1
|
Behzad H, Asham H, Beheshtirouy S, Mashayekhi S, Jafari N, Entezari-Maleki T. Sodium-glucose cotransporter-2 inhibitors in individuals with ischemia reperfusion injury: A systematic review. Perfusion 2025; 40:701-710. [PMID: 38813587 DOI: 10.1177/02676591241257371] [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] [Indexed: 05/31/2024]
Abstract
Background: The effects of sodium-glucose cotransporter-2 (SGLT-2) inhibitors on ischemia reperfusion injury (IRI) is a novel concept and only limited number of animals studies have yet been investigated. We aimed to perform a systematic review of literature to explore the clinical studies which investigated the effects of SGLT-2 inhibitors on myocardial IRI setting.Methods: We searched MEDLINE, Embase, and Cochrane Library from inception until December 7th, 2023. ClinicalTrials.gov was also explored for ongoing studies. Two authors independently conducted the literature search, examined the studies, and evaluated the eligibility criteria. Any disagreements or uncertainties were resolved by the corresponding author. The search strategy followed the PICO process (Population, Intervention, Comparison, and Outcome) and Emtree was used to select relevant keywords.Results: Of 220 articles identified from the literature research, five articles were included in the study, of which three studies lately were retracted. The remaining studies included 1229 participants, with 209 receiving SGLT-2 inhibitors and 1090 not receiving them. All of the participants were diabetic patients admitted with acute myocardial infarction (AMI), undergoing percutaneous coronary intervention (PCI). The results demonstrated that the use of SGLT-2 inhibitors is associated with lower troponin levels, and higher rates of ST resolution. The results of the studies also showed smaller infarct sizes, lower inflammatory biomarkers and improved left ventricular function at discharge among SGLT-2 inhibitor users.Conclusion: In line with in vivo and ex vivo findings, the results of this systematic review supported benefits of SGLT-2 inhibitors in IRI through reducing infarct size and inflammatory biomarkers. However, further clinical trials are warranted to provide robust evidence.
Collapse
Affiliation(s)
- Hossein Behzad
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hila Asham
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samineh Beheshtirouy
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sina Mashayekhi
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Negin Jafari
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Taher Entezari-Maleki
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
2
|
Bhalraam U, Veerni RB, Paddock S, Meng J, Piepoli M, López-Fernández T, Tsampasian V, Vassiliou VS. Impact of sodium-glucose cotransporter-2 inhibitors on heart failure outcomes in cancer patients and survivors: a systematic review and meta-analysis. Eur J Prev Cardiol 2025:zwaf026. [PMID: 40044419 DOI: 10.1093/eurjpc/zwaf026] [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: 09/07/2024] [Revised: 11/30/2024] [Accepted: 01/28/2025] [Indexed: 03/20/2025]
Abstract
AIMS Sodium-glucose cotransporter-2 inhibitors (SGLT2i) are recognized for their cardiovascular benefits. This systematic review and meta-analysis evaluated the impact of SGLT2i on heart failure (HF) outcomes in cancer patients and survivors, focusing on HF hospitalization and new HF diagnoses. METHODS AND RESULTS A comprehensive search of PubMed, MEDLINE, and Embase via Ovid, and the Cochrane Library was conducted up to 5 June 2024, focusing on studies involving cancer patients and survivors treated with SGLT2i. The search criterion used was [(SGLT2) OR (Sodium glucose cotransporter 2 inhibitors) OR (canagliflozin) OR (dapagliflozin) OR (empagliflozin) OR (ertugliflozin) AND (cancer)]. The primary outcomes assessed were HF hospitalization and new HF diagnoses. The search yielded 1880 studies, from which 13 studies encompassing 88 273 patients were included. SGLT2i use reduced HF hospitalizations by 51% (RR 0.49, 95% CI 0.36-0.66, I² = 28%, P < 0.01) and new HF diagnoses by 71% (RR 0.29, 95% CI 0.10-0.87, I² = 71%). Multi-variate meta-regression analysis suggested that among breast cancer populations, studies with ≥50% of patients on anthracyclines exhibited a 99% reduction in HF hospitalization risk compared with similar studies that included <50% of patients on anthracyclines (RR 0.0085, 95% CI: 0.0001-0.2645, P = 0.0081). CONCLUSION SGLT2i significantly lower the risk of HF hospitalization and new HF diagnoses among cancer patients and survivors, with particularly pronounced benefits in breast cancer patients receiving anthracycline-based chemotherapy. These findings support the need for prospective trials to further investigate the integration of SGLT2i into cancer patient management to enhance cardiovascular outcomes.
Collapse
Affiliation(s)
- U Bhalraam
- Department of Medicine, Norwich Medical School, University of East Anglia, Norwich, UK
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
| | - Rathna B Veerni
- Department of Medicine, Ninewells Hospital and Medical School, Dundee, UK
| | - Sophie Paddock
- Department of Medicine, Norwich Medical School, University of East Anglia, Norwich, UK
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
| | - James Meng
- Department of Medicine, Norwich Medical School, University of East Anglia, Norwich, UK
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
| | - Massimo Piepoli
- University Cardiology Department, IRCCS Policlinico San Donato, Via Morandi 30, 20097 San Donato Milanese, Milan, Italy
- Department of Biomedical Sciences for Health, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Teresa López-Fernández
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
- Cardiology Department, Quiron Pozuelo University Hospital, Madrid, Spain
| | - Vasiliki Tsampasian
- Department of Medicine, Norwich Medical School, University of East Anglia, Norwich, UK
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
| | - Vassilios S Vassiliou
- Department of Medicine, Norwich Medical School, University of East Anglia, Norwich, UK
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
| |
Collapse
|
3
|
Ansari HUH, Samad MA, Mahboob E, Zulfiqar E, Qazi SU, Ahsan A, Ahmed M, Ahmed F, Ahmed R, Ali S, Alam M, Rana JS, Fonarow GC. Sodium-glucose cotransporter 2 inhibitors in patients with type 2 diabetes and myocardial infarction undergoing percutaneous coronary intervention: A systematic review and meta-analysis. Am J Prev Cardiol 2025; 21:100927. [PMID: 39867488 PMCID: PMC11757226 DOI: 10.1016/j.ajpc.2024.100927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/21/2024] [Accepted: 12/30/2024] [Indexed: 01/28/2025] Open
Abstract
Background Sodium-glucose cotransporter 2 inhibitors (SGLT2i) have shown benefits in improving cardiovascular (CV) outcomes in patients with heart failure (HF) and may mitigate symptom progression in myocardial infarction (MI). However, their effectiveness in patients with type 2 diabetes and MI undergoing percutaneous coronary intervention (PCI) is unclear. Methods To identify eligible studies, a comprehensive search of electronic databases, PubMed, Cochrane Library, Scopus and Embase, was conducted from inception until May 2024. Results were presented as risk ratios (RR) and their corresponding 95 % confidence intervals (CIs). Results Our analysis included 8 observational studies comprising 24,229 patients. The results indicated that SGLT2i with PCI was associated with a significantly reduced risk of all-cause death (RR=0.61; 95 % CI=0.54 to 0.68), CV death (RR=0.46; 95 % CI=0.22 to 0.94), major adverse cardiovascular events (RR=0.80;95 % CI: 0.66 to 0.96), HF-related hospitalizations (RR=0.63; 95 % CI=0.44 to 0.90), stroke (RR=0.77; 95 % CI: 0.62 to 0.96) and acute kidney injury (RR=0.46; 95 % CI: 0.25 to 0.84) compared to PCI without SGLT2i use. However, the risk of revascularization remained comparable between the groups. Conclusion Our study demonstrates that SGLT2i with PCI in patients with type 2 diabetes and MI are associated with improved CV outcomes compared to PCI without SGLT2i use. Randomized controlled trials are required to confirm the improvement in outcomes with SGLT2i therapy combined with PCI in patients with MI and diabetes.
Collapse
Affiliation(s)
| | | | - Eman Mahboob
- Dow University of Health Sciences, Karachi, Pakistan
| | | | | | - Areeba Ahsan
- Foundation University Medical College, Islamabad, Pakistan
| | | | - Faizan Ahmed
- Division of Cardiology, Duke University Hospital, Durham, NC, USA
| | - Raheel Ahmed
- Department of Cardiology, Royal Brompton Hospital, London, UK
- National Heart and Lung Institute, Imperial College London, UK
| | - Shafaqat Ali
- Department of Cardiology, Louisiana State University, Shreveport, USA
| | - Mahboob Alam
- Department of Cardiology, Baylor College of Medicine, Houston, TX, USA
| | - Jamal S. Rana
- Division of Cardiology, Kaiser Permanente Northern California, Oakland, CA, USA
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Gregg C. Fonarow
- Ahmanson-UCLA Cardiomyopathy Center, Division of Cardiology, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
4
|
Jia Q, Zuo A, Song H, Zhang C, Fu X, Hu K, An F. Effects of sodium-glucose cotransporter-2 inhibitors in myocardial infarction patients: A systematic review and meta-analysis. Diabetes Obes Metab 2025; 27:1276-1286. [PMID: 39691984 DOI: 10.1111/dom.16122] [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: 10/06/2024] [Revised: 11/17/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024]
Abstract
AIMS Sodium-glucose cotransporter-2 (SGLT2) inhibitors are known to improve cardiovascular outcomes in individuals with heart failure (HF), type 2 diabetes mellitus (T2DM) and chronic kidney disease (CKD). However, their efficacy following myocardial infarction (MI) remains unclear. MATERIALS AND METHODS A systematic search was conducted using PubMed, Embase, Cochrane Library, Web of Science and ClinicalTrials.gov. Primary outcomes included hospitalization for heart failure (HHF), cardiovascular (CV) death, a composite of HHF or CV death, all-cause death, major cardiovascular events (MACE), recurrent MI, severe arrhythmia, renal injury and stroke. Secondary outcomes targeted improvements in left ventricular ejection fraction (LVEF) and left ventricular end-diastolic volume (LVEDV). RESULTS Thirteen studies comprising 22 370 patients were included. Meta-analysis revealed that SGLT2 inhibitors reduced HHF (RR 0.69, 95% CI 0.61 to 0.78, p < 0.001), combined HHF or CV death (RR 0.87, 95% CI 0.77 to 0.99, p = 0.028), all-cause mortality (RR 0.82, 95% CI 0.73 to 0.93, p = 0.002), MACE (RR 0.68, 95% CI 0.53 to 0.88, p = 0.004), recurrent MI (RR 0.81, 95% CI 0.69 to 0.94, p = 0.007), severe arrhythmia (RR 0.54, 95% CI 0.34 to 0.85, p = 0.009) and renal injury (RR 0.68, 95% CI 0.53 to 0.87, p = 0.002). Improvement in LVEF (MD 3.96%, 95% CI 2.52 to 5.40; p < 0.001) and LVEDV (MD -5.52 mL, 95% CI -10.21 to -0.83; p = 0.021) was notably greater in the SGLT2 inhibitors group. CONCLUSIONS In post-MI patients, we first found that SGLT2 inhibitors significantly lowered the risk of HHF, combined CV death or HHF, all-cause death, MACE, recurrent MI, severe arrhythmias and renal injury. Additionally, SGLT2 inhibitors improved LVEF and LVEDV.
Collapse
Affiliation(s)
- Qiufeng Jia
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Ankai Zuo
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hui Song
- Department of Cardiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Chengrui Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiangrui Fu
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Keqing Hu
- Department of Cardiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Fengshuang An
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
5
|
Khan U, Amin AM, Mohamed Taha A, Khlidj Y, M. AlBarakat M, Elewidi M, Abuelazm M, Turkmani M, Abdelazeem B, Laeeq R. The effect of sodium-glucose co-transporter 2 inhibitors on clinical outcomes after acute myocardial infarction: a systematic review and meta-analysis of randomized controlled trials. Future Cardiol 2025; 21:177-190. [PMID: 39939290 PMCID: PMC11875467 DOI: 10.1080/14796678.2025.2464449] [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: 10/21/2024] [Accepted: 02/05/2025] [Indexed: 02/14/2025] Open
Abstract
INTRODUCTION Sodium-glucose cotransporter 2 inhibitors (SGLT2is) reduce cardiovascular events, especially in diabetic patients. However, the cardioprotective effects of early SGLT2i administration following acute myocardial infarction (AMI) remain unclear. OBJECTIVE This study aims to investigate the impact of SGLT2is on clinical outcomes in patients post-AMI. METHODS A comprehensive search was conducted in PubMed, CENTRAL, WOS, Scopus, and EMBASE up to April 2024. Risk ratio (RR) was used for dichotomous outcomes and mean difference (MD) for continuous outcomes, with 95% confidence intervals (CI). RESULTS Seven studies with 11,407 patients were included. SGLT2is did not significantly reduce the incidence of major adverse cardiovascular events (MACE) (RR = 0.94, 95% CI [0.68, 1.29], p = 0.69), all-cause mortality (RR = 1.01, 95% CI [0.84, 1.21], p = 0.93), or stroke (RR = 0.61, 95% CI [0.29,1.28], p = 0.19). However, SGLT2is significantly reduced the risk of heart failure (RR = 0.76, 95% CI [0.63, 0.91], p < 0.01) and improved left ventricular ejection fraction (MD = 1.86, 95% CI [1.58, 2.14], p < 0.01). CONCLUSION In post-AMI patients, SGLT2is do not significantly affect MACE or mortality but are associated with reduced heart failure risk and improved ejection fraction. PROTOCOL REGISTRATION PROSPERO identifier number: CRD42024506806.
Collapse
Affiliation(s)
- Ubaid Khan
- Division of Cardiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | - Yehya Khlidj
- Faculty of Medicine, University of Algiers, Algiers, Algeria
| | - Majd M. AlBarakat
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | | | | | - Mustafa Turkmani
- Faculty of Medicine, Michigan State University, East Lansing, MI, USA
- Department of Internal Medicine, McLaren Health Care, Oakland, MI, USA
| | - Basel Abdelazeem
- Department of Cardiology, West Virginia University Morgantown, West Virginia, USA
| | - Rida Laeeq
- Department of Cardiology, West Virginia University Morgantown, West Virginia, USA
| |
Collapse
|
6
|
Kianfar T, Hasan R, Azizi Y, Ramezani F. The Effect of Dapagliflozin on Heart Function in Animal Models of Cardiac Ischemia, A Systematic Review and Meta-analysis. Curr Rev Clin Exp Pharmacol 2025; 20:72-88. [PMID: 39882703 DOI: 10.2174/0127724328313815240723044625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/01/2024] [Accepted: 06/11/2024] [Indexed: 01/31/2025]
Abstract
INTRODUCTION In this study, a meta-analysis was conducted to investigate the therapeutic effect of Dapagliflozin (DAPA) on animals suffering from myocardial ischemia reperfusion compared to the group that did not receive treatment. METHODS According to the inclusion and exclusion criteria two researchers performed the primary and secondary screening based on the title abstract and full text. After data extraction, meta-analysis was performed using STATA software. Standardized mean differences were used to analyze the results of the reported studies. Subgroup analysis and quality control of articles were also conducted. RESULTS A total of 21 separate experiments showed that DAPA increased mean fractional shortening (%FS) and ejection fraction (%EF) compared to the untreated animals. A significant reduction in the weight and size of the infarcted area and significant increases in dp/dt+, dp/dt-, left ventricular end-systolic internal dimensions (LVIDs), left ventricular end-diastolic internal dimensions (LVIDd), Volume systole and Volume diastole were observed in treated animals. CONCLUSION DAPA has the potential to become a candidate for the treatment of post-ischemic heart damage, pending animal and human studies to validate this.
Collapse
Affiliation(s)
- Tina Kianfar
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Raquibul Hasan
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA
- Department of Biomedical Sciences, School of Medicine, Mercer University, Macon, GA, USA
| | - Yaser Azizi
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ramezani
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
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
|
8
|
Prosperi S, D’Amato A, Labbro Francia A, Monosilio S, Cestiè C, Marek Iannucci S, Netti L, Angotti D, Filomena D, Mariani MV, Myftari V, Germanò R, Cimino S, Mancone M, Badagliacca R, Maestrini V, Severino P, Vizza CD. Sodium-Glucose Cotransporter 2 Inhibitor Therapy in Different Scenarios of Heart Failure: An Overview of the Current Literature. Int J Mol Sci 2024; 25:11458. [PMID: 39519011 PMCID: PMC11546829 DOI: 10.3390/ijms252111458] [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: 08/31/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Heart failure (HF) is a complex syndrome that requires tailored and patient-centered treatment. Sodium-glucose cotransporter 2 inhibitors (SGLT2is) constitute one of the four pillars of the medical treatment of HF. However, the 2023 ESC guidelines treat HF as a single entity without making clear distinctions in phenotypes according to etiology. This creates a "gap in knowledge", causing much debate about the applicability of these drugs in peculiar clinical settings that are etiological and/or predisposing clinical conditions for HF. Furthermore, considering the variety of etiologies and different pathophysiological backgrounds of HF, one might question whether the use of SGLT2is is equally beneficial in all types of HF and whether certain drug-related properties may be exploited in different contexts. For example, SGLT2is can improve the metabolic and inflammatory state, which is fundamental in ischemic heart disease. Anti-inflammatory power can also play a paramount role in myocarditis or cardiotoxicity, while improving the congestive state and reducing filling pressure may be even more fundamental in restrictive heart disease or advanced heart disease. This review aims to gather the evidence currently present in the literature concerning the advantages or the disadvantages of using these drugs in these particular clinical settings, with the goal being an optimized and highly personalized treatment for HF.
