as well as the cardioprotective effect of miR-125b* mimic in vivo. LINKED ARTICLES This article is part of a themed issue Non-coding RNA Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.2/issuetoc.
Collapse
Affiliation(s)
- Tamara Szabados
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - András Makkos
- Pharmahungary Group, Szeged, Hungary
- Cardiometabolic and HUN-REN-SU and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, and Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Bence Ágg
- Pharmahungary Group, Szeged, Hungary
- Cardiometabolic and HUN-REN-SU and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, and Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Bettina Benczik
- Pharmahungary Group, Szeged, Hungary
- Cardiometabolic and HUN-REN-SU and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, and Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Gábor G Brenner
- Cardiometabolic and HUN-REN-SU and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, and Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Márta Szabó
- Cardiometabolic and HUN-REN-SU and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, and Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Barnabás Váradi
- Cardiometabolic and HUN-REN-SU and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, and Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Imre Vörös
- Cardiometabolic and HUN-REN-SU and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, and Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Kamilla Gömöri
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Zoltán V Varga
- Cardiometabolic and HUN-REN-SU and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, and Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Anikó Görbe
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Pharmahungary Group, Szeged, Hungary
- Cardiometabolic and HUN-REN-SU and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, and Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Péter Bencsik
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Péter Ferdinandy
- Pharmahungary Group, Szeged, Hungary
- Cardiometabolic and HUN-REN-SU and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, and Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| |
Collapse
|
22
|
Nikolaou PE, Konijnenberg LS, Kostopoulos IV, Miliotis M, Mylonas N, Georgoulis A, Pavlidis G, Kuster CT, van Reijmersdal VP, Luiken TT, Agapaki A, Roverts R, Orologas N, Grigoriadis D, Pallot G, Boucher P, Kostomitsopoulos N, Pieper MP, Germain S, Loukas Y, Dotsikas Y, Ikonomidis I, Hatzigeorgiou AG, Tsitsilonis O, Zuurbier CJ, Nijveldt R, van Royen N, Andreadou I. Empagliflozin in Acute Myocardial Infarction Reduces No-Reflow and Preserves Cardiac Function by Preventing Endothelial Damage. JACC Basic Transl Sci 2025; 10:43-61. [PMID: 39958474 PMCID: PMC11830260 DOI: 10.1016/j.jacbts.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 02/18/2025]
Abstract
Empagliflozin treatment before acute myocardial infarction mainly targets the endothelial cell transcriptome. Empagliflozin treatment before and after myocardial infarction decreased no reflow and microvascular injury, leading to reduced infiltration of inflammatory cells, reduced infarct size, and improved cardiac function in mice. In diabetic patients receiving empagliflozin after myocardial infarction, perfused boundary region, flow-mediated dilation, and global longitudinal strain were improved.
Collapse
Affiliation(s)
- Panagiota Efstathia Nikolaou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
- Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Ioannis V. Kostopoulos
- Section of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Marios Miliotis
- DIANA-Lab, Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
| | - Nikolaos Mylonas
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasios Georgoulis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - George Pavlidis
- Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Carolien T.A. Kuster
- Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Tom T.J. Luiken
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Anna Agapaki
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Rona Roverts
- Electron Microscopy Center, Radboud UMC Technology Center, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Nikolaos Orologas
- Section of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitris Grigoriadis
- DIANA-Lab, Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
| | - Gaëtan Pallot
- Center for Interdisciplinary Research in Biology, College de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Pierre Boucher
- Center for Interdisciplinary Research in Biology, College de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Nikolaos Kostomitsopoulos
- Laboratory Animal Facilities, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | | - Stéphane Germain
- Center for Interdisciplinary Research in Biology, College de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Yannis Loukas
- Laboratory of Pharmaceutical Analysis, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Yannis Dotsikas
- Laboratory of Pharmaceutical Analysis, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Ignatios Ikonomidis
- Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Artemis G. Hatzigeorgiou
- DIANA-Lab, Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
| | - Ourania Tsitsilonis
- Section of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Coert J. Zuurbier
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, the Netherlands
| | - Robin Nijveldt
- Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Niels van Royen
- Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
23
|
Braczko F, Skyschally A, Lieder H, Kather JN, Kleinbongard P, Heusch G. Deep learning segmentation model for quantification of infarct size in pigs with myocardial ischemia/reperfusion. Basic Res Cardiol 2024; 119:923-936. [PMID: 39348000 PMCID: PMC11628591 DOI: 10.1007/s00395-024-01081-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/05/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
Infarct size (IS) is the most robust end point for evaluating the success of preclinical studies on cardioprotection. The gold standard for IS quantification in ischemia/reperfusion (I/R) experiments is triphenyl tetrazolium chloride (TTC) staining, typically done manually. This study aimed to determine if automation through deep learning segmentation is a time-saving and valid alternative to standard IS quantification. High-resolution images from TTC-stained, macroscopic heart slices were retrospectively collected from pig experiments (n = 390) with I/R without/with cardioprotection to cover a wide IS range. Existing IS data from pig experiments, quantified using a standard method of manual and subsequent digital labeling of film-scan annotations, were used as reference. To automate the evaluation process with the aim to be more objective and save time, a deep learning pipeline was implemented; the collected images (n = 3869) were pre-processed by cropping and labeled (image annotations). To ensure their usability as training data for a deep learning segmentation model, IS was quantified from image annotations and compared to IS quantified using the existing film-scan annotations. A supervised deep learning segmentation model based on dynamic U-Net architecture was developed and trained. The evaluation of the trained model was performed by fivefold cross-validation (n = 220 experiments) and testing on an independent test set (n = 170 experiments). Performance metrics (Dice similarity coefficient [DSC], pixel accuracy [ACC], average precision [mAP]) were calculated. IS was then quantified from predictions and compared to IS quantified from image annotations (linear regression, Pearson's r; analysis of covariance; Bland-Altman plots). Performance metrics near 1 indicated a strong model performance on cross-validated data (DSC: 0.90, ACC: 0.98, mAP: 0.90) and on the test set data (DSC: 0.89, ACC: 0.98, mAP: 0.93). IS quantified from predictions correlated well with IS quantified from image annotations in all data sets (cross-validation: r = 0.98; test data set: r = 0.95) and analysis of covariance identified no significant differences. The model reduced the IS quantification time per experiment from approximately 90 min to 20 s. The model was further tested on a preliminary test set from experiments in isolated, saline-perfused rat hearts with regional I/R without/with cardioprotection (n = 27). There was also no significant difference in IS between image annotations and predictions, but the performance on the test set data from rat hearts was lower (DSC: 0.66, ACC: 0.91, mAP: 0.65). IS quantification using a deep learning segmentation model is a valid and time-efficient alternative to manual and subsequent digital labeling.
Collapse
Affiliation(s)
- Felix Braczko
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Hufelandstr. 55, 45122, Essen, Germany
| | - Andreas Skyschally
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Hufelandstr. 55, 45122, Essen, Germany
| | - Helmut Lieder
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Hufelandstr. 55, 45122, Essen, Germany
| | - Jakob Nikolas Kather
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Hufelandstr. 55, 45122, Essen, Germany
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Hufelandstr. 55, 45122, Essen, Germany.
| |
Collapse
|
24
|
Buja LM. Pathobiology of myocardial and cardiomyocyte injury in ischemic heart disease: Perspective from seventy years of cell injury research. Exp Mol Pathol 2024; 140:104944. [PMID: 39577392 DOI: 10.1016/j.yexmp.2024.104944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/11/2024] [Accepted: 11/08/2024] [Indexed: 11/24/2024]
Abstract
This review presents a perspective on the pathobiology of acute myocardial infarction, a major manifestation of ischemic heart disease, and related mechanisms of ischemic and toxic cardiomyocyte injury, based on advances and insights that have accrued over the last seventy years, including my sixty years of involvement in the field as a physician-scientist-pathologist. This analysis is based on integration of my research within the broader context of research in the field. A particular focus has been on direct measurements in cardiomyocytes of electrolyte content by electron probe X-ray microanalysis (EPXMA) and Ca2+ fluxes by fura-2 microspectrofluorometry. These studies established that increased intracellular Ca2+ develops at a transitional stage in the progression of cardiomyocyte injury in association with ATP depletion, other electrolyte alterations, altered cell volume regulation, and altered membrane phospholipid composition. Subsequent increase in total calcium with mitochondrial calcium accumulation can occur. These alterations are characteristic of oncosis, which is an initial pre-lethal state of cell injury with cell swelling due to cell membrane dysfunction in ATP depleted cells; oncosis rapidly progresses to necrosis/necroptosis with physical disruption of the cell membrane, unless the adverse stimulus is rapidly reversed. The observed sequential changes fit a three-stage model of membrane injury leading to irreversible cell injury. The data establish oncosis as the primary mode of cardiomyocyte injury in evolving myocardial infarcts. Oncosis also has been documented to be the typical form of non-ischemic cell injury due to toxins. Cardiomyocytes with less energy impairment have the capability of undergoing apoptosis and autophagic death as well as oncosis, as is seen in pathological remodeling in chronic heart failure. Work is ongoing to apply the insights from experimental studies to better understand and ameliorate myocardial ischemia and reperfusion injury in patients. The perspective and insights in this review are derived from basic principles of pathology, an integrative discipline focused on mechanisms of disease affecting the cell, the organizing unit of living organisms.
Collapse
Affiliation(s)
- L Maximilian Buja
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth-Houston), Houston, TX, United States of America.
| |
Collapse
|
25
|
Bosa Ojeda F, Méndez Vargas C, Lacalzada Almeida J, Izquierdo Gómez MM, Jiménez Sosa A, Rodríguez Jiménez C, Sánchez‐Grande Flecha A, Bosa Santana M, Yanes Bowden G. Efficacy and safety of levosimendan in patients with ST-segment elevation myocardial infarction undergoing primary percutaneous coronary intervention: The LEVOCEST trial. Catheter Cardiovasc Interv 2024; 104:1414-1422. [PMID: 39425551 PMCID: PMC11667407 DOI: 10.1002/ccd.31267] [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: 08/09/2024] [Revised: 09/30/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Primary angioplasty is the standard procedure for patients with ST-segment elevation myocardial infarction (STEMI). However, myocardial reperfusion results in additional cell damage. Levosimendan, due to its pleiotropic effects, may be a therapeutic alternative to prevent this damage. The objective of this study was to evaluate whether this drug can reduce infarct size in patients with STEMI. METHODS Patients were randomized to receive a 24-h infusion of either levosimendan (0.1 μg/kg/min) or placebo after the primary angioplasty. The main objective was to assess the size of the infarct by cardiac resonance at 30 days and 6 months after the event. Other variables such as left ventricular ejection fraction (LVEF) and adverse ventricular remodeling (AVR) were assessed by speckle-tracking echocardiography and magnetic resonance. Major adverse cardiovascular events (MACE) were also collected. RESULTS 157 patients were analysed (levosimendan, n = 79; placebo, n = 78). We found that after 6 months, patients treated with levosimendan had a greater reduction in infarct size (13.19% ± 9.5% vs.11.79% ± 9%, p = 0.001), compared with those in the placebo group (13.35% ± 7.1% vs. 13.43% ± 7.8%, p = 0.38). There were no significant differences in MACE between both groups. CONCLUSIONS Levosimendan is a safe and effective therapeutic option for reducing infarct size in patients with STEMI.
Collapse
Affiliation(s)
- Francisco Bosa Ojeda
- Department of Interventional CardiologyUniversity Hospital of the Canary IslandsLa Laguna, Santa Cruz de TenerifeCanary IslandsSpain
- Department of CardiologyUniversity Hospital of the Canary IslandsLa Laguna, Santa Cruz de TenerifeCanary IslandsSpain
| | - Corabel Méndez Vargas
- Department of Interventional CardiologyUniversity Hospital of the Canary IslandsLa Laguna, Santa Cruz de TenerifeCanary IslandsSpain
- Department of CardiologyUniversity Hospital of the Canary IslandsLa Laguna, Santa Cruz de TenerifeCanary IslandsSpain
| | - Juan Lacalzada Almeida
- Department of CardiologyUniversity Hospital of the Canary IslandsLa Laguna, Santa Cruz de TenerifeCanary IslandsSpain
| | - María M. Izquierdo Gómez
- Department of CardiologyUniversity Hospital of the Canary IslandsLa Laguna, Santa Cruz de TenerifeCanary IslandsSpain
| | - Alejandro Jiménez Sosa
- Research UnitUniversity Hospital of the Canary IslandsLa Laguna, Santa Cruz de TenerifeCanary IslandsSpain
| | - Consuelo Rodríguez Jiménez
- Clinical Pharmacology ServiceClinical Research and Clinical Trials Unit of the University Hospital of the Canary IslandsCanary IslandsSpain
| | - Alejandro Sánchez‐Grande Flecha
- Department of Interventional CardiologyUniversity Hospital of the Canary IslandsLa Laguna, Santa Cruz de TenerifeCanary IslandsSpain
- Department of CardiologyUniversity Hospital of the Canary IslandsLa Laguna, Santa Cruz de TenerifeCanary IslandsSpain
| | - Marta Bosa Santana
- Emergency DepartmentUniversity Hospital Nuestra Señora de CandelariaCanary IslandsSpain
| | - Geoffrey Yanes Bowden
- Department of Interventional CardiologyUniversity Hospital of the Canary IslandsLa Laguna, Santa Cruz de TenerifeCanary IslandsSpain
- Department of CardiologyUniversity Hospital of the Canary IslandsLa Laguna, Santa Cruz de TenerifeCanary IslandsSpain
| |
Collapse
|
26
|
Bolli R, Tang XL. CAESAR lives on with IMPACT: bringing rigor and relevance to cardioprotection research. Basic Res Cardiol 2024; 119:889-892. [PMID: 39422733 DOI: 10.1007/s00395-024-01082-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 09/27/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024]
Affiliation(s)
- Roberto Bolli
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY, 40202, USA.
| | - Xian-Liang Tang
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY, 40202, USA
| |
Collapse
|
27
|
Kleinbongard P, Arriola CG, Badimon L, Crisostomo V, Giricz Z, Gyöngyösi M, Heusch G, Ibanez B, Kiss A, de Kleijn DPV, Podesser BK, Carracedo RR, Rodríguez-Sinovas A, Ruiz-Meana M, Sanchez Margallo FM, Vilahur G, Zamorano JL, Zaragoza C, Ferdinandy P, Hausenloy DJ. The IMproving Preclinical Assessment of Cardioprotective Therapies (IMPACT): multicenter pig study on the effect of ischemic preconditioning. Basic Res Cardiol 2024; 119:893-909. [PMID: 39422732 PMCID: PMC11628588 DOI: 10.1007/s00395-024-01083-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 10/19/2024]
Abstract
Numerous cardioprotective interventions have been reported to reduce myocardial infarct size (IS) in pre-clinical studies. However, their translation for the benefit of patients with acute myocardial infarction (AMI) has been largely disappointing. One reason for the lack of translation is the lack of rigor and reproducibility in pre-clinical studies. To address this, we have established the European IMproving Preclinical Assessment of Cardioprotective Therapies (IMPACT) pig AMI network with centralized randomization and blinded core laboratory IS analysis and validated the network with ischemic preconditioning (IPC) as a positive control. Ten sites in the COST Innovators Grant (IG16225) network participated in the IMPACT network. Three sites were excluded from the final analysis through quality control of infarct images and use of pre-defined exclusion criteria. Using a centrally generated randomization list, pigs were allocated to myocardial ischemia/reperfusion (I/R, N = 5/site) or IPC + I/R (N = 5/site). The primary endpoint was IS [% area-at-risk (AAR)], as quantified by triphenyl-tetrazolium-chloride (TTC) staining in a centralized, blinded core laboratory (5 sites), or IS [% left-ventricular mass (LV)], as quantified by a centralized, blinded cardiac magnetic resonance (CMR) core laboratory (2 sites). In pooled analyses, IPC significantly reduced IS when compared to I/R (57 ± 14 versus 32 ± 19 [%AAR] N = 25 pigs/group; p < 0.001; 25 ± 13 versus 14 ± 8 [%LV]; N = 10 pigs/group; p = 0.021). In site-specific analyses, in 4 of the 5 sites, IS was significantly reduced by IPC when compared to I/R when quantified by TTC and in 1 of 2 sites when quantified by CMR. A pig AMI multicenter European network with centralized randomization and core blinded IS analysis was established and validated with the aim to improve the reproducibility of cardioprotective interventions in pre-clinical studies and the translation of cardioprotection for patient benefit.
Collapse
Affiliation(s)
- Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, University of Duisburg-Essen, Hufelandstr. 55, 45122, Essen, Germany.
| | - Carlos Galán Arriola
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, CIBER de Enfermedades Cardiovasculares (CIBERCV), Melchor Fernández Almagro 9, 28029, Madrid, Spain.
| | - Lina Badimon
- Research Institute Hospital de La Santa Creu I Sant Pau-IIB Sant Pau, and CIBER Enfermedades Cardiovasculares, Barcelona, Spain
| | - Veronica Crisostomo
- Cardiovascular Area, Jesús Usón Minimally Invasive Surgery Centre (CCMIJU), Cáceres, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), RICORS-TERAV Network, ISCIII, Madrid, Spain
| | - Zoltán Giricz
- Cardiovascular and Metabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Mariann Gyöngyösi
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090, Vienna, Austria
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, University of Duisburg-Essen, Hufelandstr. 55, 45122, Essen, Germany
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Attila Kiss
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | | | - Bruno K Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Rafael Ramírez Carracedo
- Unidad de Investigación Cardiovascular, Departamento de Cardiología, Hospital Ramón y Cajal (IRYCIS), Universidad Francisco de Vitoria, Madrid, Spain
| | - Antonio Rodríguez-Sinovas
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Marisol Ruiz-Meana
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Francisco M Sanchez Margallo
- CIBER de Enfermedades Cardiovasculares (CIBERCV), RICORS-TERAV Network, ISCIII, Madrid, Spain
- Jesús Usón Minimally Invasive Surgery Centre (CCMIJU), Cáceres, Spain
| | - Gemma Vilahur
- Research Institute Hospital de La Santa Creu I Sant Pau-IIB Sant Pau, and CIBER Enfermedades Cardiovasculares, Barcelona, Spain
| | | | - Carlos Zaragoza
- Unidad de Investigación Cardiovascular, Departamento de Cardiología, Hospital Ramón y Cajal (IRYCIS), Universidad Francisco de Vitoria, Madrid, Spain
| | - Peter Ferdinandy
- Pharmahungary Group, Szeged, Hungary.
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad Tér 4, Budapest, 1089, Hungary.
- Center for Pharmacology and Drug Research and Development, Semmelweis University, Budapest, Hungary.
| | - Derek J Hausenloy
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore.
- National Heart Research Institute Singapore, National Heart Centre, Singapore, Singapore.
- Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore.
- The Hatter Cardiovascular Institute, University College London, London, UK.
| |
Collapse
|
28
|
Liu F, Lv LF, Bi FF, Pan QM, Jing ZH, Cui C, Cao M, Yu T, Li J, He YJ, Xiao HW, Tian H, Wu Y, Shan HL, Zhou YH. ITFG2 as a NEDD4-2 inhibitor: Preserving calcium homeostasis to prevent myocardial ischemic injury. Biochem Pharmacol 2024; 230:116597. [PMID: 39477020 DOI: 10.1016/j.bcp.2024.116597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/15/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
This study aimed to investigate the role of ITFG2, a protein highly expressed in cardiac tissues, in myocardial ischemic injury and its potential interactions with NEDD4-2. An in vivo myocardial infarction (MI) model was induced in mice via left anterior descending artery ligation, and ITFG2 expression was modulated using adeno-associated virus AAV9 vectors. Echocardiography was used to assess cardiac function, and primary mouse cardiomyocytes were cultured and subjected to hypoxia. ITFG2 expression was found to be significantly reduced following MI and in hypoxia-treated neonatal cardiomyocytes. Overexpression of ITFG2 improved cardiac contractility, reduced apoptosis, and stabilized calcium levels by inhibiting NEDD4-2-mediated ubiquitination of SERCA2a. Conversely, ITFG2 knockdown exacerbated calcium overload and cardiac dysfunction. Mechanistically, ITFG2 binds to NEDD4-2, decreasing its interaction with SERCA2a and preventing SERCA2a degradation. These findings suggest that ITFG2 acts as a critical inhibitor of NEDD4-2, preserving SERCA2a function and maintaining calcium homeostasis in cardiomyocytes under ischemic conditions. Therefore, ITFG2 may represent a potential therapeutic target for preventing myocardial ischemic injury and improving outcomes in MI patients.
Collapse
Affiliation(s)
- Fu Liu
- Department of Basic Medicine, Institute of Respiratory Diseases Xiamen Medical College of Respiratory Diseases, Xiamen Medical College, Xiamen, Fujian 361023, PR China
| | - Li-Fang Lv
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150081, PR China; Department of Physiology, Department of Basic Medicine, Harbin Medical University, Harbin, Heilongjiang 150081, PR China
| | - Fang-Fang Bi
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150081, PR China
| | - Qing-Ming Pan
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150081, PR China
| | - Ze-Hong Jing
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150081, PR China
| | - Chen Cui
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150081, PR China
| | - Miao Cao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150081, PR China
| | - Tong Yu
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, 201620, PR China
| | - Jin Li
- Department of Basic Medicine, Institute of Respiratory Diseases Xiamen Medical College of Respiratory Diseases, Xiamen Medical College, Xiamen, Fujian 361023, PR China; Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, PR China
| | - Yi-Jie He
- Department of Basic Medicine, Institute of Respiratory Diseases Xiamen Medical College of Respiratory Diseases, Xiamen Medical College, Xiamen, Fujian 361023, PR China; Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, PR China
| | - Hong-Wen Xiao
- Department of Basic Medicine, Institute of Respiratory Diseases Xiamen Medical College of Respiratory Diseases, Xiamen Medical College, Xiamen, Fujian 361023, PR China
| | - Hua Tian
- Department of Basic Medicine, Institute of Respiratory Diseases Xiamen Medical College of Respiratory Diseases, Xiamen Medical College, Xiamen, Fujian 361023, PR China
| | - Yun Wu
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, PR China
| | - Hong-Li Shan
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, 201620, PR China.
| | - Yu-Hong Zhou
- Department of Basic Medicine, Institute of Respiratory Diseases Xiamen Medical College of Respiratory Diseases, Xiamen Medical College, Xiamen, Fujian 361023, PR China.