Collapse
Affiliation(s)
- Silvia Prosperi
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Andrea D’Amato
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
- Department of Cardiology, Ospedale Fabrizio Spaziani, 03100 Frosinone, Italy
| | - Aurora Labbro Francia
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Sara Monosilio
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Claudia Cestiè
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Stefanie Marek Iannucci
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Lucrezia Netti
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Danilo Angotti
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Domenico Filomena
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Marco Valerio Mariani
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Vincenzo Myftari
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Rosanna Germanò
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Sara Cimino
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Massimo Mancone
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Roberto Badagliacca
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Viviana Maestrini
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Paolo Severino
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Carmine Dario Vizza
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| |
Collapse
|
9
|
Mouffokes A, Soliman Y, Amer BE, Umar TP, Gonnah AR, Ellabban MH, Abdelazeem B. The effect of Empagliflozin on echocardiographic parameters in diabetic patients after acute myocardial infarction: A systematic review and meta-analysis with trial sequential analysis. Ir J Med Sci 2024; 193:2223-2238. [PMID: 38958683 DOI: 10.1007/s11845-024-03744-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024]
Abstract
Patients with diabetes mellitus (DM) are at higher risk of cardiovascular events, particularly acute myocardial infarction (MI). Sodium-glucose cotransporter 2 inhibitors (SGLT2i) can improve cardiac outcomes among heart failure individuals, however, the effects on acute myocardial infarction remain unclear. This meta-analysis investigates the impact of empagliflozin in diabetic patients following acute myocardial infarction. We comprehensively searched PubMed, Scopus, Cochrane, and Web of Science through August 10th, 2023. We included studies comparing empagliflozin versus placebo in diabetes patients with acute myocardial infarction. We used Revman to report the data as mean difference (MD) and 95% confidence interval (CI), and our effect size with a random effects model. Additionally, we performed Trial Sequential Analysis (TSA) to test the robustness of the results. The study protocol was published on PROSPERO with ID: CRD42023447733. Five studies with a total of 751 patients were included in our analysis. Empagliflozin was effective to improve LVEF% (MD: 1.80, 95% CI [0.50, 3.10], p = 0.007), left ventricular end-diastolic volume (LVEDV) (MD: -9.93, 95% CI [-16.07, -3.80], p = 0.002), and left ventricular end-systolic volume (LVESV) (MD: -7.91, 95% CI [-11.93, -3.88], p = 0.0001). However, there was no difference between empagliflozin and placebo groups in terms of NT-pro BNP (MD: - 136.59, 95% CI [-293.43, 20.25], p = 0.09), and HbA1c (MD: -0.72, 95% CI [-1.73, 0.29], p = 0.16). Additionally, empagliflozin did not prevent hospitalization due to heart failure (RR: 0.59, 95% CI [0.16, 2.24], p = 0.44, I-squared = 0%), and mortality (RR: 1.34, 95% CI [0.15,11.90], p = 0.79, I-squared = 25%). Empagliflozin initiation in diabetic patients following acute MI may improve echocardiographic parameters. However, empagliflozin might not be effective in heart failure prevention and optimal glycemic control in this patient population. Further large-scale trials are warranted to ascertain our findings.
Collapse
Affiliation(s)
- Adel Mouffokes
- Faculty of Medicine, University of Oran Ahmed Ben Bella 1, Oran, Algeria.
| | | | | | - Tungki Pratama Umar
- Faculty of Medicine, Sriwijaya University, Palembang, Indonesia
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, London, UK
| | | | | | - Basel Abdelazeem
- Department of Cardiology, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
10
|
Almalki A, Arjun S, Harding I, Jasem H, Kolatsi-Joannou M, Jafree DJ, Pomeranz G, Long DA, Yellon DM, Bell RM. SGLT1 contributes to glucose-mediated exacerbation of ischemia-reperfusion injury in ex vivo rat heart. Basic Res Cardiol 2024; 119:733-749. [PMID: 39088085 PMCID: PMC11461679 DOI: 10.1007/s00395-024-01071-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: 11/21/2023] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
Hyperglycaemia is common during acute coronary syndromes (ACS) irrespective of diabetic status and portends excess infarct size and mortality, but the mechanisms underlying this effect are poorly understood. We hypothesized that sodium/glucose linked transporter-1 (SGLT1) might contribute to the effect of high-glucose during ACS and examined this using an ex-vivo rodent heart model of ischaemia-reperfusion injury. Langendorff-perfused rat hearts were subjected to 35 min ischemia and 2 h reperfusion, with variable glucose and reciprocal mannitol given during reperfusion in the presence of pharmacological inhibitors of SGLT1. Myocardial SGLT1 expression was determined in rat by rtPCR, RNAscope and immunohistochemistry, as well as in human by single-cell transcriptomic analysis. High glucose in non-diabetic rat heart exacerbated reperfusion injury, significantly increasing infarct size from 45 ± 3 to 65 ± 4% at 11-22 mmol/L glucose, respectively (p < 0.01), an association absent in diabetic heart (32 ± 1-37 ± 5%, p = NS). Rat heart expressed SGLT1 RNA and protein in vascular endothelium and cardiomyocytes, with similar expression found in human myocardium by single-nucleus RNA-sequencing. Rat SGLT1 expression was significantly reduced in diabetic versus non-diabetic heart (0.608 ± 0.08 compared with 1.116 ± 0.13 probe/nuclei, p < 0.01). Pharmacological inhibitors phlorizin, canagliflozin or mizagliflozoin in non-diabetic heart revealed that blockade of SGLT1 but not SGLT2, abrogated glucose-mediated excess reperfusion injury. Elevated glucose is injurious to the rat heart during reperfusion, exacerbating myocardial infarction in non-diabetic heart, whereas the diabetic heart is resistant to raised glucose, a finding which may be explained by lower myocardial SGLT1 expression. SGLT1 is expressed in vascular endothelium and cardiomyocytes and inhibiting SGLT1 abrogates excess glucose-mediated infarction. These data highlight SGLT1 as a potential clinical translational target to improve morbidity/mortality outcomes in hyperglycemic ACS patients.
Collapse
Affiliation(s)
- Alhanoof Almalki
- Hatter Cardiovascular Institute, Institute for Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Sapna Arjun
- Hatter Cardiovascular Institute, Institute for Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Idris Harding
- Hatter Cardiovascular Institute, Institute for Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Hussain Jasem
- Hatter Cardiovascular Institute, Institute for Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Maria Kolatsi-Joannou
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Daniyal J Jafree
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
- UCL Centre for Kidney and Bladder Health, London, UK
- UCL MB/PhD Programme, Faculty of Medical Sciences, University College London, London, UK
| | - Gideon Pomeranz
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
- UCL Centre for Kidney and Bladder Health, London, UK
| | - David A Long
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
- UCL Centre for Kidney and Bladder Health, London, UK
| | - Derek M Yellon
- Hatter Cardiovascular Institute, Institute for Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Robert M Bell
- Hatter Cardiovascular Institute, Institute for Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK.
| |
Collapse
|
11
|
Hayashida T, Kuroko Y, Shimizu S, Akiyama T, Suezawa T, Kioka Y, Kotani Y, Shishido T, Kasahara S. Effects of dapagliflozin on myoglobin efflux from cardiomyocyte during myocardial ischemia/reperfusion in anesthetized rats. Sci Rep 2024; 14:16337. [PMID: 39014025 PMCID: PMC11253006 DOI: 10.1038/s41598-024-67195-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/09/2024] [Indexed: 07/18/2024] Open
Abstract
It has been suggested that sodium-glucose cotransporter 2 (SGLT2) inhibitors have cardioprotective effects during myocardial ischemia/reperfusion (I/R) independent of glucose-lowering action. However, the effects of SGLT2 inhibitors on structural damage to cardiomyocytes in the ischemic region during I/R remain unknown. We applied a microdialysis technique to the heart of anesthetized rats and investigated the effects of an SGLT2 inhibitor, dapagliflozin, on myocardial interstitial myoglobin levels in the ischemic region during coronary occlusion followed by reperfusion. Dapagliflozin was administered systemically (40 μg/body iv) or locally via a dialysis probe (100 μM and 1 mM) 30 min before coronary occlusion. In the vehicle group, coronary occlusion increased the dialysate myoglobin concentration in the ischemic region. Reperfusion further increased the dialysate myoglobin concentration. Intravenous administration of dapagliflozin reduced dialysate myoglobin concentration during ischemia and at 0-15 min after reperfusion, but local administration (100 μM and 1 mM) did not. Therefore, acute systemic administration of dapagliflozin prior to ischemia has cardioprotective effects on structural damage during I/R.
Collapse
Affiliation(s)
- Tomohiro Hayashida
- Department of Cardiovascular Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
- Department of Cardiovascular Surgery, Fukuyama City Hospital, Fukuyama, Hiroshima, 721-8511, Japan
| | - Yosuke Kuroko
- Department of Cardiovascular Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences and Okayama University Hospital, Okayama, 700-8558, Japan
| | - Shuji Shimizu
- Department of Cardiovascular Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan.
- Department of Research Promotion and Management, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan.
| | - Tsuyoshi Akiyama
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center, Suita, Osaka, 564-8565, Japan
| | - Takanori Suezawa
- Department of Cardiovascular Surgery, Fukuyama City Hospital, Fukuyama, Hiroshima, 721-8511, Japan
| | - Yukio Kioka
- Department of Cardiovascular Surgery, Fukuyama City Hospital, Fukuyama, Hiroshima, 721-8511, Japan
| | - Yasuhiro Kotani
- Department of Cardiovascular Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences and Okayama University Hospital, Okayama, 700-8558, Japan
| | - Toshiaki Shishido
- Department of Research Promotion and Management, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Shingo Kasahara
- Department of Cardiovascular Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences and Okayama University Hospital, Okayama, 700-8558, Japan
| |
Collapse
|
12
|
Ionică LN, Lința AV, Bătrîn AD, Hâncu IM, Lolescu BM, Dănilă MD, Petrescu L, Mozoș IM, Sturza A, Muntean DM. The Off-Target Cardioprotective Mechanisms of Sodium-Glucose Cotransporter 2 Inhibitors: An Overview. Int J Mol Sci 2024; 25:7711. [PMID: 39062954 PMCID: PMC11277154 DOI: 10.3390/ijms25147711] [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: 06/20/2024] [Revised: 07/08/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Sodium-glucose cotransporter 2 inhibitors (SGLT2i), a novel class of glucose-lowering drugs, have revolutionized the management of heart failure with reduced and preserved ejection fraction, regardless of the presence of diabetes, and are currently incorporated in the heart failure guidelines. While these drugs have consistently demonstrated their ability to decrease heart failure hospitalizations in several landmark clinical trials, their cardioprotective effects are far from having been completely elucidated. In the past decade, a growing body of experimental research has sought to address the molecular and cellular mechanisms of SGLT2i in order to provide a better understanding of the off-target acute and chronic cardiac benefits, beyond the on-target renal effect responsible for blood glucose reduction. The present narrative review addresses the direct cardioprotective effects of SGLT2i, delving into the off-target mechanisms of the drugs currently approved for heart failure therapy, and provides insights into future perspectives.
Collapse
Affiliation(s)
- Loredana N. Ionică
- Department of Internal Medicine-Medical Semiotics, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania;
- Doctoral School Medicine-Pharmacy, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq., No. 2, 300041 Timișoara, Romania; (A.V.L.); (A.D.B.); (I.M.H.); (B.M.L.)
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (M.D.D.); (L.P.); (I.M.M.); (D.M.M.)
| | - Adina V. Lința
- Doctoral School Medicine-Pharmacy, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq., No. 2, 300041 Timișoara, Romania; (A.V.L.); (A.D.B.); (I.M.H.); (B.M.L.)
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (M.D.D.); (L.P.); (I.M.M.); (D.M.M.)
- Department of Functional Sciences-Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Alina D. Bătrîn
- Doctoral School Medicine-Pharmacy, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq., No. 2, 300041 Timișoara, Romania; (A.V.L.); (A.D.B.); (I.M.H.); (B.M.L.)
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (M.D.D.); (L.P.); (I.M.M.); (D.M.M.)
| | - Iasmina M. Hâncu
- Doctoral School Medicine-Pharmacy, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq., No. 2, 300041 Timișoara, Romania; (A.V.L.); (A.D.B.); (I.M.H.); (B.M.L.)
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (M.D.D.); (L.P.); (I.M.M.); (D.M.M.)
- Department of Functional Sciences-Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Bogdan M. Lolescu
- Doctoral School Medicine-Pharmacy, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq., No. 2, 300041 Timișoara, Romania; (A.V.L.); (A.D.B.); (I.M.H.); (B.M.L.)
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (M.D.D.); (L.P.); (I.M.M.); (D.M.M.)
| | - Maria D. Dănilă
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (M.D.D.); (L.P.); (I.M.M.); (D.M.M.)
- Department of Functional Sciences-Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Lucian Petrescu
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (M.D.D.); (L.P.); (I.M.M.); (D.M.M.)
| | - Ioana M. Mozoș
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (M.D.D.); (L.P.); (I.M.M.); (D.M.M.)
- Department of Functional Sciences-Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Adrian Sturza
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (M.D.D.); (L.P.); (I.M.M.); (D.M.M.)
- Department of Functional Sciences-Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Danina M. Muntean
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (M.D.D.); (L.P.); (I.M.M.); (D.M.M.)
- Department of Functional Sciences-Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania
| |
Collapse
|
13
|
Aziri B, Begic E, Stanetic B, Mladenovic Z, Kovacevic-Preradovic T. Sodium-glucose cotransporter-2 inhibitors: a swinging pendulum in the treatment of acute myocardial infarction. Minerva Cardiol Angiol 2024; 72:237-250. [PMID: 37161919 DOI: 10.23736/s2724-5683.22.06200-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
INTRODUCTION Sodium-glucose cotransporter-2 (SGLT2) inhibitors are integral in treating patients with heart failure, regardless of the existence of diabetes mellitus. In light of their benefits on the heart muscle, the question of their effect on acute coronary syndrome is raised, and a hypothesis as to whether they can be implemented in its treatment is proposed. The aim of the article was to indicate the potential of using SGLT2 inhibitors in the treatment of myocardial infarction (MI). EVIDENCE ACQUISITION A PubMed search for articles published between October 2017 and May 2022 was conducted using the following keywords: "SGLT2 inhibitors," "Acute Coronary Syndrome," "Treatment," "Prognosis." Reference lists of identified articles were searched for further articles. EVIDENCE SYNTHESIS Reports from clinical trials and animal studies thus far investigating mechanistic pathways of SGLT2 inhibitors' effect in relation to acute myocardial infarction were interplayed to extract relevant findings and analyze the safety of this therapy in acute coronary syndrome (ACS) patients. CONCLUSIONS SGLT2 inhibitors indicate beneficial effects in acute cardiovascular incident by various mechanisms, and early initiation of therapy may improve outcomes for AMI survivors.