| |
Collapse
|
29
|
Erkens R, Duse DA, Brum A, Chadt A, Becher S, Siragusa M, Quast C, Müssig J, Roden M, Cortese-Krott M, Ibáñez B, Lammert E, Fleming I, Jung C, Al-Hasani H, Heusch G, Kelm M. Inhibition of proline-rich tyrosine kinase 2 restores cardioprotection by remote ischaemic preconditioning in type 2 diabetes. Br J Pharmacol 2024; 181:4174-4194. [PMID: 38956895 DOI: 10.1111/bph.16483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/26/2024] [Accepted: 05/24/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND AND PURPOSE Remote ischaemic preconditioning (rIPC) for cardioprotection is severely impaired in diabetes, and therapeutic options to restore it are lacking. The vascular endothelium plays a key role in rIPC. Given that the activity of endothelial nitric oxide synthase (eNOS) is inhibited by proline-rich tyrosine kinase 2 (Pyk2), we hypothesized that pharmacological Pyk2 inhibition could restore eNOS activity and thus restore remote cardioprotection in diabetes. EXPERIMENTAL APPROACH New Zealand obese (NZO) mice that demonstrated key features of diabetes were studied. The consequence of Pyk2 inhibition on endothelial function, rIPC and infarct size after myocardial infarction were evaluated. The impact of plasma from mice and humans with or without diabetes was assessed in isolated buffer perfused murine hearts and aortic rings. KEY RESULTS Plasma from nondiabetic mice and humans, both subjected to rIPC, caused remote tissue protection. Similar to diabetic humans, NZO mice demonstrated endothelial dysfunction. NZO mice had reduced circulating nitrite levels, elevated arterial blood pressure and a larger infarct size after ischaemia and reperfusion than BL6 mice. Pyk2 increased the phosphorylation of eNOS at its inhibitory site (Tyr656), limiting its activity in diabetes. The cardioprotective effects of rIPC were abolished in diabetic NZO mice. Pharmacological Pyk2 inhibition restored endothelial function and rescued cardioprotective effects of rIPC. CONCLUSION AND IMPLICATIONS Endothelial function and remote tissue protection are impaired in diabetes. Pyk2 is a novel target for treating endothelial dysfunction and restoring cardioprotection through rIPC in diabetes.
Collapse
Affiliation(s)
- Ralf Erkens
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Dragos Andrei Duse
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Amanda Brum
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Alexandra Chadt
- Institute for Clinical Biochemistry and Pathobiochemistry, Deutsches Diabetes Zentrum at Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Partner Duesseldorf, Neuherberg, Germany
| | - Stefanie Becher
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Mauro Siragusa
- Center for Molecular Medicine, Institute for Vascular Signalling, Goethe University Frankfurt, Frankfurt, Germany
- German Centre for Cardiovascular Research, Partner site RhineMain, Frankfurt, Germany
| | - Christine Quast
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Johanna Müssig
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Michael Roden
- German Center for Diabetes Research (DZD e.V.), Partner Duesseldorf, Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty, Heinrich-Heine University and University Hospital Duesseldorf, Duesseldorf, Germany
- Institute for Clinical Diabetology, Deutsches Diabetes Zentrum at Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Miriam Cortese-Krott
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
- CARID Cardiovascular Research Institute Duesseldorf, Duesseldorf, Germany
| | - Borja Ibáñez
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Eckhard Lammert
- German Center for Diabetes Research (DZD e.V.), Partner Duesseldorf, Neuherberg, Germany
- Institute of Metabolic Physiology, Heinrich-Heine University, Duesseldorf, Germany
| | - Ingrid Fleming
- Center for Molecular Medicine, Institute for Vascular Signalling, Goethe University Frankfurt, Frankfurt, Germany
- German Centre for Cardiovascular Research, Partner site RhineMain, Frankfurt, Germany
| | - Christian Jung
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Hadi Al-Hasani
- Institute for Clinical Biochemistry and Pathobiochemistry, Deutsches Diabetes Zentrum at Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Partner Duesseldorf, Neuherberg, Germany
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University School of Medicine Essen, Essen, Germany
| | - Malte Kelm
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
- CARID Cardiovascular Research Institute Duesseldorf, Duesseldorf, Germany
| |
Collapse
|
30
|
Jin Y, Tan M, Yin Y, Lin C, Zhao Y, Zhang J, Jiang T, Li H, He M. Oroxylin A alleviates myocardial ischemia–reperfusion injury by quelling ferroptosis via activating the DUSP10/ MAPK‐Nrf2 pathway. Phytother Res 2024; 38:5290-5308. [DOI: pmid:39225191 doi:10.1002/ptr.8315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 07/26/2024] [Indexed: 06/06/2025]
Abstract
AbstractReperfusion therapy is the primary treatment strategy for acute myocardial infarction (AMI). Paradoxically, it can lead to myocardial damage, namely myocardial ischemia/reperfusion injury (MIRI). This study explored whether oroxylin A (OA) protects the myocardium after MIRI by inhibiting ferroptosis and the underlying mechanism. In vivo, we established an MIRI model to investigate the protective effect of OA. In vitro, H9C2 cells were used to explore the regulation of ferroptosis by OA through immunofluorescence staining, western blotting, assay kits, etc. Additionally, RNA sequencing analysis (RNA‐seq) and network pharmacology analyses were conducted to elucidate the molecular mechanisms. Our results showed that MIRI caused cardiac structural and functional damage in rats. MIRI promoted ferroptosis, which was consistently observed in vitro. However, pretreatment with OA reversed these effects. The mitogen‐activated protein kinases (MAPK) signaling pathway participated in the MIRI process, with dual‐specificity phosphatase 10 (DUSP10) found to regulate it. Further confirmation was provided by knocking down DUSP10 using small interfering RNA (siRNA), demonstrating the activation of the DUSP10/MAPK‐Nrf2 pathway by OA to protect H9C2 cells from ferroptosis. Our research has demonstrated the mitigating effect of OA on MIRI and the improvement of myocardial function for the first time. The inhibition of ferroptosis has been identified as one of the mechanisms through which OA exerts its myocardial protective effects. Moreover, we have first unveiled that DUSP10 serves as an upstream target involved in mediating ferroptosis, and the regulation of the DUSP10/MAPK‐Nrf2 pathway by OA is crucial in inhibiting ferroptosis to protect the myocardium.
Collapse
Affiliation(s)
- Yifeng Jin
- Department of Cardiology The First Affiliated Hospital of Soochow University Suzhou Jiangsu P. R. China
- Department of General Practice The First Affiliated Hospital of Soochow University Suzhou Jiangsu P. R. China
| | - Mingyue Tan
- Department of Cardiology The First Affiliated Hospital of Soochow University Suzhou Jiangsu P. R. China
- Department of Geriatrics, Southwest Hospital The Third Military Medical University (Army Medical University) Chongqing P. R. China
| | - Yunfei Yin
- Department of Cardiology The First Affiliated Hospital of Soochow University Suzhou Jiangsu P. R. China
| | - Chen Lin
- Jinjihu Business District Squadron Suzhou Industrial Park Food and Drug Safety Inspection Team Suzhou Jiangsu P. R. China
| | - Yongjian Zhao
- Department of Cardiology The First Affiliated Hospital of Soochow University Suzhou Jiangsu P. R. China
| | - Jun Zhang
- Department of Cardiology The First Affiliated Hospital of Soochow University Suzhou Jiangsu P. R. China
| | - Tingbo Jiang
- Department of Cardiology The First Affiliated Hospital of Soochow University Suzhou Jiangsu P. R. China
| | - Hongxia Li
- Department of Cardiology The First Affiliated Hospital of Soochow University Suzhou Jiangsu P. R. China
| | - Mingqing He
- Department of Gerontology The First Affiliated Hospital of Soochow University Suzhou Jiangsu P. R. China
| |
Collapse
|
31
|
Jin Y, Tan M, Yin Y, Lin C, Zhao Y, Zhang J, Jiang T, Li H, He M. Oroxylin A alleviates myocardial ischemia-reperfusion injury by quelling ferroptosis via activating the DUSP10/MAPK-Nrf2 pathway. Phytother Res 2024; 38:5290-5308. [PMID: 39225191 DOI: 10.1002/ptr.8315] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 06/30/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
Reperfusion therapy is the primary treatment strategy for acute myocardial infarction (AMI). Paradoxically, it can lead to myocardial damage, namely myocardial ischemia/reperfusion injury (MIRI). This study explored whether oroxylin A (OA) protects the myocardium after MIRI by inhibiting ferroptosis and the underlying mechanism. In vivo, we established an MIRI model to investigate the protective effect of OA. In vitro, H9C2 cells were used to explore the regulation of ferroptosis by OA through immunofluorescence staining, western blotting, assay kits, etc. Additionally, RNA sequencing analysis (RNA-seq) and network pharmacology analyses were conducted to elucidate the molecular mechanisms. Our results showed that MIRI caused cardiac structural and functional damage in rats. MIRI promoted ferroptosis, which was consistently observed in vitro. However, pretreatment with OA reversed these effects. The mitogen-activated protein kinases (MAPK) signaling pathway participated in the MIRI process, with dual-specificity phosphatase 10 (DUSP10) found to regulate it. Further confirmation was provided by knocking down DUSP10 using small interfering RNA (siRNA), demonstrating the activation of the DUSP10/MAPK-Nrf2 pathway by OA to protect H9C2 cells from ferroptosis. Our research has demonstrated the mitigating effect of OA on MIRI and the improvement of myocardial function for the first time. The inhibition of ferroptosis has been identified as one of the mechanisms through which OA exerts its myocardial protective effects. Moreover, we have first unveiled that DUSP10 serves as an upstream target involved in mediating ferroptosis, and the regulation of the DUSP10/MAPK-Nrf2 pathway by OA is crucial in inhibiting ferroptosis to protect the myocardium.
Collapse
Affiliation(s)
- Yifeng Jin
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
- Department of General Practice, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
| | - Mingyue Tan
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
- Department of Geriatrics, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
| | - Yunfei Yin
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
| | - Chen Lin
- Jinjihu Business District Squadron, Suzhou Industrial Park Food and Drug Safety Inspection Team, Suzhou, Jiangsu, P. R. China
| | - Yongjian Zhao
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
| | - Jun Zhang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
| | - Tingbo Jiang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
| | - Hongxia Li
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
| | - Mingqing He
- Department of Gerontology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
| |
Collapse
|
32
|
Christodoulou A, Nikolaou PE, Symeonidi L, Katogiannis K, Pechlivani L, Nikou T, Varela A, Chania C, Zerikiotis S, Efentakis P, Vlachodimitropoulos D, Katsoulas N, Agapaki A, Dimitriou C, Tsoumani M, Kostomitsopoulos N, Davos CH, Skaltsounis AL, Tselepis A, Halabalaki M, Tseti I, Iliodromitis EK, Ikonomidis I, Andreadou I. Cardioprotective potential of oleuropein, hydroxytyrosol, oleocanthal and their combination: Unravelling complementary effects on acute myocardial infarction and metabolic syndrome. Redox Biol 2024; 76:103311. [PMID: 39153251 PMCID: PMC11378258 DOI: 10.1016/j.redox.2024.103311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024] Open
Abstract
Clinical studies have previously established the role of olive products in cardiovascular disease (CVD) prevention, whilst the identification of the responsible constituents for the beneficial effects is still pending. We sought to assess and compare the cardioprotective potential of oleuropein (OL), hydroxytyrosol (HT), oleocanthal (OC) and oleanolic Acid (OA), regarding Ischemia/Reperfusion Injury (IRI) and CVD risk factors alleviation. The scope of the study was to design a potent and safe combinatorial therapy for high-cardiovascular-risk patients on a bench-to-bedside approach. We evaluated the IRI-limiting potential of 6-weeks treatment with OL, HT, OC or OA at nutritional doses, in healthy and metabolic syndrome (MS)-burdened mice. Three combinatorial regimens were designed and the mixture with preponderant benefits (OL-HT-OC, Combo 2), including infarct sparing and antiglycemic potency, compared to the isolated compounds, was further investigated for its anti-atherosclerotic effects. In vivo experiments revealed that the combination regimen of Combo 2 presented the most favorable effects in limiting infarct size and hyperglycemia, which was selected to be further investigated in the clinical setting in Chronic Coronary Artery Syndrome (CCAS) patients. Cardiac function, inflammation markers and oxidative stress were assessed at baseline and after 4 weeks of treatment with the OL-HT-OC supplement in the clinical study. We found that OL, OC and OA significantly reduced infarct size in vivo compared to Controls. OL exhibited antihyperglycemic properties and OA attenuated hypercholesterolemia. OL-HT-OA, OL-HT-OC and OL-HT-OC-OA combination regimens were cardioprotective, whereas only OL-HT-OC mitigated hyperglycemia. Combo 2 cardioprotection was attributed to apoptosis suppression, enhanced antioxidant effects and upregulation of antioxidant enzymes. Additionally, it reduced atherosclerotic plaque extent in vivo. OL-HT-OC supplement ameliorated cardiac, vascular and endothelial function in the small-scale clinical study. Conclusively, OL-HT-OC combination therapy exerts potent cardioprotective, antihyperglycemic and anti-atherosclerotic properties in vivo, with remarkable and clinically translatable cardiovascular benefits in high-risk patients.
Collapse
Affiliation(s)
- Andriana Christodoulou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou, Athens, Greece
| | - Panagiota-Efstathia Nikolaou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou, Athens, Greece
| | - Lydia Symeonidi
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou, Athens, Greece
| | - Konstantinos Katogiannis
- Laboratory of Echocardiography and Preventive Cardiology, Second Cardiology Department, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Louisa Pechlivani
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Theodora Nikou
- Division of Pharmacognosy and Natural Products Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Aimilia Varela
- Cardiovascular Research Laboratory, Biomedical Research Foundation Academy of Athens (BRFAA), Athens, Greece
| | - Christina Chania
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou, Athens, Greece
| | - Stelios Zerikiotis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou, Athens, Greece
| | - Panagiotis Efentakis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou, Athens, Greece
| | - Dimitris Vlachodimitropoulos
- Laboratory of Forensic Medicine and Toxicology, Medical School National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Katsoulas
- Laboratory of Forensic Medicine and Toxicology, Medical School National and Kapodistrian University of Athens, Athens, Greece
| | - Anna Agapaki
- Histochemistry Unit, Biomedical Research Foundation, Academy of Athens (BRFAA), Athens, Greece
| | - Costantinos Dimitriou
- Centre of Clinical Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Maria Tsoumani
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou, Athens, Greece
| | - Nikolaos Kostomitsopoulos
- Centre of Clinical Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Constantinos H Davos
- Cardiovascular Research Laboratory, Biomedical Research Foundation Academy of Athens (BRFAA), Athens, Greece
| | - Alexios Leandros Skaltsounis
- Division of Pharmacognosy and Natural Products Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Alexandros Tselepis
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Maria Halabalaki
- Division of Pharmacognosy and Natural Products Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | - Ignatios Ikonomidis
- Laboratory of Echocardiography and Preventive Cardiology, Second Cardiology Department, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou, Athens, Greece.
| |
Collapse
|
33
|
Köhler D, Leiss V, Beichert L, Killinger S, Grothe D, Kushwaha R, Schröter A, Roslan A, Eggstein C, Focken J, Granja T, Devanathan V, Schittek B, Lukowski R, Weigelin B, Rosenberger P, Nürnberg B, Beer-Hammer S. Targeting Gα i2 in neutrophils protects from myocardial ischemia reperfusion injury. Basic Res Cardiol 2024; 119:717-732. [PMID: 38811421 PMCID: PMC11461587 DOI: 10.1007/s00395-024-01057-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 05/31/2024]
Abstract
Neutrophils are not only involved in immune defense against infection but also contribute to the exacerbation of tissue damage after ischemia and reperfusion. We have previously shown that genetic ablation of regulatory Gαi proteins in mice has both protective and deleterious effects on myocardial ischemia reperfusion injury (mIRI), depending on which isoform is deleted. To deepen and analyze these findings in more detail the contribution of Gαi2 proteins in resident cardiac vs circulating blood cells for mIRI was first studied in bone marrow chimeras. In fact, the absence of Gαi2 in all blood cells reduced the extent of mIRI (22,9% infarct size of area at risk (AAR) Gnai2-/- → wt vs 44.0% wt → wt; p < 0.001) whereas the absence of Gαi2 in non-hematopoietic cells increased the infarct damage (66.5% wt → Gnai2-/- vs 44.0% wt → wt; p < 0.001). Previously we have reported the impact of platelet Gαi2 for mIRI. Here, we show that infarct size was substantially reduced when Gαi2 signaling was either genetically ablated in neutrophils/macrophages using LysM-driven Cre recombinase (AAR: 17.9% Gnai2fl/fl LysM-Cre+/tg vs 42.0% Gnai2fl/fl; p < 0.01) or selectively blocked with specific antibodies directed against Gαi2 (AAR: 19.0% (anti-Gαi2) vs 49.0% (IgG); p < 0.001). In addition, the number of platelet-neutrophil complexes (PNCs) in the infarcted area were reduced in both, genetically modified (PNCs: 18 (Gnai2fl/fl; LysM-Cre+/tg) vs 31 (Gnai2fl/fl); p < 0.001) and in anti-Gαi2 antibody-treated (PNCs: 9 (anti-Gαi2) vs 33 (IgG); p < 0.001) mice. Of note, significant infarct-limiting effects were achieved with a single anti-Gαi2 antibody challenge immediately prior to vessel reperfusion without affecting bleeding time, heart rate or cellular distribution of neutrophils. Finally, anti-Gαi2 antibody treatment also inhibited transendothelial migration of human neutrophils (25,885 (IgG) vs 13,225 (anti-Gαi2) neutrophils; p < 0.001), collectively suggesting that a therapeutic concept of functional Gαi2 inhibition during thrombolysis and reperfusion in patients with myocardial infarction should be further considered.
Collapse
Affiliation(s)
- David Köhler
- Department of Anesthesiology and Intensive Care Medicine, Eberhard Karls University, Tübingen, Germany
| | - Veronika Leiss
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute for Experimental and Clinical Pharmacology and Pharmacogenomic, Eberhard Karls University, and Interfaculty Center of Pharmacogenomic and Drug Research, Wilhelmstrasse 56, 72074, Tübingen, Germany
| | - Lukas Beichert
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute for Experimental and Clinical Pharmacology and Pharmacogenomic, Eberhard Karls University, and Interfaculty Center of Pharmacogenomic and Drug Research, Wilhelmstrasse 56, 72074, Tübingen, Germany
| | - Simon Killinger
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute for Experimental and Clinical Pharmacology and Pharmacogenomic, Eberhard Karls University, and Interfaculty Center of Pharmacogenomic and Drug Research, Wilhelmstrasse 56, 72074, Tübingen, Germany
| | - Daniela Grothe
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute for Experimental and Clinical Pharmacology and Pharmacogenomic, Eberhard Karls University, and Interfaculty Center of Pharmacogenomic and Drug Research, Wilhelmstrasse 56, 72074, Tübingen, Germany
| | - Ragini Kushwaha
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute for Experimental and Clinical Pharmacology and Pharmacogenomic, Eberhard Karls University, and Interfaculty Center of Pharmacogenomic and Drug Research, Wilhelmstrasse 56, 72074, Tübingen, Germany
| | - Agnes Schröter
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute for Experimental and Clinical Pharmacology and Pharmacogenomic, Eberhard Karls University, and Interfaculty Center of Pharmacogenomic and Drug Research, Wilhelmstrasse 56, 72074, Tübingen, Germany
| | - Anna Roslan
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Claudia Eggstein
- Department of Anesthesiology and Intensive Care Medicine, Eberhard Karls University, Tübingen, Germany
| | - Jule Focken
- Division of Dermatooncology, Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Tiago Granja
- Department of Anesthesiology and Intensive Care Medicine, Eberhard Karls University, Tübingen, Germany
| | - Vasudharani Devanathan
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute for Experimental and Clinical Pharmacology and Pharmacogenomic, Eberhard Karls University, and Interfaculty Center of Pharmacogenomic and Drug Research, Wilhelmstrasse 56, 72074, Tübingen, Germany
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, 517507, India
| | - Birgit Schittek
- Division of Dermatooncology, Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Robert Lukowski
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Bettina Weigelin
- Department of Preclinical Imaging and Radiopharmacy, Multiscale Immunoimaging, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Peter Rosenberger
- Department of Anesthesiology and Intensive Care Medicine, Eberhard Karls University, Tübingen, Germany
| | - Bernd Nürnberg
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute for Experimental and Clinical Pharmacology and Pharmacogenomic, Eberhard Karls University, and Interfaculty Center of Pharmacogenomic and Drug Research, Wilhelmstrasse 56, 72074, Tübingen, Germany
| | - Sandra Beer-Hammer
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute for Experimental and Clinical Pharmacology and Pharmacogenomic, Eberhard Karls University, and Interfaculty Center of Pharmacogenomic and Drug Research, Wilhelmstrasse 56, 72074, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany.
| |
Collapse
|
34
|
Wang Q, Tong Q, Jiang Z, Tang B. Predictive value of miR-636 in patients with acute myocardial infarction undergoing percutaneous coronary intervention and its bioinformatics analysis. J Cardiothorac Surg 2024; 19:572. [PMID: 39354590 PMCID: PMC11443705 DOI: 10.1186/s13019-024-03079-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 09/15/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) play an important role in the pathogenesis of cardiovascular diseases such as acute myocardial infarction (AMI). Percutaneous coronary intervention (PCI) is currently the most direct and effective procedure to treat AMI, but the occurrence of postoperative cardiovascular events (MACE) affects patients' quality of life. The objective of this study was to identify a new biomarker that could provide a theoretical basis for the prevention of MACE in patients with AMI undergoing PCI. METHODS 142 AMI patients who underwent PCI and 130 healthy volunteers were selected as study subjects. Detection of miR-636 expression level by fluorescence quantitative PCR. ROC, Kaplan-Meier and Cox regression analyses were applied to evaluate the diagnostic and prognostic value of miR-636 for AMI. The miR-636 target genes were predicted and enriched for GO function and KEGG pathway. RESULTS MiR-636 expression levels were elevated in patients with AMI. ROC curve analysis showed that miR-636 had a feasible diagnostic value in distinguishing AMI patients from healthy controls miR-636 expression levels were elevated in patients who developed MACEs. ROC results showed that miR-636 had significant diagnostic value in differentiating AMI patients with and without MACEs after PCI treatment. GO and KEGG enrichment analyses showed that miR-636 may transmit information to vesicles formed by the cell membrane. CONCLUSIONS MiR-636 expression serves as a biomarker for diagnosing AMI and predicting the occurrence of MACE after PCI.