Collapse
Affiliation(s)
- Buena Aziri
- Department of Pharmacology, Sarajevo Medical School, Sarajevo School of Science and Technology, Sarajevo, Bosnia and Herzegovina
| | - Edin Begic
- Department of Pharmacology, Sarajevo Medical School, Sarajevo School of Science and Technology, Sarajevo, Bosnia and Herzegovina -
- Department of Cardiology, Prim. Dr. Abdulah Nakas General Hospital, Sarajevo, Bosnia and Herzegovina
| | - Bojan Stanetic
- Department of Cardiology, University Clinical Centre of the Republic of Srpska, Banja Luka, Bosnia and Herzegovina
- Department of Internal Medicine, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Zorica Mladenovic
- Department of Cardiology, Military Medical Academy, University of Defense, Belgrade, Serbia
| | - Tamara Kovacevic-Preradovic
- Department of Cardiology, University Clinical Centre of the Republic of Srpska, Banja Luka, Bosnia and Herzegovina
- Department of Internal Medicine, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| |
Collapse
|
14
|
Egashira T, Ichinomiya T, Yokoyama A, Matsumoto S, Higashijima U, Sekino M, Murata H, Yoshitomi O, Sato S, Hara T. Cardioprotective Effects of Sodium-Glucose Cotransporter Subtype Inhibition on Ischemic and Pharmacological Preconditioning. Cureus 2024; 16:e59757. [PMID: 38841006 PMCID: PMC11152766 DOI: 10.7759/cureus.59757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Sodium-glucose cotransporter (SGLT) 2 inhibitors partially inhibit SGLT1 expression; however, whether a clinical dose of SGLT2 inhibitor abrogates ischemic preconditioning (IPC) is unknown, and the pharmacological cardioprotective effect under SGLT1 inhibition has not been examined. In this study, we investigated whether a clinical dose of tofogliflozin abrogates IPC and whether pharmacological preconditioning with olprinone has cardioprotective effects under SGLT1 inhibition. METHODS Male Wistar rats were divided into seven groups (seven rats per group) and subjected to the following treatments before inducing ischemia/reperfusion (I/R; 30 minutes of coronary artery occlusion followed by 120 minutes of reperfusion): saline infusion control treatment (Con); ischemic preconditioning (IPC); IPC after phlorizin infusion (IPC+Phl); IPC after low-dose tofogliflozin infusion (IPC+L-Tof); IPC after high-dose tofogliflozin infusion (IPC+H-Tof); olprinone infusion (Olp); and Olp infusion after phlorizin infusion (Olp+Phl). RESULTS The infarct size was significantly decreased in the IPC group, but not in the IPC+Phl group. In contrast, the infarct size decreased in the IPC+L-Tof and IPC+H-Tof groups. Additionally, Olp reduced the infarct size, and the effect was preserved in Olp+Phl groups. Phosphorylated AMP-activated protein kinase (AMPK) expression was lower in the IPC+Phl group compared to that in the IPC group. CONCLUSION The cardioprotective effect of IPC was attenuated by strong SGLT1 inhibition, but the effect was preserved under a clinical dose of highly selective SGLT2 inhibitor. Olprinone exerts a cardioprotective effect even under strong SGLT1 inhibition.
Collapse
Affiliation(s)
- Takashi Egashira
- Department of Anesthesiology and Intensive Care Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, JPN
| | - Taiga Ichinomiya
- Department of Anesthesiology and Intensive Care Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, JPN
| | - Akihiro Yokoyama
- Department of Anesthesiology and Intensive Care Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, JPN
| | - Sojiro Matsumoto
- Department of Anesthesiology and Intensive Care Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, JPN
| | - Ushio Higashijima
- Department of Anesthesiology and Intensive Care Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, JPN
| | - Motohiro Sekino
- Department of Anesthesiology and Intensive Care Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, JPN
| | - Hiroaki Murata
- Department of Anesthesiology and Intensive Care Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, JPN
| | - Osamu Yoshitomi
- Department of Anesthesiology and Intensive Care Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, JPN
| | - Shuntaro Sato
- Clinical Research Center, Nagasaki University Hospital, Nagasaki, JPN
| | - Tetsuya Hara
- Department of Anesthesiology and Intensive Care Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, JPN
| |
Collapse
|
15
|
Alsereidi FR, Khashim Z, Marzook H, Gupta A, Al-Rawi AM, Ramadan MM, Saleh MA. Targeting inflammatory signaling pathways with SGLT2 inhibitors: Insights into cardiovascular health and cardiac cell improvement. Curr Probl Cardiol 2024; 49:102524. [PMID: 38492622 DOI: 10.1016/j.cpcardiol.2024.102524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 03/13/2024] [Indexed: 03/18/2024]
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors have attracted significant attention for their broader therapeutic impact beyond simply controlling blood sugar levels, particularly in their ability to influence inflammatory pathways. This review delves into the anti-inflammatory properties of SGLT2 inhibitors, with a specific focus on canagliflozin, empagliflozin, and dapagliflozin. One of the key mechanisms through which SGLT2 inhibitors exert their anti-inflammatory effects is by activating AMP-activated protein kinase (AMPK), a crucial regulator of both cellular energy balance and inflammation. Activation of AMPK by these inhibitors leads to the suppression of pro-inflammatory pathways and a decrease in inflammatory mediators. Notably, SGLT2 inhibitors have demonstrated the ability to inhibit the release of cytokines in an AMPK-dependent manner, underscoring their direct influence on inflammatory signaling. Beyond AMPK activation, SGLT2 inhibitors also modulate several other inflammatory pathways, including the NLRP3 inflammasome, expression of Toll-like receptor 4 (TLR-4), and activation of NF-κB (Nuclear factor kappa B). This multifaceted approach contributes to their efficacy in reducing inflammation and managing associated complications in conditions such as diabetes and cardiovascular disorders. Several human and animal studies provide support for the anti-inflammatory effects of SGLT2 inhibitors, demonstrating protective effects on various cardiac cells. Additionally, these inhibitors exhibit direct anti-inflammatory effects by modulating immune cells. Overall, SGLT2 inhibitors emerge as promising therapeutic agents for targeting inflammation in a range of pathological conditions. Further research, particularly focusing on the molecular-level pathways of inflammation, is necessary to fully understand their mechanisms of action and optimize their therapeutic potential in inflammatory diseases.
Collapse
Affiliation(s)
- Fatmah R Alsereidi
- Cardiovascular Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Zenith Khashim
- Department of Physiology and Biomedical Engineering, Mayo Clinic Rochester, Rochester, MN, United States
| | - Hezlin Marzook
- Cardiovascular Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Anamika Gupta
- Cardiovascular Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Ahmed M Al-Rawi
- Cardiovascular Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mahmoud M Ramadan
- Cardiovascular Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates; Department of Cardiology, Faculty of Medicine, Mansoura University, 35516 Egypt
| | - Mohamed A Saleh
- Cardiovascular Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516 Egypt.
| |
Collapse
|
16
|
Barrère-Lemaire S, Vincent A, Jorgensen C, Piot C, Nargeot J, Djouad F. Mesenchymal stromal cells for improvement of cardiac function following acute myocardial infarction: a matter of timing. Physiol Rev 2024; 104:659-725. [PMID: 37589393 DOI: 10.1152/physrev.00009.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/05/2023] [Accepted: 08/16/2023] [Indexed: 08/18/2023] Open
Abstract
Acute myocardial infarction (AMI) is the leading cause of cardiovascular death and remains the most common cause of heart failure. Reopening of the occluded artery, i.e., reperfusion, is the only way to save the myocardium. However, the expected benefits of reducing infarct size are disappointing due to the reperfusion paradox, which also induces specific cell death. These ischemia-reperfusion (I/R) lesions can account for up to 50% of final infarct size, a major determinant for both mortality and the risk of heart failure (morbidity). In this review, we provide a detailed description of the cell death and inflammation mechanisms as features of I/R injury and cardioprotective strategies such as ischemic postconditioning as well as their underlying mechanisms. Due to their biological properties, the use of mesenchymal stromal/stem cells (MSCs) has been considered a potential therapeutic approach in AMI. Despite promising results and evidence of safety in preclinical studies using MSCs, the effects reported in clinical trials are not conclusive and even inconsistent. These discrepancies were attributed to many parameters such as donor age, in vitro culture, and storage time as well as injection time window after AMI, which alter MSC therapeutic properties. In the context of AMI, future directions will be to generate MSCs with enhanced properties to limit cell death in myocardial tissue and thereby reduce infarct size and improve the healing phase to increase postinfarct myocardial performance.
Collapse
Affiliation(s)
- Stéphanie Barrère-Lemaire
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Anne Vincent
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Christian Jorgensen
- Institute of Regenerative Medicine and Biotherapies, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Centre Hospitalier Universitaire Montpellier, Montpellier, France
| | - Christophe Piot
- Département de Cardiologie Interventionnelle, Clinique du Millénaire, Montpellier, France
| | - Joël Nargeot
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Farida Djouad
- Institute of Regenerative Medicine and Biotherapies, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Centre Hospitalier Universitaire Montpellier, Montpellier, France
| |
Collapse
|
17
|
Liu X, Zhang G, Li D, Ruan Z, Wu B. Effect of 24 h glucose fluctuations on 30-day and 1-year mortality in patients with acute myocardial infarction: an analysis from the MIMIC-III database. Front Cardiovasc Med 2024; 11:1371606. [PMID: 38572310 PMCID: PMC10987860 DOI: 10.3389/fcvm.2024.1371606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/11/2024] [Indexed: 04/05/2024] Open
Abstract
Background It is recognized that patients' blood glucose fluctuates over time during acute disease episodes, especially during the outbreak of cardiovascular events, regardless of the presence of an abnormal blood glucose profile prior to admission to the hospital. Glucose fluctuations in patients with acute myocardial infarction (AMI) in the intensive care unit (ICU) are currently not adequately monitored and studied. We focused on blood glucose fluctuation values within 24 h of admission to assess their association with 30-day and 1-year mortality. Methods Data of patients with AMI aged 18 years or older from the Critical Care Medical Information Marketplace database III V1.4 were available for analysis in this research. Glucose data were obtained by measurement. A total of 390 of them were treated with PCI. The principal consequence was 30-day and 1-year mortality in patients with AMI. The effect of different glucose fluctuations within 24 h of admission on mortality was predicted by constructing a multivariate Cox regression model with four model adjustments and Kaplan-Meier survival curves. Additionally, we performed curve-fitting analyses to show the correlation between blood glucose fluctuations and risk of death. Results We selected 1,699 AMI patients into our study through screening. The included population was categorized into three groups based on the tertiles of blood glucose fluctuation values within 24 h of admission to the ICU. The three groups were <25 mg/dl, 25-88 mg/dl and >88 mg/dl. By cox regression analysis, the group with the highest blood glucose fluctuation values (>88 mg/dl) had the most significant increase in 30-day and 1-year mortality after excluding confounding factors (30-day mortality adjusted HR = 2.11; 95% CI = 1.49-2.98 p < 0.001; 1-year mortality adjusted HR = 1.83; 95% CI = 1.40-2.39 p < 0.001). As demonstrated by the Kaplan-Meier survival curves, the group with the greatest fluctuations in blood glucose has the worst 30-day and 1-year prognosis. Conclusions The extent of glucose fluctuations in patients with AMI in the first 24 h after ICU admission is an essential predictor as to 30-day as well as 1-year mortality. When blood glucose fluctuates more than 88 mg/dl within 24 h, mortality increases significantly with the range of blood glucose fluctuations.
Collapse
Affiliation(s)
- Xiaohe Liu
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Guihong Zhang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Dan Li
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zhishen Ruan
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Bo Wu
- Department of Cardiovascular Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
18
|
Li S, Zheng Y, Kang Y, He X, Zheng Y, Jiang M, Xu X, Ma L, Wang X, Zhang K, Shao X, Fang J, Jiang Y. Electroacupuncture alleviates streptozotocin-induced diabetic neuropathic pain via suppressing phosphorylated CaMKIIα in rats. Neuroreport 2024; 35:258-268. [PMID: 38305135 PMCID: PMC10852042 DOI: 10.1097/wnr.0000000000002000] [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] [Accepted: 01/02/2024] [Indexed: 02/03/2024]
Abstract
Diabetic neuropathic pain (DNP) is a frequent complication of diabetes. Calcium/calmodulin-dependent protein kinase II α (CaMKIIα), a multi-functional serine/threonine kinase subunit, is mainly located in the surface layer of the spinal cord dorsal horn (SCDH) and the primary sensory neurons in dorsal root ganglion (DRG). Numerous studies have indicated electroacupuncture (EA) takes effect in various kinds of pain. In this research, we explored whether CaMKIIα on rats' SCDH and DRG participated in DNP and further explored the mechanisms underlying the analgesic effects of EA. The DNP model in rats was successfully established by intraperitoneal injection of streptozotocin. Certain DNP rats were treated with intrathecal injections of KN93, a CaMKII antagonist, and some of the DNP rats received EA intervention. The general conditions, behaviors, the expressions of CaMKIIα and phosphorylated CaMKIIα (p-CaMKIIα) were evaluated. DNP rats' paw withdrawal threshold was reduced and the expressions of p-CaMKIIα in SCDH and DRG were upregulated compared with the Normal group, while the level of CaMKIIα showed no significance. KN93 attenuated DNP rats' hyperalgesia and reduced the expressions of p-CaMKIIα. We also found EA attenuated the hyperalgesia of DNP rats and reduced the expressions of p-CaMKIIα. The above findings suggest that p-CaMKIIα in SCDH and DRG is involved in DNP. The analgesic effect of EA in DNP might be related to the downregulation of p-CaMKIIα expression level. Our study further supports that EA can be an effective clinical treatment for DNP.
Collapse
Affiliation(s)
- Siyi Li
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Yinmu Zheng
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Yurong Kang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Xiaofen He
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Yu Zheng
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Minjian Jiang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Xinnan Xu
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Liqian Ma
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Xiaoxiang Wang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Kunlong Zhang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Xiaomei Shao
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Jianqiao Fang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Yongliang Jiang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
19
|
Zeng R, Wang Y, Chen J, Liu Q. Furin knockdown inhibited EndMT and abnormal proliferation and migration of endothelial cells. Clin Hemorheol Microcirc 2024; 88:59-70. [PMID: 38820014 DOI: 10.3233/ch-242171] [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] [Indexed: 06/02/2024]
Abstract
BACKGROUND In the pathogenesis of atherosclerotic cardiovascular disorders, vascular endothelium is crucial. A critical step in the development of atherosclerosis is endothelial dysfunction. Furin may play a factor in vascular remodeling, inflammatory cell infiltration, regulation of plaque stability, and atherosclerosis by affecting the adhesion and migration of endothelial cells. It is yet unknown, though, how furin contributes to endothelial dysfunction. METHODS We stimulated endothelial cells with oxidized modified lipoprotein (ox-LDL). Endothelial-to-mesenchymal transition (EndMT) was found using immunofluorescence (IF) and western blot (WB). Furin expression level and Hippo/YAP signal activation were found using reverse transcription-quantitative PCR (RT-qPCR) and WB, respectively. To achieve the goal of furin knockdown, we transfected siRNA using the RNA transmate reagent. Following furin knockdown, cell proliferation, and migration were assessed by the CCK-8, scratch assay, and transwell gold assay, respectively. WB and IF both picked up on EndMT. WB and RT-qPCR, respectively, were used to find furin's expression level. We chose the important micrornas that can regulate furin and we then confirmed them using RT-qPCR. RESULTS EndMT was created by ox-LDL, evidenced by the up-regulation of mesenchymal cell markers and the down-regulation of endothelial cell markers. Furin expression levels in both protein and mRNA were increased, and the Hippo/YAP signaling pathway was turned on. Furin knockdown dramatically reduced the aberrant migration and proliferation of endothelial cells by ox-LDL stimulation. Furin knockdown can also suppress ox-LDL-induced EndMT, up-regulate indicators of endothelial cells, and down-regulate markers of mesenchymal cells. After ox-LDL stimulation and siRNA transfection, furin's expression level was up-regulated and down-regulated. CONCLUSION Our study demonstrated that furin knockdown could affect ox-LDL-induced abnormal endothelial cell proliferation, migration, and EndMT. This implies that furin plays an important role in endothelial dysfunction.
Collapse
Affiliation(s)
- Rui Zeng
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yimin Wang
- Rehabilitation Area of the Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jun Chen
- Rehabilitation Area of the Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Qiang Liu
- Rehabilitation Area of the Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
20
|
Aklilu AM, Kumar S, Yamamoto Y, Moledina DG, Sinha F, Testani JM, Wilson FP. Outcomes Associated with Sodium-Glucose Cotransporter-2 Inhibitor Use in Acute Heart Failure Hospitalizations Complicated by AKI. KIDNEY360 2023; 4:1371-1381. [PMID: 37644648 PMCID: PMC10615381 DOI: 10.34067/kid.0000000000000250] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 08/19/2023] [Indexed: 08/31/2023]
Abstract
Key Points In a multicenter retrospective cohort study of adults hospitalized with acute heart failure, exposure to sodium-glucose cotransporter-2 inhibitor during AKI was associated with lower risk of 30-day mortality. Exposure to sodium-glucose cotransporter-2 inhibitor during acute heart failure–associated AKI was associated with no difference in time to renal recovery. The findings were reproducible in inverse probability-weighted analysis. Background Although sodium-glucose cotransporter-2 inhibitor (SGLT2i) use during acute heart failure (AHF) hospitalizations is associated with symptomatic improvement, reduction in rehospitalizations, and mortality, these medications are often withheld during AKI because of concerns about worsening GFR. We aimed to investigate the safety of SGLT2i exposure during AKI among patients hospitalized with AHF. We hypothesized that SGLT2i exposure would not worsen mortality but may prolong return of creatinine to baseline. Methods This was a retrospective study of adults hospitalized across five Yale New Haven Health System hospitals between January 2020 and May 2022 with AHF complicated by Kidney Disease Improving Global Outcomes–defined AKI. Patients with stage 5 CKD and those with potential contraindications to SGLT2i were excluded. We tested the association of SGLT2i use with kidney function recovery at 14 days and death at 30 days using time-varying, multivariable Cox-regression analyses. Results Of 3305 individuals hospitalized with AHF and AKI, 356 received SGLT2i after AKI diagnosis either as initiation or continuation. The rate of renal recovery was not significantly different among those exposed and unexposed to SGLT2i after AKI (adjusted hazard ratio, 0.94; 95% confidence interval, 0.79 to 1.11; P = 0.46). SGLT2i exposure was associated with lower risk of 30-day mortality (adjusted hazard ratio, 0.45; 95% confidence interval, 0.23 to 0.87; P = 0.02). Sensitivity analyses using an inverse probability-weighted time-varying Cox regression analysis and using alternate definitions of AHF with different NT-proBNP cutoffs yielded similar results. Rates of renal recovery were similar between the exposed and unexposed cohorts regardless of the proximity of SGLT2i exposure to AKI diagnosis. Conclusion In adults experiencing AHF-associated AKI, exposure to SGLT2i was associated with decreased mortality and no delay in renal recovery. Prospective studies are needed to elucidate the effect of SGLT2i exposure during AKI, particularly during heart failure hospitalizations.