Collapse
Affiliation(s)
- Qi Wang
- Department of Cardiovascular Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, No. 365, Renmin East Road, Jinhua, 321100, China
| | - Qiang Tong
- Department of Cardiovascular Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, No. 365, Renmin East Road, Jinhua, 321100, China
| | - Zenan Jiang
- Department of Cardiovascular Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, No. 365, Renmin East Road, Jinhua, 321100, China
| | - Biao Tang
- Department of Cardiovascular Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, No. 365, Renmin East Road, Jinhua, 321100, China.
| |
Collapse
|
35
|
Pyrpyris N, Dimitriadis K, Iliakis P, Theofilis P, Beneki E, Terentes-Printzios D, Sakalidis A, Antonopoulos A, Aznaouridis K, Tsioufis K. Hypothermia for Cardioprotection in Acute Coronary Syndrome Patients: From Bench to Bedside. J Clin Med 2024; 13:5390. [PMID: 39336877 PMCID: PMC11432135 DOI: 10.3390/jcm13185390] [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: 08/18/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Early revascularization for patients with acute myocardial infarction (AMI) is of outmost importance in limiting infarct size and associated complications, as well as for improving long-term survival and outcomes. However, reperfusion itself may further damage the myocardium and increase the infarct size, a condition commonly recognized as myocardial reperfusion injury. Several strategies have been developed for limiting the associated with reperfusion myocardial damage, including hypothermia. Hypothermia has been shown to limit the degree of infarct size increase, when started before reperfusion, in several animal models. Systemic hypothermia, however, failed to show any benefit, due to adverse events and potentially insufficient myocardial cooling. Recently, the novel technique of intracoronary selective hypothermia is being tested, with preclinical and clinical results being of particular interest. Therefore, in this review, we will describe the pathophysiology of myocardial reperfusion injury and the cardioprotective mechanics of hypothermia, report the animal and clinical evidence in both systemic and selective hypothermia and discuss the potential future directions and clinical perspectives in the context of cardioprotection for myocardial reperfusion injury.
Collapse
Affiliation(s)
| | - Kyriakos Dimitriadis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27 Athens, Greece; (N.P.); (P.I.); (P.T.); (E.B.); (D.T.-P.); (A.S.); (A.A.); (K.A.); (K.T.)
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Lieder HR, Paket U, Skyschally A, Rink AD, Baars T, Neuhäuser M, Kleinbongard P, Heusch G. Vago-splenic signal transduction of cardioprotection in humans. Eur Heart J 2024; 45:3164-3177. [PMID: 38842545 DOI: 10.1093/eurheartj/ehae250] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/13/2024] [Accepted: 04/08/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND AND AIMS The spleen serves as an important relay organ that releases cardioprotective factor(s) upon vagal activation during remote ischaemic conditioning (RIC) in rats and pigs. The translation of these findings to humans was attempted. METHODS Remote ischaemic conditioning or electrical auricular tragus stimulation (ATS) were performed in 10 healthy young volunteers, 10 volunteers with splenectomy, and 20 matched controls. Venous blood samples were taken before and after RIC/ATS or placebo, and a plasma dialysate was infused into isolated perfused rat hearts subjected to global ischaemia/reperfusion. RESULTS Neither left nor right RIC or ATS altered heart rate and heart rate variability in the study cohorts. With the plasma dialysate prepared before RIC or ATS, respectively, infarct size (% ventricular mass) in the recipient rat heart was 36 ± 6% (left RIC), 34 ± 3% (right RIC) or 31 ± 5% (left ATS), 35 ± 5% (right ATS), and decreased with the plasma dialysate from healthy volunteers after RIC or ATS to 20 ± 4% (left RIC), 23 ± 6% (right RIC) or to 19 ± 4% (left ATS), 26 ± 9% (right ATS); infarct size was still reduced with plasma dialysate 4 days after ATS and 9 days after RIC. In a subgroup of six healthy volunteers, such infarct size reduction was abrogated by intravenous atropine. Infarct size reduction by RIC or ATS was also abrogated in 10 volunteers with splenectomy, but not in their 20 matched controls. CONCLUSIONS In humans, vagal innervation and the spleen as a relay organ are decisive for the cardioprotective signal transduction of RIC and ATS.
Collapse
Affiliation(s)
- Helmut Raphael Lieder
- Institute for Pathophysiology, West German Heart and Vascular Centre, University of Essen Medical School, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Umut Paket
- Institute for Pathophysiology, West German Heart and Vascular Centre, University of Essen Medical School, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Andreas Skyschally
- Institute for Pathophysiology, West German Heart and Vascular Centre, University of Essen Medical School, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Andreas D Rink
- Department of General, Visceral and Transplant Surgery, University of Essen Medical School, University of Duisburg-Essen, Essen, Germany
| | - Theodor Baars
- Private Practice of General and Internal Medicine, Kölner Straße 68, Essen, Germany
| | - Markus Neuhäuser
- Department of Mathematics and Technology, Koblenz University of Applied Sciences, Rhein-Ahr-Campus, Remagen, Germany
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Centre, University of Essen Medical School, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Centre, University of Essen Medical School, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| |
Collapse
|
37
|
Gastaldi S, Giordano M, Blua F, Rubeo C, Boscaro V, Femminò S, Comità S, Gianquinto E, Landolfi V, Marini E, Gallicchio M, Spyrakis F, Pagliaro P, Bertinaria M, Penna C. Novel NLRP3 inhibitor INF195: Low doses provide effective protection against myocardial ischemia/reperfusion injury. Vascul Pharmacol 2024; 156:107397. [PMID: 38897555 DOI: 10.1016/j.vph.2024.107397] [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: 02/19/2024] [Revised: 05/21/2024] [Accepted: 06/16/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Several factors contribute to ischemia/reperfusion injury (IRI), including activation of the NLRP3 inflammasome and its byproducts, such as interleukin-1β (IL-1β) and caspase-1. However, NLRP3 may paradoxically exhibit cardioprotective properties. This study aimed to assess the protective effects of the novel NLRP3 inhibitor, INF195, both in vitro and ex vivo. METHODS To investigate the relationship between NLRP3 and myocardial IRI, we synthetized a series of novel NLRP3 inhibitors, and investigated their putative binding mode via docking studies. Through in vitro studies we identified INF195 as optimal for NLRP3 inhibition. We measured infarct-size in isolated mouse hearts subjected to 30-min global ischemia/one-hour reperfusion in the presence of three different doses of INF195 (5, 10, or 20-μM). We analyzed caspase-1 and IL-1β concentration in cardiac tissue homogenates by ELISA. Statistical significance was determined using one-way ANOVA followed by Tukey's test. RESULTS AND CONCLUSION INF195 reduces NLRP3-induced pyroptosis in human macrophages. Heart pre-treatment with 5 and 10-μM INF195 significantly reduces both infarct size and IL-1β levels. Data suggest that intracardiac NLRP3 activation contributes to IRI and that low doses of INF195 exert cardioprotective effects by reducing infarct size. However, at 20-μM, INF195 efficacy declines, leading to a lack of cardioprotection. Research is required to determine if high doses of INF195 have off-target effects or dual roles, potentially eliminating both harmful and cardioprotective functions of NLRP3. Our findings highlight the potential of a new chemical scaffold, amenable to further optimization, to provide NLRP3 inhibition and cardioprotection in the ischemia/reperfusion setting.
Collapse
Affiliation(s)
- Simone Gastaldi
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Torino, Italy
| | - Magalì Giordano
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Federica Blua
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Torino, Italy
| | - Chiara Rubeo
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Valentina Boscaro
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Torino, Italy
| | - Saveria Femminò
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Stefano Comità
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Eleonora Gianquinto
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Torino, Italy
| | - Vanessa Landolfi
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Elisabetta Marini
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Torino, Italy
| | - Margherita Gallicchio
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Torino, Italy
| | - Francesca Spyrakis
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Torino, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043 Torino, Italy; INRC, Bologna, Italy.
| | - Massimo Bertinaria
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Torino, Italy; Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043 Torino, Italy.
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043 Torino, Italy; INRC, Bologna, Italy
| |
Collapse
|
38
|
Zhang J, Zhao Y, Yan L, Tan M, Jin Y, Yin Y, Han L, Ma X, Li Y, Yang T, Jiang T, Li H. Corosolic acid attenuates cardiac ischemia/reperfusion injury through the PHB2/PINK1/parkin/mitophagy pathway. iScience 2024; 27:110448. [PMID: 39091464 PMCID: PMC11293524 DOI: 10.1016/j.isci.2024.110448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/20/2024] [Accepted: 07/01/2024] [Indexed: 08/04/2024] Open
Abstract
Despite advances in treatment, myocardial infarction remains the leading cause of heart failure and death worldwide, and the restoration of coronary blood flow can also cause heart damage. In this study, we found that corosolic acid (CA), also known as plant insulin, significantly protects the heart from ischemia-reperfusion (I/R) injury. In addition, CA can inhibit oxidative stress and improve mitochondrial structure and function in cardiomyocytes. Subsequently, our study demonstrated that CA improved the expression of the mitophagy-related proteins Prohibitin 2 (PHB2), PTEN-induced putative kinase protein-1 (PINK1), and Parkin. Meanwhile, through molecular docking, we found an excellent binding between CA and PHB2 protein. Finally, the knockdown of PHB2 eliminated the protective effect of CA on hypoxia-reoxygenation in cardiomyocytes. Taken together, our study reveals that CA increases mitophagy in cardiomyocytes via the PHB2/PINK1/Parkin signaling pathway, inhibits oxidative stress response, and maintains mitochondrial function, thereby improving cardiac function after I/R.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, P.R. China
| | - Yongjian Zhao
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, P.R. China
| | - Lin Yan
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, P.R. China
| | - Mingyue Tan
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, P.R. China
| | - Yifeng Jin
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, P.R. China
| | - Yunfei Yin
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, P.R. China
| | - Lianhua Han
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, P.R. China
| | - Xiao Ma
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, P.R. China
| | - Yimin Li
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, P.R. China
| | - Tianke Yang
- Department of Ophthalmology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, P.R. China
| | - Tingbo Jiang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, P.R. China
| | - Hongxia Li
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
39
|
Zhang J, Zhao Y, Yan L, Tan M, Jin Y, Yin Y, Han L, Ma X, Li Y, Yang T, Jiang T, Li H. Corosolic acid attenuates cardiac ischemia/reperfusion injury through the PHB2/PINK1/parkin/mitophagy pathway. iScience 2024; 27:110448. [DOI: pmid: 39091464 doi: 10.1016/j.isci.2024.110448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2025] Open
|
40
|
Abe J, Vujic A, Prag HA, Murphy MP, Krieg T. Malonate given at reperfusion prevents post-myocardial infarction heart failure by decreasing ischemia/reperfusion injury. Basic Res Cardiol 2024; 119:691-697. [PMID: 38864895 PMCID: PMC11319474 DOI: 10.1007/s00395-024-01063-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/13/2024]
Abstract
The mitochondrial metabolite succinate is a key driver of ischemia/reperfusion injury (IRI). Targeting succinate metabolism by inhibiting succinate dehydrogenase (SDH) upon reperfusion using malonate is an effective therapeutic strategy to achieve cardioprotection in the short term (< 24 h reperfusion) in mouse and pig in vivo myocardial infarction (MI) models. We aimed to assess whether inhibiting IRI with malonate given upon reperfusion could prevent post-MI heart failure (HF) assessed after 28 days. Male C57BL/6 J mice were subjected to 30 min left anterior coronary artery (LAD) occlusion, before reperfusion for 28 days. Malonate or without-malonate control was infused as a single dose upon reperfusion. Cardiac function was assessed by echocardiography and fibrosis by Masson's trichrome staining. Reperfusion without malonate significantly reduced ejection fraction (~ 47%), fractional shortening (~ 23%) and elevated collagen deposition 28 days post-MI. Malonate, administered as a single infusion (16 mg/kg/min for 10 min) upon reperfusion, gave a significant cardioprotective effect, with ejection fraction (~ 60%) and fractional shortening (~ 30%) preserved and less collagen deposition. Using an acidified malonate formulation, to enhance its uptake into cardiomyocytes via the monocarboxylate transporter 1, both 1.6 and 16 mg/kg/min 10 min infusion led to robust long-term cardioprotection with preserved ejection fraction (> 60%) and fractional shortening (~ 30%), as well as significantly less collagen deposition than control hearts. Malonate administration upon reperfusion prevents post-MI HF. Acidification of malonate enables lower doses of malonate to also achieve long-term cardioprotection post-MI. Therefore, the administration of acidified malonate upon reperfusion is a promising therapeutic strategy to prevent IRI and post-MI HF.
Collapse
Affiliation(s)
- Jiro Abe
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
- MRC Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
| | - Ana Vujic
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Hiran A Prag
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK.
- MRC Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK.
| | - Michael P Murphy
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK.
- MRC Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK.
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
41
|
Melberg MB, Flaa A, Andersen GØ, Sunde K, Bellomo R, Eastwood G, Olasveengen TM, Qvigstad E. Effects of mild hypercapnia on myocardial injury after out-of-hospital cardiac arrest. A sub-study of the TAME trial. Resuscitation 2024; 201:110295. [PMID: 38936652 DOI: 10.1016/j.resuscitation.2024.110295] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/21/2024] [Accepted: 06/21/2024] [Indexed: 06/29/2024]
Abstract
PURPOSE Mild hypercapnia did not improve neurological outcomes for resuscitated out-of-hospital cardiac arrest (OHCA) patients in the Targeted Therapeutic Mild Hypercapnia After Resuscitated Cardiac Arrest (TAME) trial. However, the effects of hypercapnic acidosis on myocardial injury in patients with cardiac arrest is unexplored. We investigated whether mild hypercapnia compared to normocapnia, following emergency coronary intervention, increased myocardial injury in comatose OHCA-patients with AMI. METHODS Single-centre, prospective, pre-planned sub-study of the TAME trial. Patients were randomised to targeted mild hypercapnia (PaCO2 = 6.7-7.3 kPa) or normocapnia (PaCO2 = 4.7-6.0 kPa) for 24 h. Myocardial injury was assessed with high-sensitive cardiac troponin T (hs-cTnT) measured at baseline, 24, 48 and 72 h. Haemodynamics were assessed with right heart catheterisation and blood-gas analyses every 4th hour for 48 h. RESULTS We included 125 OHCA-patients. 57 (46%) had an AMI, with 31 and 26 patients randomised to hypercapnia and normocapnia, respectively. Median peak hs-cTnT in AMI-patients was 58% lower in the hypercapnia-group: 2136 (IQR: 861-4462) versus 5165 ng/L (IQR: 2773-7519), p = 0.007. Lower average area under the hs-cTnT curve was observed in the hypercapnia-group: 2353 (95% CI 1388-3319) versus 4953 ng/L (95% CI 3566-6341), P-group = 0.002. Hypercapnia was associated with increased cardiac power output (CPO) and lower lactate levels in patients with AMI (P-group < 0.05). hs-cTnT, lactate and CPO were not significantly different between intervention groups in OHCA-patients without AMI (p > 0.05). CONCLUSIONS Mild hypercapnia was not associated with increased myocardial injury in resuscitated OHCA-patients. In AMI-patients, mild hypercapnia was associated with lower hs-cTnT and lactate, and improved cardiac performance. TRIAL REGISTRATION NUMBER NCT03114033.
Collapse
Affiliation(s)
- Mathias Baumann Melberg
- Department of Research and Development, Division of Emergencies and Critical Care, Department of Cardiology, Division of Medicine, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Norway.
| | - Arnljot Flaa
- Department of Research and Development, Division of Emergencies and Critical Care, Department of Cardiology, Division of Medicine, Oslo University Hospital, Oslo, Norway
| | - Geir Øystein Andersen
- Department of Research and Development, Division of Emergencies and Critical Care, Department of Cardiology, Division of Medicine, Oslo University Hospital, Oslo, Norway
| | - Kjetil Sunde
- Department of Anaesthesiology and Intensive Care, Division of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway and Institute of Clinical Medicine, University of Oslo, Norway
| | - Rinaldo Bellomo
- Department of Intensive Care, Austin Hospital, Heidelberg, Victoria, Australia; Australian and New Zealand Intensive Care Research Centre, Monash University, Victoria, Melbourne, Australia; Department of Critical Care, Melbourne University, Melbourne, Australia; Department of Intensive Care, Royal Melbourne Hospital, Melbourne, Australia
| | - Glenn Eastwood
- Department of Intensive Care, Austin Hospital, Heidelberg, Victoria, Australia; Australian and New Zealand Intensive Care Research Centre, Monash University, Victoria, Melbourne, Australia
| | - Theresa Mariero Olasveengen
- Department of Anaesthesiology and Intensive Care, Division of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway and Institute of Clinical Medicine, University of Oslo, Norway
| | - Eirik Qvigstad
- Department of Research and Development, Division of Emergencies and Critical Care, Department of Cardiology, Division of Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
42
|
Johnson TW, Holt J, Kleyman A, Zhou S, Sammut E, Bruno VD, Gaupp C, Stanzani G, Martin J, Arina P, Deutsch J, Ascione R, Singer M, Dyson A. Development and translation of thiometallate sulfide donors using a porcine model of coronary occlusion and reperfusion. Redox Biol 2024; 73:103167. [PMID: 38688060 PMCID: PMC11070758 DOI: 10.1016/j.redox.2024.103167] [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: 02/15/2024] [Revised: 04/12/2024] [Accepted: 04/22/2024] [Indexed: 05/02/2024] Open
Abstract
Sulfide-releasing compounds reduce reperfusion injury by decreasing mitochondria-derived reactive oxygen species production. We previously characterised ammonium tetrathiomolybdate (ATTM), a clinically used copper chelator, as a sulfide donor in rodents. Here we assessed translation to large mammals prior to clinical testing. In healthy pigs an intravenous ATTM dose escalation revealed a reproducible pharmacokinetic/pharmacodynamic (PK/PD) relationship with minimal adverse clinical or biochemical events. In a myocardial infarction (1-h occlusion of the left anterior descending coronary artery)-reperfusion model, intravenous ATTM or saline was commenced just prior to reperfusion. ATTM protected the heart (24-h histological examination) in a drug-exposure-dependent manner (r2 = 0.58, p < 0.05). Blood troponin T levels were significantly (p < 0.05) lower in ATTM-treated animals while myocardial glutathione peroxidase activity, an antioxidant selenoprotein, was elevated (p < 0.05). Overall, our study represents a significant advance in the development of sulfides as therapeutics and underlines the potential of ATTM as a novel adjunct therapy for reperfusion injury. Mechanistically, our study suggests that modulating selenoprotein activity could represent an additional mode of action of sulfide-releasing drugs.
Collapse
Affiliation(s)
- Thomas W Johnson
- Translational Biomedical Research Centre (TBRC), Faculty of Health Science, University of Bristol, UK
| | - James Holt
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Anna Kleyman
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Shengyu Zhou
- Institute of Pharmaceutical Science, King's College London, London, UK; Centre for Pharmaceutical Medicine Research, King's College London, London, UK
| | - Eva Sammut
- Translational Biomedical Research Centre (TBRC), Faculty of Health Science, University of Bristol, UK
| | - Vito Domenico Bruno
- Translational Biomedical Research Centre (TBRC), Faculty of Health Science, University of Bristol, UK
| | - Charlotte Gaupp
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Giacomo Stanzani
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - John Martin
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Pietro Arina
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Julia Deutsch
- Translational Biomedical Research Centre (TBRC), Faculty of Health Science, University of Bristol, UK
| | - Raimondo Ascione
- Translational Biomedical Research Centre (TBRC), Faculty of Health Science, University of Bristol, UK
| | - Mervyn Singer
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK.
| | - Alex Dyson
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK; Institute of Pharmaceutical Science, King's College London, London, UK; Centre for Pharmaceutical Medicine Research, King's College London, London, UK.
| |
Collapse
|
43
|
Braczko F, Fischl SR, Reinders J, Lieder HR, Kleinbongard P. Activation of the nonneuronal cholinergic cardiac system by hypoxic preconditioning protects isolated adult cardiomyocytes from hypoxia/reoxygenation injury. Am J Physiol Heart Circ Physiol 2024; 327:H70-H79. [PMID: 38700468 PMCID: PMC11380960 DOI: 10.1152/ajpheart.00211.2024] [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: 04/04/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
Activation of the vagus nerve mediates cardioprotection and attenuates myocardial ischemia/reperfusion (I/R) injury. In response to vagal activation, acetylcholine (ACh) is released from the intracardiac nervous system (ICNS) and activates intracellular cardioprotective signaling cascades. Recently, however, a nonneuronal cholinergic cardiac system (NNCCS) in cardiomyocytes has been described as an additional source of ACh. To investigate whether the NNCCS mediates cardioprotection in the absence of vagal and ICNS activation, we used a reductionist approach of isolated adult rat ventricular cardiomyocytes without neuronal cells, using hypoxic preconditioning (HPC) as a protective stimulus. Adult rat ventricular cardiomyocytes were isolated, the absence of neuronal cells was confirmed, and HPC was induced by 10/20 min hypoxia/reoxygenation (H/R) before subjection to 30/5 min H/R to simulate I/R injury. Cardiomyocyte viability was assessed by trypan blue staining at baseline and after HPC+H/R or H/R. Intra- and extracellular ACh was quantified using liquid chromatography-coupled mass spectrometry at baseline, after HPC, after hypoxia, and after reoxygenation, respectively. In a subset of experiments, muscarinic and nicotinic ACh receptor (m- and nAChR) antagonists were added during HPC or during H/R. Cardiomyocyte viability at baseline (69 ± 4%) was reduced by H/R (10 ± 3%). With HPC, cardiomyocyte viability was preserved after H/R (25 ± 6%). Intra- and extracellular ACh increased during hypoxia; HPC further increased both intra- and extracellular ACh (from 0.9 ± 0.7 to 1.5 ± 1.0 nmol/mg; from 0.7 ± 0.6 to 1.1 ± 0.7 nmol/mg, respectively). The addition of mAChR and nAChR antagonists during HPC had no impact on HPC's protection; however, protection was abrogated when antagonists were added during H/R (cardiomyocyte viability after H/R: 23 ± 5%; 13 ± 4%). In conclusion, activation of the NNCCS is involved in cardiomyocyte protection; HPC increases intra- and extracellular ACh during H/R, and m- and nAChRs are causally involved in HPC's cardiomyocyte protection during H/R. The interplay between upstream ICNS activation and NNCCS activation in myocardial cholinergic metabolism and cardioprotection needs to be investigated in future studies.NEW & NOTEWORTHY The intracardiac nervous system is considered to be involved in ischemic conditioning's cardioprotection through the release of acetylcholine (ACh). However, we demonstrate that hypoxic preconditioning (HPC) protects from hypoxia/reoxygenation injury and increases intra- and extracellular ACh during hypoxia in isolated adult ventricular rat cardiomyocytes. HPC's protection involves cardiomyocyte muscarinic and nicotinic ACh receptor activation. Thus, besides the intracardiac nervous system, a nonneuronal cholinergic cardiac system may also be causally involved in cardiomyocyte protection by ischemic conditioning.