Collapse
Affiliation(s)
- Abinet M. Aklilu
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Clinical and Translational Research Accelerator, Yale School of Medicine, New Haven, Connecticut
| | - Sanchit Kumar
- Clinical and Translational Research Accelerator, Yale School of Medicine, New Haven, Connecticut
| | - Yu Yamamoto
- Clinical and Translational Research Accelerator, Yale School of Medicine, New Haven, Connecticut
| | - Dennis G. Moledina
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Clinical and Translational Research Accelerator, Yale School of Medicine, New Haven, Connecticut
| | - Frederick Sinha
- Department of Internal Medicine II, University Medical Center Regensburg, Germany
| | - Jeffrey M. Testani
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - F. Perry Wilson
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Clinical and Translational Research Accelerator, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
21
|
Oriot P, Hermans MP, Beauloye C, Rogghe PA, Noel S, Paternotte E. Unsuspected diabetic ketoacidosis after myocardial infarction in a patient treated with SGLT2 inhibitor increased length of stay in the hospital: how can it be prevented? A case report. Eur Heart J Case Rep 2023; 7:ytad336. [PMID: 37681058 PMCID: PMC10481769 DOI: 10.1093/ehjcr/ytad336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 07/05/2023] [Accepted: 07/19/2023] [Indexed: 09/09/2023]
Abstract
Background As the use of sodium-glucose co-transporter 2 inhibitors (SGLT2is) has expanded beyond glucose-lowering therapy in type 2 diabetes mellitus (T2DM), including chronic kidney disease and heart failure, there has also been an increase in reported cases of diabetic ketoacidosis (DKA) associated with SGLT2i. Case summary A 77-year-old woman with T2DM presented to the emergency department with ST-segment elevation myocardial infarction (MI) complicated by atrial fibrillation. Her medications included empagliflozin, an SGLT2i, initiated for T2DM. Diabetic ketoacidosis was suspected on the basis of a large anion gap, despite a plasma glucose level below 200 mg/dL (11.1 mmol/L) and the absence of symptoms, including nausea and vomiting. Laboratory tests confirmed metabolic acidosis and high ketones. However, the diagnosis of euglycaemic DKA (eu-DKA) was delayed due to lack of symptoms and moderate hyperglycaemia. The patient was successfully treated according to DKA management guidelines. She was discharged on insulin, and SGLT2i was discontinued. Discussion This is a case of asymptomatic eu-DKA after acute MI (AMI). We discuss the use of SGLT2is in AMI and arrhythmias from a review of the literature and the prophylaxis of eu-DKA. Regular monitoring of blood glucose and ketones should be performed in hospitalized T2DM patients treated with SGLT2i. The SGLT2i should be stopped as soon as possible in the event of critical illness or suspected DKA in the setting of an acute illness such as AMI. To help clinicians prevent this potentially fatal disease, we propose a flowchart for the prophylactic management of eu-DKA among inpatients.
Collapse
Affiliation(s)
- Philippe Oriot
- Department of Diabetology, Mouscron Hospital Centre, Avenue de Fécamp 49, 7700 Mouscron, Belgium
| | - Michel P Hermans
- Division of Endocrinology & Nutrition, Université catholique de louvain, Avenue Hippocrate 10, 1200 Woluwe-Saint-Lambert, Brussels, Belgium
- IREC – Institut de Recherche Expérimentale et Clinique Avenue Hippocrate, 55 bte B1.55.02 - 1200 Woluwé-Saint-Lambert – Belgium
| | - Christophe Beauloye
- Division of Cardiology, Université catholique de louvain, Avenue Hippocrate 10, 1200 Woluwe-Saint-Lambert, Brussels, Belgium
- IREC – Institut de Recherche Expérimentale et Clinique Avenue Hippocrate, 55 bte B1.55.02 - 1200 Woluwé-Saint-Lambert – Belgium
| | - Pierre-Arnaud Rogghe
- Department of Critical Care Unit, Mouscron Hospital Centre, Avenue de Fécamp 49, 7700 Mouscron, Belgium
| | - Sophie Noel
- Université Catholique de Louvain (UCLouvain), Av Hippocrate 10/2806, B-1200 Brussels, Belgium
| | - Emmanuelle Paternotte
- Department of Cardiology, Mouscron Hospital Centre, Avenue de Fécamp 49, 7700 Mouscron, Belgium
| |
Collapse
|
22
|
Wang Y, Guo L, Zhang Z, Fu S, Huang P, Wang A, Liu M, Ma X. A bibliometric analysis of myocardial ischemia/reperfusion injury from 2000 to 2023. Front Cardiovasc Med 2023; 10:1180792. [PMID: 37383699 PMCID: PMC10293770 DOI: 10.3389/fcvm.2023.1180792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/22/2023] [Indexed: 06/30/2023] Open
Abstract
Background Myocardial ischemia/reperfusion injury (MIRI) refers to the more severe damage that occurs in the previously ischemic myocardium after a short-term interruption of myocardial blood supply followed by restoration of blood flow within a certain period of time. MIRI has become a major challenge affecting the therapeutic efficacy of cardiovascular surgery. Methods A scientific literature search on MIRI-related papers published from 2000 to 2023 in the Web of Science Core Collection database was conducted. VOSviewer was used for bibliometric analysis to understand the scientific development and research hotspots in this field. Results A total of 5,595 papers from 81 countries/regions, 3,840 research institutions, and 26,202 authors were included. China published the most papers, but the United States had the most significant influence. Harvard University was the leading research institution, and influential authors included Lefer David J., Hausenloy Derek J., Yellon Derek M., and others. All keywords can be divided into four different directions: risk factors, poor prognosis, mechanisms and cardioprotection. Conclusion Research on MIRI is flourishing. It is necessary to conduct an in-depth investigation of the interaction between different mechanisms and multi-target therapy will be the focus and hotspot of MIRI research in the future.
Collapse
Affiliation(s)
- Yifei Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Lijun Guo
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Zhibo Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Shuangqing Fu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Pingping Huang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Anzhu Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mi Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Xiaochang Ma
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| |
Collapse
|
23
|
Wang Q, Zuurbier CJ, Huhn R, Torregroza C, Hollmann MW, Preckel B, van den Brom CE, Weber NC. Pharmacological Cardioprotection against Ischemia Reperfusion Injury-The Search for a Clinical Effective Therapy. Cells 2023; 12:1432. [PMID: 37408266 DOI: 10.3390/cells12101432] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/10/2023] [Accepted: 05/17/2023] [Indexed: 07/07/2023] Open
Abstract
Pharmacological conditioning aims to protect the heart from myocardial ischemia-reperfusion injury (IRI). Despite extensive research in this area, today, a significant gap remains between experimental findings and clinical practice. This review provides an update on recent developments in pharmacological conditioning in the experimental setting and summarizes the clinical evidence of these cardioprotective strategies in the perioperative setting. We start describing the crucial cellular processes during ischemia and reperfusion that drive acute IRI through changes in critical compounds (∆GATP, Na+, Ca2+, pH, glycogen, succinate, glucose-6-phosphate, mitoHKII, acylcarnitines, BH4, and NAD+). These compounds all precipitate common end-effector mechanisms of IRI, such as reactive oxygen species (ROS) generation, Ca2+ overload, and mitochondrial permeability transition pore opening (mPTP). We further discuss novel promising interventions targeting these processes, with emphasis on cardiomyocytes and the endothelium. The limited translatability from basic research to clinical practice is likely due to the lack of comorbidities, comedications, and peri-operative treatments in preclinical animal models, employing only monotherapy/monointervention, and the use of no-flow (always in preclinical models) versus low-flow ischemia (often in humans). Future research should focus on improved matching between preclinical models and clinical reality, and on aligning multitarget therapy with optimized dosing and timing towards the human condition.
Collapse
Affiliation(s)
- Qian Wang
- Department of Anesthesiology-L.E.I.C.A., Amsterdam University Medical Centers, Location AMC, Cardiovascular Science, Meibergdreef 11, 1105 AZ Amsterdam, The Netherlands
| | - Coert J Zuurbier
- Department of Anesthesiology-L.E.I.C.A., Amsterdam University Medical Centers, Location AMC, Cardiovascular Science, Meibergdreef 11, 1105 AZ Amsterdam, The Netherlands
| | - Ragnar Huhn
- Department of Anesthesiology, Kerckhoff-Clinic-Center for Heart, Lung, Vascular and Rheumatic Disease, Justus-Liebig-University Giessen, Benekestr. 2-8, 61231 Bad Nauheim, Germany
| | - Carolin Torregroza
- Department of Anesthesiology, Kerckhoff-Clinic-Center for Heart, Lung, Vascular and Rheumatic Disease, Justus-Liebig-University Giessen, Benekestr. 2-8, 61231 Bad Nauheim, Germany
| | - Markus W Hollmann
- Department of Anesthesiology-L.E.I.C.A., Amsterdam University Medical Centers, Location AMC, Cardiovascular Science, Meibergdreef 11, 1105 AZ Amsterdam, The Netherlands
| | - Benedikt Preckel
- Department of Anesthesiology-L.E.I.C.A., Amsterdam University Medical Centers, Location AMC, Cardiovascular Science, Meibergdreef 11, 1105 AZ Amsterdam, The Netherlands
| | - Charissa E van den Brom
- Department of Anesthesiology-L.E.I.C.A., Amsterdam University Medical Centers, Location AMC, Cardiovascular Science, Meibergdreef 11, 1105 AZ Amsterdam, The Netherlands
| | - Nina C Weber
- Department of Anesthesiology-L.E.I.C.A., Amsterdam University Medical Centers, Location AMC, Cardiovascular Science, Meibergdreef 11, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
24
|
Chen R, Zhang Y, Zhang H, Zhou H, Tong W, Wu Y, Ma M, Chen Y. SGLT2 inhibitor dapagliflozin alleviates intramyocardial hemorrhage and adverse ventricular remodeling via suppressing hepcidin in myocardial ischemia-reperfusion injury. Eur J Pharmacol 2023; 950:175729. [PMID: 37100110 DOI: 10.1016/j.ejphar.2023.175729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/01/2023] [Accepted: 04/14/2023] [Indexed: 04/28/2023]
Abstract
Intramyocardial hemorrhage (IMH), a reperfusion therapy-associated complication, is the extravasation of red blood cells caused by severe microvascular injury. IMH is an independent predictor of adverse ventricular remodeling (AVR) after acute myocardial infarction (AMI). Hepcidin, a major regulator of iron uptake and systemic distribution, is a key factor affecting AVR. However, the role of cardiac hepcidin in the development of IMH has not been completely elucidated. This study aimed to explore if sodium-dependent glucose co-transporter 2 inhibitor (SGLT2i) exerts therapeutic effects on IMH and AVR by suppressing hepcidin and to elucidate the underlying mechanisms. SGLT2i alleviated IMH and AVR in the ischemia-reperfusion injury (IRI) mouse model. Additionally, SGLT2i downregulated the cardiac levels of hepcidin in IRI mice, suppressed M1-type macrophage polarization, and promoted M2-type macrophage polarization. The effects of hepcidin knockdown on macrophage polarization were similar to those of SGLT2i in RAW264.7 cells. SGLT2i treatment or hepcidin knockdown inhibited the expression of MMP9, an inducer of IMH and AVR, in RAW264.7 cells. Regulation of macrophage polarization and reduction of MMP9 expression by SGLT2i and hepcidin knockdown is achieved through activation of pSTAT3. In conclusion, this study demonstrated that SGLT2i alleviated IMH and AVR by regulating macrophage polarization. The potential mechanism through which SGLT2i exerted its therapeutic effect seems to involve the downregulation of MMP9 via the hepcidin-STAT3 pathway.
Collapse
Affiliation(s)
- Rundu Chen
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, China; Senior Department of Cardiology, the Sixth Medical Center of PLA General Hospital, Beijing, 100853, China.
| | - Yingqian Zhang
- Senior Department of Cardiology, the Sixth Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Haoran Zhang
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100853, China
| | - Hao Zhou
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, China; Senior Department of Cardiology, the Sixth Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Wei Tong
- Senior Department of Cardiology, the Sixth Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Yuanbin Wu
- Department of Emergency, the Seventh Medical Center, Chinese PLA General Hospital, Beijing, 100700, China
| | - Mingrui Ma
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, China; Senior Department of Cardiology, the Sixth Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Yundai Chen
- Senior Department of Cardiology, the Sixth Medical Center of PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
25
|
Sabe SA, Xu CM, Sabra M, Harris DD, Malhotra A, Aboulgheit A, Stanley M, Abid MR, Sellke FW. Canagliflozin Improves Myocardial Perfusion, Fibrosis, and Function in a Swine Model of Chronic Myocardial Ischemia. J Am Heart Assoc 2023; 12:e028623. [PMID: 36583437 PMCID: PMC9973570 DOI: 10.1161/jaha.122.028623] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/28/2022] [Indexed: 12/31/2022]
Abstract
Background Sodium-glucose cotransporter-2 inhibitors are cardioprotective independent of glucose control, as demonstrated in animal models of acute myocardial ischemia and clinical trials. The functional and molecular mechanisms of these benefits in the setting of chronic myocardial ischemia are poorly defined. The purpose of this study is to determine the effects of canagliflozin therapy on myocardial perfusion, fibrosis, and function in a large animal model of chronic myocardial ischemia. Methods and Results Yorkshire swine underwent placement of an ameroid constrictor to the left circumflex artery to induce chronic myocardial ischemia. Two weeks later, pigs received either no drug (n=8) or 300 mg sodium-glucose cotransporter-2 inhibitor canagliflozin orally, daily (n=8). Treatment continued for 5 weeks, followed by hemodynamic measurements, harvest, and tissue analysis. Canagliflozin therapy was associated with increased stroke volume and stroke work and decreased left ventricular stiffness compared with controls. The canagliflozin group had improved perfusion to ischemic myocardium compared with controls, without differences in arteriolar or capillary density. Canagliflozin was associated with decreased interstitial and perivascular fibrosis in chronically ischemic tissue, with reduced Jak/STAT (Janus kinase/signal transducer and activator of transcription) signaling compared with controls. In ischemic myocardium of the canagliflozin group, there was increased expression and activation of adenosine monophosphate-activated protein kinase, decreased activation of endothelial nitric oxide synthase, and unchanged total endothelial nitric oxide synthase. Canagliflozin therapy reduced total protein oxidation and increased expression of mitochondrial antioxidant superoxide dismutase 2 compared with controls. Conclusions In the setting of chronic myocardial ischemia, canagliflozin therapy improves myocardial function and perfusion to ischemic territory, without changes in collateralization. Attenuation of fibrosis via reduced Jak/STAT signaling, activation of adenosine monophosphate-activated protein kinase, and antioxidant signaling may contribute to these effects.