Collapse
Affiliation(s)
- Felix Braczko
- Institute for Pathophysiology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Sara Romina Fischl
- Institute for Pathophysiology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Jörg Reinders
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Dortmund, Germany
| | - Helmut Raphael Lieder
- Institute for Pathophysiology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
44
|
Brosinsky P, Heger J, Sydykov A, Weiss A, Klatt S, Czech L, Kraut S, Schermuly RT, Schlüter KD, Schulz R. Does Cell-Type-Specific Silencing of Monoamine Oxidase B Interfere with the Development of Right Ventricle (RV) Hypertrophy or Right Ventricle Failure in Pulmonary Hypertension? Int J Mol Sci 2024; 25:6212. [PMID: 38892401 PMCID: PMC11172614 DOI: 10.3390/ijms25116212] [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: 04/16/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Increased mitochondrial reactive oxygen species (ROS) formation is important for the development of right ventricular (RV) hypertrophy (RVH) and failure (RVF) during pulmonary hypertension (PH). ROS molecules are produced in different compartments within the cell, with mitochondria known to produce the strongest ROS signal. Among ROS-forming mitochondrial proteins, outer-mitochondrial-membrane-located monoamine oxidases (MAOs, type A or B) are capable of degrading neurotransmitters, thereby producing large amounts of ROS. In mice, MAO-B is the dominant isoform, which is present in almost all cell types within the heart. We analyzed the effect of an inducible cardiomyocyte-specific knockout of MAO-B (cmMAO-B KO) for the development of RVH and RVF in mice. Right ventricular hypertrophy was induced by pulmonary artery banding (PAB). RV dimensions and function were measured through echocardiography. ROS production (dihydroethidium staining), protein kinase activity (PamStation device), and systemic hemodynamics (in vivo catheterization) were assessed. A significant decrease in ROS formation was measured in cmMAO-B KO mice during PAB compared to Cre-negative littermates, which was associated with reduced activity of protein kinases involved in hypertrophic growth. In contrast to littermates in which the RV was dilated and hypertrophied following PAB, RV dimensions were unaffected in response to PAB in cmMAO-B KO mice, and no decline in RV systolic function otherwise seen in littermates during PAB was measured in cmMAO-B KO mice. In conclusion, cmMAO-B KO mice are protected against RV dilatation, hypertrophy, and dysfunction following RV pressure overload compared to littermates. These results support the hypothesis that cmMAO-B is a key player in causing RV hypertrophy and failure during PH.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Disease Models, Animal
- Heart Failure/metabolism
- Heart Failure/etiology
- Heart Failure/genetics
- Heart Failure/pathology
- Heart Ventricles/pathology
- Heart Ventricles/metabolism
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/genetics
- Hypertrophy, Right Ventricular/etiology
- Hypertrophy, Right Ventricular/pathology
- Mice, Knockout
- Monoamine Oxidase/genetics
- Monoamine Oxidase/metabolism
- Monoamine Oxidase/deficiency
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Reactive Oxygen Species/metabolism
- Ventricular Dysfunction, Right/metabolism
- Ventricular Dysfunction, Right/genetics
- Ventricular Dysfunction, Right/etiology
- Ventricular Dysfunction, Right/pathology
Collapse
Affiliation(s)
- Paulin Brosinsky
- Physiologisches Institut, Justus-Liebig-Universität, 35392 Gießen, Germany; (J.H.); (L.C.); (K.-D.S.); (R.S.)
| | - Jacqueline Heger
- Physiologisches Institut, Justus-Liebig-Universität, 35392 Gießen, Germany; (J.H.); (L.C.); (K.-D.S.); (R.S.)
| | - Akylbek Sydykov
- Excellence Cluster Cardiopulmonary System (ECCPS), Justus-Liebig-Universität, 35392 Gießen, Germany; (A.S.); (A.W.); (S.K.); (R.T.S.)
| | - Astrid Weiss
- Excellence Cluster Cardiopulmonary System (ECCPS), Justus-Liebig-Universität, 35392 Gießen, Germany; (A.S.); (A.W.); (S.K.); (R.T.S.)
| | - Stephan Klatt
- Vascular Research Centre, Goethe Universität, 60590 Frankfurt, Germany;
| | - Laureen Czech
- Physiologisches Institut, Justus-Liebig-Universität, 35392 Gießen, Germany; (J.H.); (L.C.); (K.-D.S.); (R.S.)
| | - Simone Kraut
- Excellence Cluster Cardiopulmonary System (ECCPS), Justus-Liebig-Universität, 35392 Gießen, Germany; (A.S.); (A.W.); (S.K.); (R.T.S.)
| | - Ralph Theo Schermuly
- Excellence Cluster Cardiopulmonary System (ECCPS), Justus-Liebig-Universität, 35392 Gießen, Germany; (A.S.); (A.W.); (S.K.); (R.T.S.)
| | - Klaus-Dieter Schlüter
- Physiologisches Institut, Justus-Liebig-Universität, 35392 Gießen, Germany; (J.H.); (L.C.); (K.-D.S.); (R.S.)
| | - Rainer Schulz
- Physiologisches Institut, Justus-Liebig-Universität, 35392 Gießen, Germany; (J.H.); (L.C.); (K.-D.S.); (R.S.)
| |
Collapse
|
45
|
Schnitter F, Stangl F, Noeske E, Bille M, Stadtmüller A, Vogt N, Sicklinger F, Leuschner F, Frey A, Schreiber L, Frantz S, Beyersdorf N, Ramos G, Gladow N, Hofmann U. Characterizing the immune response to myocardial infarction in pigs. Basic Res Cardiol 2024; 119:453-479. [PMID: 38491291 PMCID: PMC11143055 DOI: 10.1007/s00395-024-01036-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 01/30/2024] [Accepted: 01/30/2024] [Indexed: 03/18/2024]
Abstract
Though myocardial infarction (MI) in pigs is a well-established translational large animal model, it has not yet been widely used for immunotherapy studies, and a comprehensive description of the immune response to MI in this species is lacking. We induced MI in Landrace pigs by balloon occlusion of the left anterior descending artery over 90 min. Within 14 days, the necrotic myocardium was progressively replaced by scar tissue with involvement of myofibroblasts. We characterized the immune response in the heart ex vivo by (immuno)histology, flow cytometry, and RNA sequencing of myocardial tissue on days 3, 7, and 14 after MI. Besides a clear predominance of myeloid cells among heart-infiltrating leukocytes, we detected activated T cells and an increasing proportion of CD4+ Foxp3+ regulatory T cells (Treg), especially in the infarct core-findings that closely mirror what has been observed in mice and humans after MI. Transcriptome data indicated inflammatory activity that was persistent but markedly changing in character over time and linked to extracellular matrix biology. Analysis of lymphocytes in heart-draining lymph nodes revealed significantly higher proliferation rates of T helper cell subsets, including Treg on day 7 after MI, compared to sham controls. Elevated frequencies of myeloid progenitors in the spleen suggest that it might be a site of emergency myelopoiesis after MI in pigs, as previously shown in mice. We thus provide a first description of the immune response to MI in pigs, and our results can aid future research using the species for preclinical immunotherapy studies.
Collapse
Affiliation(s)
- Florian Schnitter
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany.
| | - Franziska Stangl
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Elisabeth Noeske
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Maya Bille
- Comprehensive Heart Failure Center, Department of Cardiovascular Imaging, University Hospital Würzburg, Würzburg, Germany
| | - Anja Stadtmüller
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Niklas Vogt
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Florian Sicklinger
- Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg, Heidelberg, Germany
| | - Florian Leuschner
- Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg, Heidelberg, Germany
| | - Anna Frey
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Laura Schreiber
- Comprehensive Heart Failure Center, Department of Cardiovascular Imaging, University Hospital Würzburg, Würzburg, Germany
| | - Stefan Frantz
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Niklas Beyersdorf
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Gustavo Ramos
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Nadine Gladow
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Ulrich Hofmann
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
46
|
Rubeo C, Hoti G, Giordano M, Molinar C, Aragno M, Mantuano B, Comità S, Femminò S, Cavalli R, Trotta F, Penna C, Pagliaro P. Enhancing Heart Transplantation: Utilizing Gas-Loaded Nanocarriers to Mitigate Cold/Hypoxia Stress. Int J Mol Sci 2024; 25:5685. [PMID: 38891873 PMCID: PMC11171608 DOI: 10.3390/ijms25115685] [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: 04/26/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Gas-loaded nanocarriers (G-LN) show promise in improving heart transplantation (HTx) outcomes. Given their success in reducing cell death during normothermic hypoxia/reoxygenation (H/R) in vitro, we tested their integration into cardioplegic solutions and static cold storage (SCS) during simulated HTx. Wistar rat hearts underwent four hours of SCS with four G-LN variants: O2- or N2-cyclic-nigerosyl-nigerose-nanomonomers (CNN), and O2- or N2-cyclic-nigerosyl-nigerose-nanosponges (CNN-NS). We monitored physiological-hemodynamic parameters and molecular markers during reperfusion to assess cell damage/protection. Hearts treated with nanomonomers (N2-CNN or O2-CNN) showed improvements in left ventricular developed pressure (LVDP) and a trend towards faster recovery of the rate pressure product (RPP) compared to controls. However, nanosponges (N2-CNN-NS or O2-CNN-NS) did not show similar improvements. None of the groups exhibited an increase in diastolic left ventricular pressure (contracture index) during reperfusion. Redox markers and apoptosis/autophagy pathways indicated an increase in Beclin 1 for O2-CNN and in p22phox for N2-CNN, suggesting alterations in autophagy and the redox environment during late reperfusion, which might explain the gradual decline in heart performance. The study highlights the potential of nanomonomers to improve early cardiac performance and mitigate cold/H/R-induced stunning in HTx. These early improvements suggest a promising avenue for increasing HTx success. Nevertheless, further research and optimization are needed before clinical application.
Collapse
Affiliation(s)
- Chiara Rubeo
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (C.R.); (M.G.); (M.A.); (B.M.); (S.C.); (S.F.)
| | - Gjylije Hoti
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Torino, Italy; (G.H.); (C.M.); (R.C.)
| | - Magalì Giordano
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (C.R.); (M.G.); (M.A.); (B.M.); (S.C.); (S.F.)
| | - Chiara Molinar
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Torino, Italy; (G.H.); (C.M.); (R.C.)
| | - Manuela Aragno
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (C.R.); (M.G.); (M.A.); (B.M.); (S.C.); (S.F.)
| | - Beatrice Mantuano
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (C.R.); (M.G.); (M.A.); (B.M.); (S.C.); (S.F.)
| | - Stefano Comità
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (C.R.); (M.G.); (M.A.); (B.M.); (S.C.); (S.F.)
| | - Saveria Femminò
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (C.R.); (M.G.); (M.A.); (B.M.); (S.C.); (S.F.)
| | - Roberta Cavalli
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Torino, Italy; (G.H.); (C.M.); (R.C.)
| | - Francesco Trotta
- Department of Chemistry, University of Turin, Via P. Giuria 7, 10125 Torino, Italy;
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (C.R.); (M.G.); (M.A.); (B.M.); (S.C.); (S.F.)
- National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (C.R.); (M.G.); (M.A.); (B.M.); (S.C.); (S.F.)