Collapse
Affiliation(s)
- Sharif A. Sabe
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island HospitalAlpert Medical School of Brown University, Rhode Island HospitalProvidenceRI
| | - Cynthia M. Xu
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island HospitalAlpert Medical School of Brown University, Rhode Island HospitalProvidenceRI
| | - Mohamed Sabra
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island HospitalAlpert Medical School of Brown University, Rhode Island HospitalProvidenceRI
| | - Dwight Douglas Harris
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island HospitalAlpert Medical School of Brown University, Rhode Island HospitalProvidenceRI
| | - Akshay Malhotra
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island HospitalAlpert Medical School of Brown University, Rhode Island HospitalProvidenceRI
| | - Ahmed Aboulgheit
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island HospitalAlpert Medical School of Brown University, Rhode Island HospitalProvidenceRI
| | - Madigan Stanley
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island HospitalAlpert Medical School of Brown University, Rhode Island HospitalProvidenceRI
| | - M. Ruhul Abid
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island HospitalAlpert Medical School of Brown University, Rhode Island HospitalProvidenceRI
| | - Frank W. Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island HospitalAlpert Medical School of Brown University, Rhode Island HospitalProvidenceRI
| |
Collapse
|
26
|
Paolisso P, Bergamaschi L, Gragnano F, Gallinoro E, Cesaro A, Sardu C, Mileva N, Foà A, Armillotta M, Sansonetti A, Amicone S, Impellizzeri A, Esposito G, Morici N, Andrea OJ, Casella G, Mauro C, Vassilev D, Galie N, Santulli G, Marfella R, Calabrò P, Pizzi C, Barbato E. Outcomes in diabetic patients treated with SGLT2-Inhibitors with acute myocardial infarction undergoing PCI: The SGLT2-I AMI PROTECT Registry. Pharmacol Res 2023; 187:106597. [PMID: 36470546 PMCID: PMC9946774 DOI: 10.1016/j.phrs.2022.106597] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/27/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
AIMS To investigate in-hospital and long-term prognosis in T2DM patients presenting with acute myocardial infarction (AMI) treated with SGLT2-I versus other oral anti-diabetic agents (non-SGLT2-I users). METHODS In this multicenter international registry all consecutive diabetic AMI patients undergoing percutaneous coronary intervention between 2018 and 2021 were enrolled and, based on the admission anti-diabetic therapy, divided into SGLT-I users versus non-SGLT2-I users. The primary endpoint was defined as a composite of cardiovascular death, recurrent AMI, and hospitalization for HF (MACE). Secondary outcomes included i) in-hospital cardiovascular death, recurrent AMI, occurrence of arrhythmias, and contrast-induced acute kidney injury (CI-AKI); ii) long-term cardiovascular mortality, recurrent AMI, heart failure (HF) hospitalization. RESULTS The study population consisted of 646 AMI patients (with or without ST-segment elevation): 111 SGLT2-I users and 535 non-SGLT-I users. The use of SGLT2-I was associated with a significantly lower in-hospital cardiovascular death, arrhythmic burden, and occurrence of CI-AKI (all p < 0.05). During a median follow-up of 24 ± 13 months, the primary composite endpoint, as well as cardiovascular mortality and HF hospitalization were lower for SGLT2-I users compared to non-SGLT2-I patients (p < 0.04 for all). After adjusting for confounding factors, the use of SGLT2-I was identified as independent predictor of reduced MACE occurrence (HR=0.57; 95%CI:0.33-0.99; p = 0.039) and HF hospitalization (HR=0.46; 95%CI:0.21-0.98; p = 0.041). CONCLUSIONS In T2DM AMI patients, the use of SGLT2-I was associated with a lower risk of adverse cardiovascular outcomes during index hospitalization and long-term follow-up. Our findings provide new insights into the cardioprotective effects of SGLT2-I in the setting of AMI. REGISTRATION Data are part of the observational international registry: SGLT2-I AMI PROTECT. CLINICALTRIALS gov Identifier: NCT05261867.
Collapse
Affiliation(s)
- Pasquale Paolisso
- Cardiovascular Center Aalst, OLV-Clinic, Aalst, Belgium; Dept. of Advanced Biomedical Sciences, University Federico II, Naples, Italy
| | - Luca Bergamaschi
- Unit of Cardiology, Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, Sant'Orsola-Malpighi Hospital, IRCCS, Bologna, Italy
| | - Felice Gragnano
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Naples, Italy; Division of Cardiology, A.O.R.N. "Sant'Anna e San Sebastiano", Caserta, Italy
| | - Emanuele Gallinoro
- Cardiovascular Center Aalst, OLV-Clinic, Aalst, Belgium; Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Arturo Cesaro
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Naples, Italy; Division of Cardiology, A.O.R.N. "Sant'Anna e San Sebastiano", Caserta, Italy
| | - Celestino Sardu
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Niya Mileva
- Cardiology Clinic, "Alexandrovska" University Hospital, Medical University of Sofia, Sofia, Bulgaria
| | - Alberto Foà
- Unit of Cardiology, Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, Sant'Orsola-Malpighi Hospital, IRCCS, Bologna, Italy
| | - Matteo Armillotta
- Unit of Cardiology, Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, Sant'Orsola-Malpighi Hospital, IRCCS, Bologna, Italy
| | - Angelo Sansonetti
- Unit of Cardiology, Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, Sant'Orsola-Malpighi Hospital, IRCCS, Bologna, Italy
| | - Sara Amicone
- Unit of Cardiology, Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, Sant'Orsola-Malpighi Hospital, IRCCS, Bologna, Italy
| | - Andrea Impellizzeri
- Unit of Cardiology, Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, Sant'Orsola-Malpighi Hospital, IRCCS, Bologna, Italy
| | - Giuseppe Esposito
- Dept. of Advanced Biomedical Sciences, University Federico II, Naples, Italy; Interventional Cardiology Unit, De Gasperis Cardio Center, Niguarda Hospital, Milan, Italy
| | - Nuccia Morici
- IRCCS S. Maria Nascente - Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| | - Oreglia Jacopo Andrea
- Interventional Cardiology Unit, De Gasperis Cardio Center, Niguarda Hospital, Milan, Italy
| | | | - Ciro Mauro
- Department of Cardiology, Hospital Cardarelli, Naples, Italy
| | | | - Nazzareno Galie
- Unit of Cardiology, Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, Sant'Orsola-Malpighi Hospital, IRCCS, Bologna, Italy
| | - Gaetano Santulli
- Dept. of Advanced Biomedical Sciences, University Federico II, Naples, Italy; International Translational Research and Medical Education (ITME) Consortium, Naples, Italy; Department of Medicine (Division of Cardiology) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein-Sinai Diabetes Research Center, The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, New York, USA
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy; Mediterranea Cardiocentro, Naples, Italy
| | - Paolo Calabrò
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Naples, Italy; Division of Cardiology, A.O.R.N. "Sant'Anna e San Sebastiano", Caserta, Italy
| | - Carmine Pizzi
- Unit of Cardiology, Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, Sant'Orsola-Malpighi Hospital, IRCCS, Bologna, Italy.
| | | |
Collapse
|
27
|
Ferdinandy P, Andreadou I, Baxter GF, Bøtker HE, Davidson SM, Dobrev D, Gersh BJ, Heusch G, Lecour S, Ruiz-Meana M, Zuurbier CJ, Hausenloy DJ, Schulz R. Interaction of Cardiovascular Nonmodifiable Risk Factors, Comorbidities and Comedications With Ischemia/Reperfusion Injury and Cardioprotection by Pharmacological Treatments and Ischemic Conditioning. Pharmacol Rev 2023; 75:159-216. [PMID: 36753049 PMCID: PMC9832381 DOI: 10.1124/pharmrev.121.000348] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 08/07/2022] [Accepted: 09/12/2022] [Indexed: 12/13/2022] Open
Abstract
Preconditioning, postconditioning, and remote conditioning of the myocardium enhance the ability of the heart to withstand a prolonged ischemia/reperfusion insult and the potential to provide novel therapeutic paradigms for cardioprotection. While many signaling pathways leading to endogenous cardioprotection have been elucidated in experimental studies over the past 30 years, no cardioprotective drug is on the market yet for that indication. One likely major reason for this failure to translate cardioprotection into patient benefit is the lack of rigorous and systematic preclinical evaluation of promising cardioprotective therapies prior to their clinical evaluation, since ischemic heart disease in humans is a complex disorder caused by or associated with cardiovascular risk factors and comorbidities. These risk factors and comorbidities induce fundamental alterations in cellular signaling cascades that affect the development of ischemia/reperfusion injury and responses to cardioprotective interventions. Moreover, some of the medications used to treat these comorbidities may impact on cardioprotection by again modifying cellular signaling pathways. The aim of this article is to review the recent evidence that cardiovascular risk factors as well as comorbidities and their medications may modify the response to cardioprotective interventions. We emphasize the critical need for taking into account the presence of cardiovascular risk factors as well as comorbidities and their concomitant medications when designing preclinical studies for the identification and validation of cardioprotective drug targets and clinical studies. This will hopefully maximize the success rate of developing rational approaches to effective cardioprotective therapies for the majority of patients with multiple comorbidities. SIGNIFICANCE STATEMENT: Ischemic heart disease is a major cause of mortality; however, there are still no cardioprotective drugs on the market. Most studies on cardioprotection have been undertaken in animal models of ischemia/reperfusion in the absence of comorbidities; however, ischemic heart disease develops with other systemic disorders (e.g., hypertension, hyperlipidemia, diabetes, atherosclerosis). Here we focus on the preclinical and clinical evidence showing how these comorbidities and their routine medications affect ischemia/reperfusion injury and interfere with cardioprotective strategies.
Collapse
Affiliation(s)
- Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Ioanna Andreadou
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Gary F Baxter
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Hans Erik Bøtker
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Sean M Davidson
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Dobromir Dobrev
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Bernard J Gersh
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Gerd Heusch
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Sandrine Lecour
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Marisol Ruiz-Meana
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Coert J Zuurbier
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Derek J Hausenloy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Rainer Schulz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| |
Collapse
|
28
|
Chen W, Zhang Y, Wang Z, Tan M, Lin J, Qian X, Li H, Jiang T. Dapagliflozin alleviates myocardial ischemia/reperfusion injury by reducing ferroptosis via MAPK signaling inhibition. Front Pharmacol 2023; 14:1078205. [PMID: 36891270 PMCID: PMC9986553 DOI: 10.3389/fphar.2023.1078205] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 02/08/2023] [Indexed: 02/22/2023] Open
Abstract
Reperfusion is essential for ischemic myocardium but paradoxically leads to myocardial damage that worsens cardiac functions. Ferroptosis often occurs in cardiomyocytes during ischemia/reperfusion (I/R). The SGLT2 inhibitor dapagliflozin (DAPA) exerts cardioprotective effects independent of hypoglycemia. Here, we investigated the effect and potential mechanism of DAPA against myocardial ischemia/reperfusion injury (MIRI)-related ferroptosis using the MIRI rat model and hypoxia/reoxygenation (H/R)-induced H9C2 cardiomyocytes. Our results show that DAPA significantly ameliorated myocardial injury, reperfusion arrhythmia, and cardiac function, as evidenced by alleviated ST-segment elevation, ameliorated cardiac injury biomarkers including cTnT and BNP and pathological features, prevented H/R-triggered cell viability loss in vitro. In vitro and in vivo experiments showed that DAPA inhibited ferroptosis by upregulating the SLC7A11/GPX4 axis and FTH and inhibiting ACSL4. DAPA notably mitigated oxidative stress, lipid peroxidation, ferrous iron overload, and reduced ferroptosis. Subsequently, network pharmacology and bioinformatics analysis suggested that the MAPK signaling pathway was a potential target of DAPA and a common mechanism of MIRI and ferroptosis. DAPA treatment significantly reduced MAPK phosphorylation in vitro and in vivo, suggesting that DAPA might protect against MIRI by reducing ferroptosis through the MAPK signaling pathway.
Collapse
Affiliation(s)
- Weixiang Chen
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yue Zhang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zuoxiang Wang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Mingyue Tan
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jia Lin
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaodong Qian
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hongxia Li
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Tingbo Jiang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
29
|
Sang H, Wan Y, Ma Z, Zhang S, Zhao Q. Cost-effectiveness of empagliflozin for the treatment of heart failure with reduced ejection fraction in China. Front Cardiovasc Med 2022; 9:1022020. [PMID: 36465457 PMCID: PMC9708714 DOI: 10.3389/fcvm.2022.1022020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 11/01/2022] [Indexed: 09/01/2023] Open
Abstract
AIM To determine the pharmacoeconomics of empagliflozin for the treatment of heart failure (HF) with reduced ejection fraction in China and to provide evidence-based reference for clinical rational drug selection and medical decision-making. RESEARCH DESIGN AND METHODS We used the Markov model to evaluate the cost-effectiveness of empagliflozin for the treatment of HF with reduced ejection fraction (HFrEF). We evaluated the cost-effectiveness of the standard treatment in addition to empagliflozin (empagliflozin group) vs. the cost-effectiveness of the standard treatment alone (standard treatment group). RESULTS We found that each additional quality-adjusted life year (QALY) in the empagliflozin group costed $3,842.20 more, which was less than China's gross domestic product (GDP) per capita in 2021 ($11,981). The steady-state mortality in the two groups was the key factor affecting the incremental cost-effectiveness ratio (ICER). Probabilistic sensitivity analysis revealed that when the willingness-to-pay (WTP) threshold was one time the GDP per capita in 2021 ($11,981) and three times the GDP per capita in 2021 ($35,943), the probability of the empagliflozin group being cost-effective was 85.8 and 91.6%, respectively. CONCLUSION Compared with the standard treatment alone, the addition of empagliflozin to the standard treatment was more cost-effective for the treatment of HFrEF in China.
Collapse
Affiliation(s)
- Haiqiang Sang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiming Wan
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenzhou Ma
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shengye Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qiuping Zhao
- Department of Cardiology, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| |
Collapse
|
30
|
Wu YJ, Wang SB, Wang LS. SGLT2 Inhibitors: New Hope for the Treatment of Acute Myocardial Infarction? Am J Cardiovasc Drugs 2022; 22:601-613. [PMID: 35947249 DOI: 10.1007/s40256-022-00545-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/19/2022] [Indexed: 11/01/2022]
Abstract
Among all of the new antidiabetic drugs, an increasing number of studies have evaluated the relationship between the sodium-glucose cotransporter 2 inhibitors (SGLT2i) and acute myocardial infarction (AMI). Since SGLT2i like empagliflozin, canagliflozin, and recently, dapagliflozin have shown impressive positive effects in patients with chronic heart failure with reduced ejection fraction (HFrEF), it has increased research interest to explore the cardiac molecular mechanisms underlying the clinical benefits and attracted more attention to the effects of SGLT2i on a series of cardiovascular events. Experimental and clinical data on SGLT2i treatment after AMI is limited. This is a review of the clinical and preclinical effects of SGLT2i, focusing on available data on the effects of SGLT2i in AMI patients with a brief overview of ongoing trials.
Collapse
Affiliation(s)
- Yu-Jie Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Si-Bo Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Lian-Sheng Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
31
|
Scisciola L, Cataldo V, Taktaz F, Fontanella RA, Pesapane A, Ghosh P, Franzese M, Puocci A, De Angelis A, Sportiello L, Marfella R, Barbieri M. Anti-inflammatory role of SGLT2 inhibitors as part of their anti-atherosclerotic activity: Data from basic science and clinical trials. Front Cardiovasc Med 2022; 9:1008922. [PMID: 36148061 PMCID: PMC9485634 DOI: 10.3389/fcvm.2022.1008922] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 08/16/2022] [Indexed: 01/10/2023] Open
Abstract
Atherosclerosis is a progressive inflammatory disease leading to mortality and morbidity in the civilized world. Atherosclerosis manifests as an accumulation of plaques in the intimal layer of the arterial wall that, by its subsequent erosion or rupture, triggers cardiovascular diseases. Diabetes mellitus is a well-known risk factor for atherosclerosis. Indeed, Type 2 diabetes mellitus patients have an increased risk of atherosclerosis and its associated-cardiovascular complications than non-diabetic patients. Sodium-glucose co-transport 2 inhibitors (SGLT2i), a novel anti-diabetic drugs, have a surprising advantage in cardiovascular effects, such as reducing cardiovascular death in a patient with or without diabetes. Numerous studies have shown that atherosclerosis is due to a significant inflammatory burden and that SGLT2i may play a role in inflammation. In fact, several experiment results have demonstrated that SGLT2i, with suppression of inflammatory mechanism, slows the progression of atherosclerosis. Therefore, SGLT2i may have a double benefit in terms of glycemic control and control of the atherosclerotic process at a myocardial and vascular level. This review elaborates on the anti-inflammatory effects of sodium-glucose co-transporter 2 inhibitors on atherosclerosis.
Collapse
Affiliation(s)
- Lucia Scisciola
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- *Correspondence: Lucia Scisciola
| | - Vittoria Cataldo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Fatemeh Taktaz
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Rosaria Anna Fontanella
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Ada Pesapane
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Puja Ghosh
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Martina Franzese
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Armando Puocci
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology “L. Donatelli”, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Liberata Sportiello
- Department of Experimental Medicine, Section of Pharmacology “L. Donatelli”, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Mediterranea Cardiocentro, Napoli, Italy
| | - Michelangela Barbieri
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| |
Collapse
|
32
|
Leancă SA, Crișu D, Petriș AO, Afrăsânie I, Genes A, Costache AD, Tesloianu DN, Costache II. Left Ventricular Remodeling after Myocardial Infarction: From Physiopathology to Treatment. Life (Basel) 2022; 12:1111. [PMID: 35892913 PMCID: PMC9332014 DOI: 10.3390/life12081111] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 12/11/2022] Open
Abstract
Myocardial infarction (MI) is the leading cause of death and morbidity worldwide, with an incidence relatively high in developed countries and rapidly growing in developing countries. The most common cause of MI is the rupture of an atherosclerotic plaque with subsequent thrombotic occlusion in the coronary circulation. This causes cardiomyocyte death and myocardial necrosis, with subsequent inflammation and fibrosis. Current therapies aim to restore coronary flow by thrombus dissolution with pharmaceutical treatment and/or intravascular stent implantation and to counteract neurohormonal activation. Despite these therapies, the injury caused by myocardial ischemia leads to left ventricular remodeling; this process involves changes in cardiac geometry, dimension and function and eventually progression to heart failure (HF). This review describes the pathophysiological mechanism that leads to cardiac remodeling and the therapeutic strategies with a role in slowing the progression of remodeling and improving cardiac structure and function.
Collapse
Affiliation(s)
- Sabina Andreea Leancă
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Daniela Crișu
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Antoniu Octavian Petriș
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Str. University nr. 16, 700083 Iasi, Romania;
| | - Irina Afrăsânie
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Antonia Genes
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Alexandru Dan Costache
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Str. University nr. 16, 700083 Iasi, Romania;
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Dan Nicolae Tesloianu
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Irina Iuliana Costache
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Str. University nr. 16, 700083 Iasi, Romania;
| |
Collapse
|
33
|
Diao H, Cheng J, Huang X, Huang B, Shao X, Zhao J, Lan D, Zhu Q, Yan M, Zhang Y, Rong X, Guo J. The Chinese medicine Fufang Zhenzhu Tiaozhi capsule protects against atherosclerosis by suppressing EndMT via modulating Akt1/β-catenin signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2022; 293:115261. [PMID: 35447198 DOI: 10.1016/j.jep.2022.115261] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/17/2022] [Accepted: 03/30/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fufang Zhenzhu Tiaozhi (FTZ) is a traditional Chinese herbal prescription that has been used to treat dyslipidemia, nonalcoholic fatty liver disease, atherosclerosis, diabetes and its complications in the clinic for almost ten years. Endothelial-mesenchymal transition (EndMT) is the key driver of atherosclerosis. However, the effects of FTZ on endothelial dysfunction and EndMT remain unknown. AIM OF THE STUDY To evaluate the therapeutic effects of FTZ against EndMT and the underlying mechanisms. MATERIALS AND METHODS An in vivo model of atherosclerosis was established by feeding ApoE-/- mice with a high-fat diet (HFD). The body weight, lipid levels, plaque area, lipid deposition and EndMT were evaluated using standard assays 12 weeks after intragastric administration of FTZ and simvastatin. Human umbilical vein endothelial cells (HUVECs) were treated with oxidized low-density lipoprotein (ox-LDL) to simulate EndMT in vitro. The degree of EndMT was assessed after treating the cells with FTZ or transfection with si-Akt1. The expression levels of genes involved in EndMT were quantified by real-time PCR or western blotting. RESULTS FTZ ameliorated dyslipidemia and endothelial dysfunction in the atherosclerotic mice. In addition, FTZ reduced body weight and the total cholesterol, triglycerides and low-density lipoprotein levels, and increased that of high-density lipoproteins. FTZ also upregulated the expression of endothelial markers (CD31 and VE-cadherin) and decreased that of mesenchymal markers (ɑ-SMA and FSP1), indicating that it inhibits EndMT. Knocking down Akt1 exacerbated EndMT and reversed the therapeutic effect of FTZ. CONCLUSION FTZ delayed atherosclerosis by inhibiting EndMT via the Akt1/β-catenin pathway.
Collapse
Affiliation(s)
- Hongtao Diao
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Jiawen Cheng
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Xueying Huang
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Bingying Huang
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Xiaoqi Shao
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Jingjing Zhao
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Dingming Lan
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Qing Zhu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, 510006, China.
| | - Meiling Yan
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Yue Zhang
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Xianglu Rong
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, 510006, China.
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, 510006, China.
| |
Collapse
|
34
|
Sodium-Glucose Cotransporter-2 Inhibitors-from the Treatment of Diabetes to Therapy of Chronic Heart Failure. J Cardiovasc Dev Dis 2022; 9:jcdd9070225. [PMID: 35877587 PMCID: PMC9325125 DOI: 10.3390/jcdd9070225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/08/2022] [Accepted: 07/12/2022] [Indexed: 02/01/2023] Open
Abstract
Sodium-glucose cotransporter-2 (SGLT2) inhibitors are currently the second-line pharmacotherapy in type 2 diabetes, particularly through their effectiveness in reducing glycemia, but also due to their cardioprotective and nephroprotective effects. In light of surprisingly satisfactory results from large, randomized trials on gliflozins, SGLT2 received the highest recommendation (Class IA) with the highest level of evidence (A) in the treatment algorithm for HF with reduced LVEF in recent ESC HF guidelines. This great breakthrough in the treatment of HF is due to different mechanisms of action of gliflozins that are reported to be able to change the natural course of HF by reducing the risk of both hospitalization and death. They are recommended regardless of the patient’s diabetes status. This review summarizes the up-to-date literature on their beneficial and pleiotropic impact on the cardiovascular system.
Collapse
|
35
|
Role of Oxidative Stress in Cardiac Dysfunction and Subcellular Defects Due to Ischemia-Reperfusion Injury. Biomedicines 2022; 10:biomedicines10071473. [PMID: 35884777 PMCID: PMC9313001 DOI: 10.3390/biomedicines10071473] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 11/17/2022] Open
Abstract
Ischemia-reperfusion (I/R) injury is well-known to be associated with impaired cardiac function, massive arrhythmias, marked alterations in cardiac metabolism and irreversible ultrastructural changes in the heart. Two major mechanisms namely oxidative stress and intracellular Ca2+-overload are considered to explain I/R-induced injury to the heart. However, it is becoming apparent that oxidative stress is the most critical pathogenic factor because it produces myocardial abnormalities directly or indirectly for the occurrence of cardiac damage. Furthermore, I/R injury has been shown to generate oxidative stress by promoting the formation of different reactive oxygen species due to defects in mitochondrial function and depressions in both endogenous antioxidant levels as well as regulatory antioxidative defense systems. It has also been demonstrated to adversely affect a wide variety of metabolic pathways and targets in cardiomyocytes, various resident structures in myocardial interstitium, as well as circulating neutrophils and leukocytes. These I/R-induced alterations in addition to myocardial inflammation may cause cell death, fibrosis, inflammation, Ca2+-handling abnormalities, activation of proteases and phospholipases, as well as subcellular remodeling and depletion of energy stores in the heart. Analysis of results from isolated hearts perfused with or without some antioxidant treatments before subjecting to I/R injury has indicated that cardiac dysfunction is associated with the development of oxidative stress, intracellular Ca2+-overload and protease activation. In addition, changes in the sarcolemma and sarcoplasmic reticulum Ca2+-handling, mitochondrial oxidative phosphorylation as well as myofibrillar Ca2+-ATPase activities in I/R hearts were attenuated by pretreatment with antioxidants. The I/R-induced alterations in cardiac function were simulated upon perfusing the hearts with oxyradical generating system or oxidant. These observations support the view that oxidative stress may be intimately involved in inducing intracellular Ca2+-overload, protease activation, subcellular remodeling, and cardiac dysfunction as a consequence of I/R injury to the heart.
Collapse
|
36
|
Nikolaou PE, Mylonas N, Makridakis M, Makrecka-Kuka M, Iliou A, Zerikiotis S, Efentakis P, Kampoukos S, Kostomitsopoulos N, Vilskersts R, Ikonomidis I, Lambadiari V, Zuurbier CJ, Latosinska A, Vlahou A, Dimitriadis G, Iliodromitis EK, Andreadou I. Cardioprotection by selective SGLT-2 inhibitors in a non-diabetic mouse model of myocardial ischemia/reperfusion injury: a class or a drug effect? Basic Res Cardiol 2022; 117:27. [PMID: 35581445 DOI: 10.1007/s00395-022-00934-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/04/2022] [Accepted: 05/04/2022] [Indexed: 02/08/2023]
Abstract
Major clinical trials with sodium glucose co-transporter-2 inhibitors (SGLT-2i) exhibit protective effects against heart failure events, whereas inconsistencies regarding the cardiovascular death outcomes are observed. Therefore, we aimed to compare the selective SGLT-2i empagliflozin (EMPA), dapagliflozin (DAPA) and ertugliflozin (ERTU) in terms of infarct size (IS) reduction and to reveal the cardioprotective mechanism in healthy non-diabetic mice. C57BL/6 mice randomly received vehicle, EMPA (10 mg/kg/day) and DAPA or ERTU orally at the stoichiometrically equivalent dose (SED) for 7 days. 24 h-glucose urinary excretion was determined to verify SGLT-2 inhibition. IS of the region at risk was measured after 30 min ischemia (I), and 120 min reperfusion (R). In a second series, the ischemic myocardium was collected (10th min of R) for shotgun proteomics and evaluation of the cardioprotective signaling. In a third series, we evaluated the oxidative phosphorylation capacity (OXPHOS) and the mitochondrial fatty acid oxidation capacity by measuring the respiratory rates. Finally, Stattic, the STAT-3 inhibitor and wortmannin were administered in both EMPA and DAPA groups to establish causal relationships in the mechanism of protection. EMPA, DAPA and ERTU at the SED led to similar SGLT-2 inhibition as inferred by the significant increase in glucose excretion. EMPA and DAPA but not ERTU reduced IS. EMPA preserved mitochondrial functionality in complex I&II linked oxidative phosphorylation. EMPA and DAPA treatment led to NF-kB, RISK, STAT-3 activation and the downstream apoptosis reduction coinciding with IS reduction. Stattic and wortmannin attenuated the cardioprotection afforded by EMPA and DAPA. Among several upstream mediators, fibroblast growth factor-2 (FGF-2) and caveolin-3 were increased by EMPA and DAPA treatment. ERTU reduced IS only when given at the double dose of the SED (20 mg/kg/day). Short-term EMPA and DAPA, but not ERTU administration at the SED reduce IS in healthy non-diabetic mice. Cardioprotection is not correlated to SGLT-2 inhibition, is STAT-3 and PI3K dependent and associated with increased FGF-2 and Cav-3 expression.
Collapse
Affiliation(s)
- Panagiota Efstathia Nikolaou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou, 15771, Athens, Greece
| | - Nikolaos Mylonas
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou, 15771, Athens, Greece
| | - Manousos Makridakis
- Centre of Systems Biology, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | | | - Aikaterini Iliou
- Faculty of Pharmacy, Section of Pharmaceutical Chemistry, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Stelios Zerikiotis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou, 15771, Athens, Greece
| | - Panagiotis Efentakis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou, 15771, Athens, Greece
| | - Stavros Kampoukos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou, 15771, Athens, Greece
| | - Nikolaos Kostomitsopoulos
- Centre of Clinical Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | | | - Ignatios Ikonomidis
- Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Vaia Lambadiari
- 2nd Department of Internal Medicine, Research Institute and Diabetes Center, National and Kapodistrian University of Athens, "Attikon" University Hospital, Athens, Greece
| | - Coert J Zuurbier
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | | | - Antonia Vlahou
- Centre of Systems Biology, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - George Dimitriadis
- 2nd Department of Internal Medicine, Research Institute and Diabetes Center, National and Kapodistrian University of Athens, "Attikon" University Hospital, Athens, Greece
| | | | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou, 15771, Athens, Greece.
| |
Collapse
|
37
|
Paolisso P, Bergamaschi L, Santulli G, Gallinoro E, Cesaro A, Gragnano F, Sardu C, Mileva N, Foà A, Armillotta M, Sansonetti A, Amicone S, Impellizzeri A, Casella G, Mauro C, Vassilev D, Marfella R, Calabrò P, Barbato E, Pizzi C. Infarct size, inflammatory burden, and admission hyperglycemia in diabetic patients with acute myocardial infarction treated with SGLT2-inhibitors: a multicenter international registry. Cardiovasc Diabetol 2022; 21:77. [PMID: 35570280 PMCID: PMC9107763 DOI: 10.1186/s12933-022-01506-8] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/13/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The inflammatory response occurring in acute myocardial infarction (AMI) has been proposed as a potential pharmacological target. Sodium-glucose co-transporter 2 inhibitors (SGLT2-I) currently receive intense clinical interest in patients with and without diabetes mellitus (DM) for their pleiotropic beneficial effects. We tested the hypothesis that SGLT2-I have anti-inflammatory effects along with glucose-lowering properties. Therefore, we investigated the link between stress hyperglycemia, inflammatory burden, and infarct size in a cohort of type 2 diabetic patients presenting with AMI treated with SGLT2-I versus other oral anti-diabetic (OAD) agents. METHODS In this multicenter international observational registry, consecutive diabetic AMI patients undergoing percutaneous coronary intervention (PCI) between 2018 and 2021 were enrolled. Based on the presence of anti-diabetic therapy at the admission, patients were divided into those receiving SGLT2-I (SGLT-I users) versus other OAD agents (non-SGLT2-I users). The following inflammatory markers were evaluated at different time points: white-blood-cell count, neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), neutrophil-to-platelet ratio (NPR), and C-reactive protein. Infarct size was assessed by echocardiography and by peak troponin levels. RESULTS The study population consisted of 583 AMI patients (with or without ST-segment elevation): 98 SGLT2-I users and 485 non-SGLT-I users. Hyperglycemia at admission was less prevalent in the SGLT2-I group. Smaller infarct size was observed in patients treated with SGLT2-I compared to non-SGLT2-I group. On admission and at 24 h, inflammatory indices were significantly higher in non-SGLT2-I users compared to SGLT2-I patients, with a significant increase in neutrophil levels at 24 h. At multivariable analysis, the use of SGLT2-I was a significant predictor of reduced inflammatory response (OR 0.457, 95% CI 0.275-0.758, p = 0.002), independently of age, admission creatinine values, and admission glycemia. Conversely, peak troponin values and NSTEMI occurrence were independent predictors of a higher inflammatory status. CONCLUSIONS Type 2 diabetic AMI patients receiving SGLT2-I exhibited significantly reduced inflammatory response and smaller infarct size compared to those receiving other OAD agents, independently of glucose-metabolic control. Our findings are hypothesis generating and provide new insights on the cardioprotective effects of SGLT2-I in the setting of coronary artery disease. TRIAL REGISTRATION Data are part of the ongoing observational registry: SGLT2-I AMI PROTECT. CLINICALTRIALS gov Identifier: NCT05261867.