- National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy
| |
Collapse
|
47
|
Seefeldt JM, Homilius C, Hansen J, Lassen TR, Jespersen NR, Jensen RV, Boedtkjer E, Bøtker HE, Nielsen R. Short-Chain Fatty Acid Butyrate Is an Inotropic Agent With Vasorelaxant and Cardioprotective Properties. J Am Heart Assoc 2024; 13:e033744. [PMID: 38686853 PMCID: PMC11179878 DOI: 10.1161/jaha.123.033744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/21/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND The heart can metabolize the microbiota-derived short-chain fatty acid butyrate. Butyrate may have beneficial effects in heart failure, but the underlying mechanisms are unknown. We tested the hypothesis that butyrate elevates cardiac output by mechanisms involving direct stimulation of cardiac contractility and vasorelaxation in rats. METHODS AND RESULTS We examined the effects of butyrate on (1) in vivo hemodynamics using parallel echocardiographic and invasive blood pressure measurements, (2) isolated perfused hearts in Langendorff systems under physiological conditions and after ischemia and reperfusion, and (3) isolated coronary arteries mounted in isometric wire myographs. We tested Na-butyrate added to injection solutions or physiological buffers and compared its effects with equimolar doses of NaCl. Butyrate at plasma concentrations of 0.56 mM increased cardiac output by 48.8±14.9%, stroke volume by 38.5±12.1%, and left ventricular ejection fraction by 39.6±6.2%, and lowered systemic vascular resistance by 33.5±6.4% without affecting blood pressure or heart rate in vivo. In the range between 0.1 and 5 mM, butyrate increased left ventricular systolic pressure by up to 23.7±3.4% in isolated perfused hearts and by 9.4±2.9% following ischemia and reperfusion, while reducing myocardial infarct size by 81.7±16.9%. Butyrate relaxed isolated coronary septal arteries concentration dependently with an EC50=0.57 mM (95% CI, 0.23-1.44). CONCLUSIONS We conclude that butyrate elevates cardiac output through mechanisms involving increased cardiac contractility and vasorelaxation. This effect of butyrate was not associated with adverse myocardial injury in damaged hearts exposed to ischemia and reperfusion.
Collapse
Affiliation(s)
- Jacob Marthinsen Seefeldt
- Department of Clinical Medicine Aarhus University Aarhus Denmark
- Department of Cardiology Aarhus University Hospital Aarhus Denmark
| | | | - Jakob Hansen
- Department of Clinical Medicine Aarhus University Aarhus Denmark
- Department of Forensic Medicine Aarhus University Hospital Aarhus Denmark
| | | | | | | | - Ebbe Boedtkjer
- Department of Biomedicine Aarhus University Aarhus Denmark
| | - Hans Erik Bøtker
- Department of Clinical Medicine Aarhus University Aarhus Denmark
- Department of Cardiology Aarhus University Hospital Aarhus Denmark
| | - Roni Nielsen
- Department of Clinical Medicine Aarhus University Aarhus Denmark
- Department of Cardiology Aarhus University Hospital Aarhus Denmark
| |
Collapse
|
48
|
Escudero DS, Fantinelli JC, Martínez VR, González Arbeláez LF, Amarillo ME, Pérez NG, Díaz RG. Hydrocortisone cardioprotection in ischaemia/reperfusion injury involves antioxidant mechanisms. Eur J Clin Invest 2024; 54:e14172. [PMID: 38293760 DOI: 10.1111/eci.14172] [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: 10/27/2023] [Revised: 12/22/2023] [Accepted: 01/12/2024] [Indexed: 02/01/2024]
Abstract
BACKGROUND Glucocorticoid (GR) and mineralocorticoid (MR) receptors are highly expressed in cardiac tissue, and both can be activated by corticosteroids. MR activation, in acute myocardial infarction (AMI), worsens cardiac function, and increase NHE activity contributing to the deleterious process. In contrast, effects of GR activation are not fully understood, probably because of the controversial scenario generated by using different doses or potencies of corticosteroids. AIMS We tested the hypothesis that an acute dose of hydrocortisone (HC), a low-potency glucocorticoid, in a murine model of AMI could be cardioprotective by regulating NHE1 activity, leading to a decrease in oxidative stress. MATERIALS AND METHODS Isolated hearts from Wistar rats were subjected to regional ischemic protocol. HC (10 nmol/L) was added to the perfusate during early reperfusion. Infarct size and oxidative stress were determined. Isolated papillary muscles from non-infarcted hearts were used to evaluate HC effect on sodium-proton exchanger 1 (NHE1) by analysing intracellular pH recovery from acute transient acidosis. RESULTS HC treatment decreased infarct size, improved cardiac mechanics, reduced oxidative stress after AMI, while restoring the decreased level of the pro-fusion mitochondrial protein MFN-2. Co-treatment with the GR-blocker Mifepristone avoided these effects. HC reduced NHE1 activity by increasing the NHE1 pro-inhibiting Ser648 phosphorylation site and its upstream kinase AKT. HC restored the decreased AKT phosphorylation and anti-apoptotic BCL-2 protein expression detected after AMI. CONCLUSIONS Our results provide the first evidence that acute HC treatment during early reperfusion induces cardioprotection against AMI, associated with a non-genomic HC-triggered NHE1 inhibition by AKT and antioxidant action that might involves mitochondrial dynamics improvement.
Collapse
Affiliation(s)
- Daiana S Escudero
- Centro de Investigaciones Cardiovasculares 'Dr. Horacio E. Cingolani', Facultad de Ciencias Médicas de La Plata, Universidad Nacional de La Plata, La Plata, Argentina
- Established Investigator of Comisión de Investigaciones Científicas (CIC), Buenos Aires, Argentina
| | - Juliana C Fantinelli
- Centro de Investigaciones Cardiovasculares 'Dr. Horacio E. Cingolani', Facultad de Ciencias Médicas de La Plata, Universidad Nacional de La Plata, La Plata, Argentina
- Established Investigators of Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Valeria R Martínez
- Centro de Investigaciones Cardiovasculares 'Dr. Horacio E. Cingolani', Facultad de Ciencias Médicas de La Plata, Universidad Nacional de La Plata, La Plata, Argentina
- Established Investigators of Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Luisa F González Arbeláez
- Centro de Investigaciones Cardiovasculares 'Dr. Horacio E. Cingolani', Facultad de Ciencias Médicas de La Plata, Universidad Nacional de La Plata, La Plata, Argentina
- Established Investigators of Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María E Amarillo
- Centro de Investigaciones Cardiovasculares 'Dr. Horacio E. Cingolani', Facultad de Ciencias Médicas de La Plata, Universidad Nacional de La Plata, La Plata, Argentina
- Fellow of Agencia Nacional de Promoción Científica y Tecnológica (Agencia I+D+i), Buenos Aires, Argentina
| | - Néstor G Pérez
- Centro de Investigaciones Cardiovasculares 'Dr. Horacio E. Cingolani', Facultad de Ciencias Médicas de La Plata, Universidad Nacional de La Plata, La Plata, Argentina
- Established Investigators of Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Romina G Díaz
- Centro de Investigaciones Cardiovasculares 'Dr. Horacio E. Cingolani', Facultad de Ciencias Médicas de La Plata, Universidad Nacional de La Plata, La Plata, Argentina
- Established Investigators of Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
49
|
Majid Z, Muhammad-Baqir B, Al-Shimerty DF, Hadi NR. The possible cardioprotective effect of ghrelin during experimental endotoxemia in mice. J Med Life 2024; 17:486-491. [PMID: 39144689 PMCID: PMC11320619 DOI: 10.25122/jml-2023-0228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 10/04/2023] [Indexed: 08/16/2024] Open
Abstract
This study aimed to evaluate the cardioprotective effects of ghrelin in septic mice, focusing on its anti-inflammatory and antioxidant properties. Thirty-five male Swiss mice (8-12 weeks old, 23-33g) were randomly assigned to five groups (n = 7 each): (1) Normal, fed usual diets, (2) Sham, subjected to anesthesia and laparotomy, (3) Sepsis, subjected to cecal ligation and puncture, (4) Vehicle, given an equivalent volume of intraperitoneal saline injections immediately after cecal ligation and puncture, and (5) Ghrelin-treated, administered 80 µg/kg ghrelin intraperitoneal injections immediately following cecal ligation and puncture. Serum levels of tumor necrosis factor-alpha (TNF-α), macrophage migration inhibitory factor (MIF), toll-like receptor 4 (TLR4), and 8-epi-prostaglandin F2 alpha (8-epi-PGF2α) were measured. The extent of cardiac damage was also evaluated histologically. The mean serum levels of TNF-α, MIF, TLR4, and 8-epi-PGF2α levels were significantly higher in the sepsis and vehicle groups than in the normal and sham groups. The levels were significantly lower in the ghrelin-treated group than in the vehicle and sepsis groups. Histological analysis revealed normal myocardial architecture in the normal and sham groups, whereas the sepsis and vehicle groups had severe myocardial injury. The ghrelin-treated group displayed histological features similar to the sham group, indicating reduced myocardial damage. Ghrelin ameliorated sepsis-induced cardiotoxicity in mice by exhibiting strong anti-inflammatory and antioxidant effects. These findings suggest that ghrelin may be a promising therapeutic candidate for the prevention of sepsis-induced cardiotoxicity.
Collapse
Affiliation(s)
| | | | | | - Najah Rayish Hadi
- Department of Pharmacology & Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq
| |
Collapse
|
50
|
Yu W, Kong Q, Jiang S, Li Y, Wang Z, Mao Q, Zhang X, Liu Q, Zhang P, Li Y, Li C, Ding Z, Liu L. HSPA12A maintains aerobic glycolytic homeostasis and Histone3 lactylation in cardiomyocytes to attenuate myocardial ischemia/reperfusion injury. JCI Insight 2024; 9:e169125. [PMID: 38421727 PMCID: PMC11128201 DOI: 10.1172/jci.insight.169125] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 02/21/2024] [Indexed: 03/02/2024] Open
Abstract
Myocardial ischemia/reperfusion (MI/R) injury is a major cause of adverse outcomes of revascularization following myocardial infarction. Anaerobic glycolysis during myocardial ischemia is well studied, but the role of aerobic glycolysis during the early phase of reperfusion is incompletely understood. Lactylation of Histone H3 (H3) is an epigenetic indicator of the glycolytic switch. Heat shock protein A12A (HSPA12A) is an atypic member of the HSP70 family. In the present study, we report that, during reperfusion following myocardial ischemia, HSPA12A was downregulated and aerobic glycolytic flux was decreased in cardiomyocytes. Notably, HSPA12A KO in mice exacerbated MI/R-induced aerobic glycolysis decrease, cardiomyocyte death, and cardiac dysfunction. Gain- and loss-of-function studies demonstrated that HSPA12A was required to support cardiomyocyte survival upon hypoxia/reoxygenation (H/R) challenge and that its protective effects were mediated by maintaining aerobic glycolytic homeostasis for H3 lactylation. Further analyses revealed that HSPA12A increased Smurf1-mediated Hif1α protein stability, thus increasing glycolytic gene expression to maintain appropriate aerobic glycolytic activity to sustain H3 lactylation during reperfusion and, ultimately, improving cardiomyocyte survival to attenuate MI/R injury.
Collapse
Affiliation(s)
- Wansu Yu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, and
| | - Qiuyue Kong
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Surong Jiang
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, and
| | - Yunfan Li
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhaohe Wang
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qian Mao
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaojin Zhang
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, and
| | - Qianhui Liu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, and
| | - Pengjun Zhang
- Department of Nuclear Medicine, Nanjing First Hospital of Nanjing Medical University, Nanjing, China
| | - Yuehua Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China
| | - Chuanfu Li
- Departments of Surgery, East Tennessee State University, Johnson City, Tennessee, USA
| | - Zhengnian Ding
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Li Liu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, and
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China
| |
Collapse
|