Collapse
Affiliation(s)
- Pasquale Paolisso
- grid.416672.00000 0004 0644 9757Cardiovascular Center Aalst, OLV-Clinic, Aalst, Belgium ,grid.4691.a0000 0001 0790 385XDepartment of Advanced Biomedical Sciences, University Federico II, Naples, Italy
| | - Luca Bergamaschi
- grid.6292.f0000 0004 1757 1758Unit of Cardiology, Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, 40138 Bologna, Italy
| | - Gaetano Santulli
- grid.4691.a0000 0001 0790 385XDepartment of Advanced Biomedical Sciences, University Federico II, Naples, Italy ,International Translational Research and Medical Education (ITME) Consortium, Naples, Italy ,grid.251993.50000000121791997Department of Medicine (Division of Cardiology) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein-Sinai Diabetes Research Center, The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, New York, USA
| | - Emanuele Gallinoro
- grid.416672.00000 0004 0644 9757Cardiovascular Center Aalst, OLV-Clinic, Aalst, Belgium ,grid.9841.40000 0001 2200 8888Department of Translational Medical Sciences, University of Campania ‘Luigi Vanvitelli’, Naples, Italy
| | - Arturo Cesaro
- grid.9841.40000 0001 2200 8888Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy ,Division of Cardiology, A.O.R.N. “Sant’Anna e San Sebastiano”, Caserta, Italy
| | - Felice Gragnano
- grid.9841.40000 0001 2200 8888Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy ,Division of Cardiology, A.O.R.N. “Sant’Anna e San Sebastiano”, Caserta, Italy
| | - Celestino Sardu
- grid.9841.40000 0001 2200 8888Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Niya Mileva
- grid.410563.50000 0004 0621 0092Cardiology Clinic, ″Alexandrovska″ University Hospital, Medical University of Sofia, Sofia, Bulgaria
| | - Alberto Foà
- grid.6292.f0000 0004 1757 1758Unit of Cardiology, Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, 40138 Bologna, Italy
| | - Matteo Armillotta
- grid.6292.f0000 0004 1757 1758Unit of Cardiology, Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, 40138 Bologna, Italy
| | - Angelo Sansonetti
- grid.6292.f0000 0004 1757 1758Unit of Cardiology, Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, 40138 Bologna, Italy
| | - Sara Amicone
- grid.6292.f0000 0004 1757 1758Unit of Cardiology, Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, 40138 Bologna, Italy
| | - Andrea Impellizzeri
- grid.6292.f0000 0004 1757 1758Unit of Cardiology, Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, 40138 Bologna, Italy
| | - Gianni Casella
- grid.416290.80000 0004 1759 7093Unit of Cardiology, Maggiore Hospital, Bologna, Italy
| | - Ciro Mauro
- grid.413172.2Department of Cardiology, Hospital Cardarelli, Naples, Italy
| | | | - Raffaele Marfella
- grid.9841.40000 0001 2200 8888Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy ,grid.477084.80000 0004 1787 3414Mediterranea Cardiocentro, Naples, Italy
| | - Paolo Calabrò
- grid.9841.40000 0001 2200 8888Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy ,Division of Cardiology, A.O.R.N. “Sant’Anna e San Sebastiano”, Caserta, Italy
| | - Emanuele Barbato
- grid.416672.00000 0004 0644 9757Cardiovascular Center Aalst, OLV-Clinic, Aalst, Belgium ,grid.4691.a0000 0001 0790 385XDepartment of Advanced Biomedical Sciences, University Federico II, Naples, Italy
| | - Carmine Pizzi
- grid.6292.f0000 0004 1757 1758Unit of Cardiology, Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, 40138 Bologna, Italy
| |
Collapse
|
38
|
Wang CC, Li Y, Qian XQ, Zhao H, Wang D, Zuo GX, Wang K. Empagliflozin alleviates myocardial I/R injury and cardiomyocyte apoptosis via inhibiting ER stress-induced autophagy and the PERK/ATF4/Beclin1 pathway. J Drug Target 2022; 30:858-872. [PMID: 35400245 DOI: 10.1080/1061186x.2022.2064479] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Cuan-Cuan Wang
- Department of Cardiology, Tianjin Fifth Central Hospital, Tianjin 300450, China
| | - Ying Li
- Department of Cardiology, Tianjin Fifth Central Hospital, Tianjin 300450, China
| | - Xiao-Qian Qian
- Department of Cardiology, Tianjin Fifth Central Hospital, Tianjin 300450, China
| | - Hui Zhao
- Department of Cardiology, Tianjin Fifth Central Hospital, Tianjin 300450, China
| | - Dong Wang
- Department of Cardiology, Tianjin Fifth Central Hospital, Tianjin 300450, China
| | - Guo-Xing Zuo
- Department of Cardiology, Tianjin Fifth Central Hospital, Tianjin 300450, China
| | - Kuan Wang
- Department of Cardiology, Tianjin Fifth Central Hospital, Tianjin 300450, China
| |
Collapse
|
39
|
Patoulias D, Papadopoulos C, Kassimis G, Doumas M. Do background ischemic heart disease and baseline left ventricular ejection fraction affect cardiovascular efficacy of SGLT-2 inhibitors in heart failure with reduced ejection fraction? J Cardiovasc Med (Hagerstown) 2022; 23:e30-e32. [PMID: 34570038 DOI: 10.2459/jcm.0000000000001260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
| | | | - George Kassimis
- Second Department of Cardiology, Aristotle University of Thessaloniki, General Hospital 'Hippokration', Thessaloniki, Greece
| | - Michael Doumas
- Second Propedeutic Department of Internal Medicine
- Veterans Affairs Medical Center, George Washington University, Washington, District of Columbia, USA
| |
Collapse
|
40
|
Marketou M, Kontaraki J, Maragkoudakis S, Danelatos C, Papadaki S, Zervakis S, Plevritaki A, Vardas P, Parthenakis F, Kochiadakis G. Effects of sodium-glucose cotransporter-2 inhibitors on cardiac structural and electrical remodeling: from myocardial cytology to cardiodiabetology. Curr Vasc Pharmacol 2021; 20:178-188. [PMID: 34961447 DOI: 10.2174/1570161120666211227125033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/04/2021] [Accepted: 12/01/2021] [Indexed: 11/22/2022]
Abstract
Sodium-glucose cotransporter 2 inhibitors (SGLT2i) have changed the clinical landscape of diabetes mellitus (DM) therapy through their favourable effects on cardiovascular outcomes. Notably, the use of SGLT2i has been linked to cardiovascular benefits regardless of DM status, while their pleiotropic actions remain to be fully elucidated. What we do know is that SGLT2i exert beneficial effects even at the level of the myocardial cell, and that these are linked to an improvement in the energy substrate, resulting in less inflammation and fibrosis. SGLT2i ameliorate myocardial extracellular matrix remodeling, cardiomyocyte stiffness and concentric hypertrophy, achieving beneficial remodeling of the left ventricle with significant implications for the pathogenesis and outcome of heart failure. Most studies show a significant improvement in markers of diastolic dysfunction along with a reduction in left ventricular hypertrophy. In addition to these effects there is electrophysiological remodeling, which explains initial data suggesting that SGLT2i have an antiarrhythmic action against both atrial and ventricular arrhythmias. However, future studies need to clarify not only the exact mechanisms of this beneficial functional, structural, and electrophysiological cardiac remodeling, but also its magnitude, and to determine whether this is a class or a drug effect.
Collapse
Affiliation(s)
- Maria Marketou
- Cardiology Department, Heraklion University Hospital, Crete Greece
| | - Joanna Kontaraki
- Laboratory of Molecular Cardiology, University of Crete, School of Medicine, Crete, Greece
| | | | | | - Sofia Papadaki
- Cardiology Department, Heraklion University Hospital, Crete Greece
| | - Stelios Zervakis
- Cardiology Department, Heraklion University Hospital, Crete Greece
| | | | - Panos Vardas
- Cardiovascular Section, Mitera Hospital, Hygeia Group, Athens Greece
| | | | | |
Collapse
|
41
|
Jiang Y, Zheng R, Sang H. Cost-Effectiveness of Adding SGLT2 Inhibitors to Standard Treatment for Heart Failure With Reduced Ejection Fraction Patients in China. Front Pharmacol 2021; 12:733681. [PMID: 34858172 PMCID: PMC8631914 DOI: 10.3389/fphar.2021.733681] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/20/2021] [Indexed: 12/11/2022] Open
Abstract
Objective: To evaluate the economics and effectiveness of adding dapagliflozin or empagliflozin to the standard treatment for heart failure (HF) for patients with reduced ejection fraction (HFrEF) in China. Methods: A Markov model was developed to project the clinical and economic outcomes of adding dapagliflozin or empagliflozin to the standard treatment for 66-year-old patients with HFrEF. A cost-utility analysis was performed based mostly on data from the empagliflozin outcome trial in patients with chronic heart failure and a reduced ejection fraction (EMPEROR-Reduced) study and the dapagliflozin and prevention of adverse outcomes in heart failure (DAPA-HF) trial. The primary outcomes were measured via total and incremental costs and quality-adjusted life years (QALYs) and the incremental cost-effectiveness ratio (ICER). Results: In China, compared to the standard treatment, although adding dapagliflozin to the standard treatment in the treatment of HFrEF was more expensive ($4,870.68 vs. $3,596.25), it was more cost-effective (3.87 QALYs vs. 3.64 QALYs), resulting in an ICER of $5,541.00 per QALY. Similarly, adding empagliflozin was more expensive ($5,021.93 vs. $4,118.86) but more cost-effective (3.66 QALYs vs. 3.53 QALYs), resulting in an ICER of $6,946.69 per QALY. A sensitivity analysis demonstrated the robustness of the model in identifying cardiovascular death as a significant driver of cost-effectiveness. A probabilistic sensitivity analysis indicated that when the willingness-to-pay was $11,008.07 per QALY, the probability of the addition of dapagliflozin or empagliflozin being cost-effective was 70.5 and 55.2%, respectively. A scenario analysis showed that the cost of hospitalization, diabetes status, and time horizon had a greater impact on ICER. Conclusion: Compared with standard treatments with or without empagliflozin, adding dapagliflozin to the standard treatment in the treatment of HFrEF in China was extremely cost-effective.
Collapse
Affiliation(s)
- Yaohui Jiang
- Department Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rujie Zheng
- Department Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Haiqiang Sang
- Department Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
42
|
Abstract
Sodium glucose cotransporter 2 (SGLT-2) inhibitors are the latest class of antidiabetic medications. They prevent glucose reabsorption in the proximal convoluted tubule to decrease blood sugar. Several animal studies revealed that SGLT-2 is profoundly involved in the inflammatory response, fibrogenesis, and regulation of numerous intracellular signaling pathways. Likewise, SGLT-2 inhibitors markedly attenuated inflammation and fibrogenesis and improved the function of damaged organ in animal studies, observational studies, and clinical trials. SGLT-2 inhibitors can decrease blood pressure and ameliorate hypertriglyceridemia and obesity. Likewise, they improve the outcome of cardiovascular diseases such as heart failure, arrhythmias, and ischemic heart disease. SGLT-2 inhibitors are associated with lower cardiovascular and all-cause mortality as well. Meanwhile, they protect against nonalcoholic fatty liver disease (NAFLD), chronic kidney disease, acute kidney injury, and improve micro- and macroalbuminuria. SGLT-2 inhibitors can reprogram numerous signaling pathways to improve NAFLD, cardiovascular diseases, and renal diseases. For instance, they enhance lipolysis, ketogenesis, mitochondrial biogenesis, and autophagy while they attenuate the renin-angiotensin-aldosterone system, lipogenesis, endoplasmic reticulum stress, oxidative stress, apoptosis, and fibrogenesis. This review explains the beneficial effects of SGLT-2 inhibitors on NAFLD and cardiovascular and renal diseases and dissects the underlying molecular mechanisms in detail. This narrative review explains the beneficial effects of SGLT-2 inhibitors on NAFLD and cardiovascular and renal diseases using the results of latest observational studies, clinical trials, and meta-analyses. Thereafter, it dissects the underlying molecular mechanisms involved in the clinical effects of SGLT-2 inhibitors on these diseases.
Collapse
Affiliation(s)
- Moein Ala
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
43
|
Comità S, Femmino S, Thairi C, Alloatti G, Boengler K, Pagliaro P, Penna C. Regulation of STAT3 and its role in cardioprotection by conditioning: focus on non-genomic roles targeting mitochondrial function. Basic Res Cardiol 2021; 116:56. [PMID: 34642818 PMCID: PMC8510947 DOI: 10.1007/s00395-021-00898-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 12/11/2022]
Abstract
Ischemia–reperfusion injury (IRI) is one of the biggest challenges for cardiovascular researchers given the huge death toll caused by myocardial ischemic disease. Cardioprotective conditioning strategies, namely pre- and post-conditioning maneuvers, represent the most important strategies for stimulating pro-survival pathways essential to preserve cardiac health. Conditioning maneuvers have proved to be fundamental for the knowledge of the molecular basis of both IRI and cardioprotection. Among this evidence, the importance of signal transducer and activator of transcription 3 (STAT3) emerged. STAT3 is not only a transcription factor but also exhibits non-genomic pro-survival functions preserving mitochondrial function from IRI. Indeed, STAT3 is emerging as an influencer of mitochondrial function to explain the cardioprotection phenomena. Studying cardioprotection, STAT3 proved to be crucial as an element of the survivor activating factor enhancement (SAFE) pathway, which converges on mitochondria and influences their function by cross-talking with other cardioprotective pathways. Clearly there are still some functional properties of STAT3 to be discovered. Therefore, in this review, we highlight the evidence that places STAT3 as a promoter of the metabolic network. In particular, we focus on the possible interactions of STAT3 with processes aimed at maintaining mitochondrial functions, including the regulation of the electron transport chain, the production of reactive oxygen species, the homeostasis of Ca2+ and the inhibition of opening of mitochondrial permeability transition pore. Then we consider the role of STAT3 and the parallels between STA3/STAT5 in cardioprotection by conditioning, giving emphasis to the human heart and confounders.
Collapse
Affiliation(s)
- Stefano Comità
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043, Torino, TO, Italy
| | - Saveria Femmino
- Department of Medical Sciences, University of Turin, Torino, Italy
| | - Cecilia Thairi
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043, Torino, TO, Italy
| | | | - Kerstin Boengler
- Institute of Physiology, University of Giessen, Giessen, Germany
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043, Torino, TO, Italy.
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043, Torino, TO, Italy.
| |
Collapse
|
44
|
de Koning MSLY, Westenbrink BD, Assa S, Garcia E, Connelly MA, van Veldhuisen DJ, Dullaart RPF, Lipsic E, van der Harst P. Association of Circulating Ketone Bodies With Functional Outcomes After ST-Segment Elevation Myocardial Infarction. J Am Coll Cardiol 2021; 78:1421-1432. [PMID: 34593124 DOI: 10.1016/j.jacc.2021.07.054] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 01/25/2023]
Abstract
BACKGROUND Circulating ketone bodies (KBs) are increased in patients with heart failure (HF), corresponding with increased cardiac KB metabolism and HF severity. However, the role of circulating KBs in ischemia/reperfusion remains unknown. OBJECTIVES This study sought to investigate longitudinal changes of KBs and their associations with functional outcomes in patients presenting with ST-segment elevation myocardial infarction (STEMI). METHODS KBs were measured in 369 participants from a randomized trial on early metformin therapy after STEMI. Nonfasting plasma concentrations of KBs (β-hydroxybutyrate, acetoacetate, and acetone) were measured by nuclear magnetic resonance spectroscopy at presentation, at 24 hours, and after 4 months. Myocardial infarct size and left ventricular ejection fraction (LVEF) were determined by cardiac magnetic resonance imaging at 4 months. Associations of circulating KBs with infarct size and LVEF were determined using multivariable linear regression analyses. RESULTS Circulating KBs were high at presentation with STEMI (median total KBs: 520 μmol/L; interquartile range [IQR]: 315-997 μmol/L). At 24 hours after reperfusion, KBs were still high compared with levels at 4-month follow-up (206 μmol/L [IQR: 174-246] vs 166 μmol/L [IQR: 143-201], respectively; P < 0.001). Increased KB concentrations at 24 hours were independently associated with larger myocardial infarct size (total KBs, per 100 μmol/L: β = 1.56; 95% confidence interval: 0.29-2.83; P = 0.016) and lower LVEF (β = -1.78; 95% CI: (-3.17 to -0.39; P = 0.012). CONCLUSIONS Circulating KBs are increased in patients presenting with STEMI. Higher KBs at 24 hours are associated with functional outcomes after STEMI, which suggests a potential role for ketone metabolism in response to myocardial ischemia. (Metabolic Modulation With Metformin to Reduce Heart Failure After Acute Myocardial Infarction: Glycometabolic Intervention as Adjunct to Primary Coronary Intervention in ST Elevation Myocardial Infarction (GIPS-III): a Randomized Controlled Trial; NCT01217307).
Collapse
Affiliation(s)
- Marie-Sophie L Y de Koning
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - B Daan Westenbrink
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Solmaz Assa
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Erwin Garcia
- Laboratory Corporation of America Holdings (Labcorp), Morrisville, North Carolina, USA
| | - Margery A Connelly
- Laboratory Corporation of America Holdings (Labcorp), Morrisville, North Carolina, USA
| | - Dirk J van Veldhuisen
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Robin P F Dullaart
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Erik Lipsic
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Pim van der Harst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
45
|
Xiao Y, Phelp P, Wang Q, Bakker D, Nederlof R, Hollmann MW, Zuurbier CJ. Cardioprotecive Properties of Known Agents in Rat Ischemia-Reperfusion Model Under Clinically Relevant Conditions: Only the NAD Precursor Nicotinamide Riboside Reduces Infarct Size in Presence of Fentanyl, Midazolam and Cangrelor, but Not Propofol. Front Cardiovasc Med 2021; 8:712478. [PMID: 34527711 PMCID: PMC8435675 DOI: 10.3389/fcvm.2021.712478] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/29/2021] [Indexed: 01/12/2023] Open
Abstract
Background: Cardioprotective strategies against ischemia-reperfusion injury (IRI) that remain effective in the clinical arena need to be developed. Therefore, maintained efficacy of cardioprotective strategies in the presence of drugs routinely used clinically (e.g., opiates, benzodiazepines, P2Y12 antagonist, propofol) need to be identified in preclinical models. Methods: Here, we examined the efficacy of promising cardioprotective compounds [fingolimod (Fingo), empagliflozin (Empa), melatonin (Mela) and nicotinamide riboside (NR)] administered i.v. as bolus before start ischemia. Infarct size as percentage of the area of risk (IS%) was determined following 25 min of left ascending coronary (LAD) ischemia and 2 h of reperfusion in a fentanyl-midazolam anesthetized IRI rat model. Plasma lactate dehydrogenase (LDH) activity at 30 min reperfusion was determined as secondary outcome parameter. Following pilot dose-response experiments of each compound (3 dosages, n = 4-6 animals per dosage), potential cardioprotective drugs at the optimal observed dosage were subsequently tested alone or in combination (n = 6-8 animals per group). The effective treatment was subsequently tested in the presence of a P2Y12 antagonist (cangrelor; n = 6/7) or propofol aesthesia (n = 6 both groups). Results: Pilot studies suggested potential cardioprotective effects for 50 mg/kg NR (p = 0.005) and 500 μg/kg melatonin (p = 0.12), but not for Empa or Fingo. Protection was subsequently tested in a new series of experiments for solvents, NR, Mela and NR+Mela. Results demonstrated that only singular NR was able to reduce IS% (30 ± 14 vs. 60 ± 16%, P = 0.009 vs. control). Mela (63 ± 18%) and NR+Mela (47 ± 15%) were unable to significantly decrease IS%. NR still reduced IS in the presence of cangrelor (51 ± 18 vs. 71 ± 4%, P = 0.016 vs. control), but lost protection in the presence of propofol anesthesia (62 ± 16 vs. 60 ± 14%, P = 0.839 vs. control). LDH activity measurements supported all IS% results. Conclusion: This observational study suggests that NR is a promising cardioprotective agent to target cardiac ischemia-reperfusion injury in clinical conditions employing opioid agonists, benzodiazepines and platelet P2Y12 inhibitors, but not propofol.
Collapse
Affiliation(s)
- Yang Xiao
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, Netherlands
| | - Philippa Phelp
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, Netherlands
| | - Qian Wang
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, Netherlands
| | - Diane Bakker
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, Netherlands
| | - Rianne Nederlof
- Institut für Herz- und Kreislaufphysiologie, Heinrich- Heine- Universität Düsseldorf, Düsseldorf, Germany
| | - Markus W Hollmann
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, Netherlands
| | - Coert J Zuurbier
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
46
|
Hoong CWS, Chua MWJ. SGLT2 Inhibitors as Calorie Restriction Mimetics: Insights on Longevity Pathways and Age-Related Diseases. Endocrinology 2021; 162:6226811. [PMID: 33857309 DOI: 10.1210/endocr/bqab079] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Indexed: 02/08/2023]
Abstract
Sodium-glucose cotransporter-2 (SGLT2) inhibitors induce glycosuria, reduce insulin levels, and promote fatty acid oxidation and ketogenesis. By promoting a nutrient deprivation state, SGLT2 inhibitors upregulate the energy deprivation sensors AMPK and SIRT1, inhibit the nutrient sensors mTOR and insulin/IGF1, and modulate the closely linked hypoxia-inducible factor (HIF)-2α/HIF-1α pathways. Phosphorylation of AMPK and upregulation of adiponectin and PPAR-α favor a reversal of the metabolic syndrome which have been linked to suppression of chronic inflammation. Downregulation of insulin/IGF1 pathways and mTOR signaling from a reduction in glucose and circulating amino acids promote cellular repair mechanisms, including autophagy and proteostasis which confer cellular stress resistance and attenuate cellular senescence. SIRT1, another energy sensor activated by NAD+ in nutrient-deficient states, is reciprocally activated by AMPK, and can deacetylate and activate transcription factors, such as PCG-1α, mitochondrial transcription factor A (TFAM), and nuclear factor E2-related factor (NRF)-2, that regulate mitochondrial biogenesis. FOXO3 transcription factor which target genes in stress resistance, is also activated by AMPK and SIRT1. Modulation of these pathways by SGLT2 inhibitors have been shown to alleviate metabolic diseases, attenuate vascular inflammation and arterial stiffness, improve mitochondrial function and reduce oxidative stress-induced tissue damage. Compared with other calorie restriction mimetics such as metformin, rapamycin, resveratrol, and NAD+ precursors, SGLT2 inhibitors appear to be the most promising in the treatment of aging-related diseases, due to their regulation of multiple longevity pathways that closely resembles that achieved by calorie restriction and their established efficacy in reducing cardiovascular events and all-cause mortality. Evidence is compelling for the role of SGLT2 inhibitors as a calorie restriction mimetic in anti-aging therapeutics.
Collapse
Affiliation(s)
- Caroline W S Hoong
- Division of Endocrinology, Department of General Medicine, Woodlands Health Campus, National Healthcare Group Singapore, Woodlands Health Campus Singapore, 768024, Singapore
| | - Marvin W J Chua
- Endocrinology Service, Department of General Medicine, Sengkang General Hospital, SingHealth Group Singapore, Sengkang General Hospital Singapore, 544886, Singapore
| |
Collapse
|
47
|
Yang Y, Jiang K, Liu X, Qin M, Xiang Y. CaMKII in Regulation of Cell Death During Myocardial Reperfusion Injury. Front Mol Biosci 2021; 8:668129. [PMID: 34141722 PMCID: PMC8204011 DOI: 10.3389/fmolb.2021.668129] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/10/2021] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide. In spite of the mature managements of myocardial infarction (MI), post-MI reperfusion (I/R) injury results in high morbidity and mortality. Cardiomyocyte Ca2+ overload is a major factor of I/R injury, initiating a cascade of events contributing to cardiomyocyte death and myocardial dysfunction. Ca2+/calmodulin-dependent protein kinase II (CaMKII) plays a critical role in cardiomyocyte death response to I/R injury, whose activation is a key feature of myocardial I/R in causing intracellular mitochondrial swelling, endoplasmic reticulum (ER) Ca2+ leakage, abnormal myofilament contraction, and other adverse reactions. CaMKII is a multifunctional serine/threonine protein kinase, and CaMKIIδ, the dominant subtype in heart, has been widely studied in the activation, location, and related pathways of cardiomyocytes death, which has been considered as a potential targets for pharmacological inhibition. In this review, we summarize a brief overview of CaMKII with various posttranslational modifications and its properties in myocardial I/R injury. We focus on the molecular mechanism of CaMKII involved in regulation of cell death induced by myocardial I/R including necroptosis and pyroptosis of cardiomyocyte. Finally, we highlight that targeting CaMKII modifications and cell death involved pathways may provide new insights to understand the conversion of cardiomyocyte fate in the setting of myocardial I/R injury.
Collapse
Affiliation(s)
- Yingjie Yang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Kai Jiang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xu Liu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Mu Qin
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yaozu Xiang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
48
|
Naidu SS, Abbott JD, Bagai J, Blankenship J, Garcia S, Iqbal SN, Kaul P, Khuddus MA, Kirkwood L, Manoukian SV, Patel MR, Skelding K, Slotwiner D, Swaminathan RV, Welt FG, Kolansky DM. SCAI expert consensus update on best practices in the cardiac catheterization laboratory: This statement was endorsed by the American College of Cardiology (ACC), the American Heart Association (AHA), and the Heart Rhythm Society (HRS) in April 2021. Catheter Cardiovasc Interv 2021; 98:255-276. [PMID: 33909349 DOI: 10.1002/ccd.29744] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 04/23/2021] [Indexed: 12/28/2022]
Abstract
The current document commissioned by the Society for Cardiovascular Angiography and Interventions (SCAI) and endorsed by the American College of Cardiology, the American Heart Association, and Heart Rhythm Society represents a comprehensive update to the 2012 and 2016 consensus documents on patient-centered best practices in the cardiac catheterization laboratory. Comprising updates to staffing and credentialing, as well as evidence-based updates to the pre-, intra-, and post-procedural logistics, clinical standards and patient flow, the document also includes an expanded section on CCL governance, administration, and approach to quality metrics. This update also acknowledges the collaboration with various specialties, including discussion of the heart team approach to management, and working with electrophysiology colleagues in particular. It is hoped that this document will be utilized by hospitals, health systems, as well as regulatory bodies involved in assuring and maintaining quality, safety, efficiency, and cost-effectiveness of patient throughput in this high volume area.
Collapse
Affiliation(s)
- Srihari S Naidu
- Department of Cardiology, Westchester Medical Center and New York Medical College, Valhalla, New York, USA
| | - J Dawn Abbott
- Cardiovascular Institute of Lifespan, Division of Cardiology, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Jayant Bagai
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - James Blankenship
- Cardiology Division, The University of New Mexico, Albuquerque, New Mexico, USA
| | | | - Sohah N Iqbal
- Mass General Brigham Salem Hospital, Salem, Massachusetts, USA
| | | | - Matheen A Khuddus
- The Cardiac and Vascular Institute and North Florida Regional Medical Center, Gainesville, Florida, USA
| | - Lorrena Kirkwood
- Department of Cardiology, Westchester Medical Center and New York Medical College, Valhalla, New York, USA
| | | | - Manesh R Patel
- Duke University Medical Center and Duke Clinical Research Institute, Durham, North Carolina, USA
| | | | - David Slotwiner
- Division of Cardiology, New York Presbyterian, Weill Cornell Medicine Population Health Sciences, Queens, New York, USA
| | - Rajesh V Swaminathan
- Duke University Medical Center and Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Frederick G Welt
- Division of Cardiovascular Medicine, University of Utah Health, Salt Lake City, Utah, USA
| | - Daniel M Kolansky
- Division of Cardiovascular Medicine, Perelman School of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
49
|
Xiao M, Zeng W, Wang J, Yao F, Peng Z, Liu G, Yu L, Wei W, Zhou S, Li K, Wu L, Zhu K, Guan Y, Chen H, Liu Z, Chen J. Exosomes Protect Against Acute Myocardial Infarction in Rats by Regulating the Renin-Angiotensin System. Stem Cells Dev 2021; 30:622-631. [PMID: 33765842 DOI: 10.1089/scd.2020.0132] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The renin-angiotensin system (RAS) has been suggested to play an important role in cardiac remodeling after acute myocardial infarction (AMI). We have confirmed that bone marrow mesenchymal stem cell-derived exosomes (BMSC-EX) had similar types of repair like effects upon tissues as BMSC, but the mechanisms remain unknown. BMSC were cultured to the third generation and were induced to release exosomes. Rats were injected with exosomes (100 μg/mL) or stem cells (1 × 106/mL) through the tail vein immediately after AMI was built, compared to those treated with physiological saline. Thereafter, all groups were analyzed for cardiac function, infarction sizes, and the levels of expression of BNP, ACE, ACE2, AngII, Ang1-7, and other factors in the plasma. After H2O2 makes contact with H9C2 cardiomyocytes, cell proliferation activity and apoptotic rates were measured by using CCK8 kits, to facilitate investigation of the effect of exosomes on H9C2 cells. In vivo, the index of cardiac remodeling and cardiac function was improved in both groups of exosomes and stem cells after AMI. Furthermore, exosomes may have helped to regulate the balance of the RAS system, upregulate ACE2-Ang1-7-Mas, and downregulate the ACE-AngII-ATIR pathway. Therefore, its effects were such as to accelerate the conversion of Ang II to Ang 1-7, thereby improving cardiac remodeling and forming sustained myocardial protection. In vitro, exosomal intervention was found to have increased the levels of activity of H9C2 cardiomyocytes under H2O2 injury and improved adverse effects of AngII upon H9C2 cells. All procedures for this study were reviewed and approved by the Institutional Animal Care and Use Committee (IACUC) at Guangdong Medical University. BMSC-EX improved cardiac remodeling and cardiac function, and had effects upon RAS system-related factors in plasma. Similarly, BMSC-EX also helped to protect H9C2 cells under attack from H2O2 or AngII, and may thus play beneficial roles by facilitating regulation of the balance of the RAS system.
Collapse
Affiliation(s)
- Mengyuan Xiao
- Cardiovascular Medicine Center and Affiliated Hospital of Guangdong Medical University, Zhangjiang, China
| | - Weikai Zeng
- Cardiovascular Medicine Center and Affiliated Hospital of Guangdong Medical University, Zhangjiang, China
| | - Junxian Wang
- Gerontology Center, Affiliated Hospital of Guangdong Medical University, Zhangjiang, China
| | - Feng Yao
- Cardiovascular Medicine Center and Affiliated Hospital of Guangdong Medical University, Zhangjiang, China
| | - Zijian Peng
- Cardiovascular Medicine Center and Affiliated Hospital of Guangdong Medical University, Zhangjiang, China
| | - Guangyan Liu
- Gerontology Center, Affiliated Hospital of Guangdong Medical University, Zhangjiang, China
| | - Liqin Yu
- Cardiovascular Medicine Center and Affiliated Hospital of Guangdong Medical University, Zhangjiang, China
| | - Wenyan Wei
- Gerontology Center, Affiliated Hospital of Guangdong Medical University, Zhangjiang, China
| | - Shengzhi Zhou
- Cardiovascular Medicine Center and Affiliated Hospital of Guangdong Medical University, Zhangjiang, China
| | - Kaize Li
- Cardiovascular Medicine Center and Affiliated Hospital of Guangdong Medical University, Zhangjiang, China
| | - Luyao Wu
- Cardiovascular Medicine Center and Affiliated Hospital of Guangdong Medical University, Zhangjiang, China
| | - Kunpeng Zhu
- Cardiovascular Medicine Center and Affiliated Hospital of Guangdong Medical University, Zhangjiang, China
| | - Yuejie Guan
- Gerontology Center, Affiliated Hospital of Guangdong Medical University, Zhangjiang, China
| | - Huanyu Chen
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhenjun Liu
- Cardiovascular Medicine Center and Affiliated Hospital of Guangdong Medical University, Zhangjiang, China
| | - Jianying Chen
- Cardiovascular Medicine Center and Affiliated Hospital of Guangdong Medical University, Zhangjiang, China
| |
Collapse
|
50
|
Sayour AA, Celeng C, Oláh A, Ruppert M, Merkely B, Radovits T. Sodium-glucose cotransporter 2 inhibitors reduce myocardial infarct size in preclinical animal models of myocardial ischaemia-reperfusion injury: a meta-analysis. Diabetologia 2021; 64:737-748. [PMID: 33483761 PMCID: PMC7940278 DOI: 10.1007/s00125-020-05359-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023]
Abstract
AIMS/HYPOTHESIS Large cardiovascular outcome trials demonstrated that the cardioprotective effects of sodium-glucose cotransporter 2 (SGLT2) inhibitors might reach beyond glucose-lowering action. In this meta-analysis, we sought to evaluate the potential infarct size-modulating effect of SGLT2 inhibitors in preclinical studies. METHODS In this preregistered meta-analysis (PROSPERO: CRD42020189124), we included placebo-controlled, interventional studies of small and large animal models of myocardial ischaemia-reperfusion injury, testing the effect of SGLT2 inhibitor treatment on myocardial infarct size (percentage of area at risk or total area). Standardised mean differences (SMDs) were calculated and pooled using random-effects method. We evaluated heterogeneity by computing Τ2 and I2 values. Meta-regression was performed to explore prespecified subgroup differences according to experimental protocols and their contribution to heterogeneity was assessed (pseudo-R2 values). RESULTS We identified ten eligible publications, reporting 16 independent controlled comparisons on a total of 224 animals. Treatment with SGLT2 inhibitor significantly reduced myocardial infarct size compared with placebo (SMD = -1.30 [95% CI -1.79, -0.81], p < 0.00001), referring to a 33% [95% CI 20%, 47%] difference. Heterogeneity was moderate (Τ2 = 0.58, I2 = 60%). SGLT2 inhibitors were only effective when administered to the intact organ system, but not to isolated hearts (p interaction <0.001, adjusted pseudo-R2 = 47%). While acute administration significantly reduced infarct size, chronic treatment was superior (p interaction <0.001, adjusted pseudo-R2 = 85%). The medications significantly reduced infarct size in both diabetic and non-diabetic animals, favouring the former (p interaction = 0.030, adjusted pseudo-R2 = 12%). Treatment was equally effective in rats and mice, as well as in a porcine model. Individual study quality scores were not related to effect estimates (p = 0.33). The overall effect estimate remained large even after adjusting for severe forms of publication bias. CONCLUSIONS/INTERPRETATION The glucose-lowering SGLT2 inhibitors reduce myocardial infarct size in animal models independent of diabetes. Future in vivo studies should focus on clinical translation by exploring whether SGLT2 inhibitors limit infarct size in animals with relevant comorbidities, on top of loading doses of antiplatelet agents. Mechanistic studies should elucidate the potential relationship between the infarct size-lowering effect of SGLT2 inhibitors and the intact organ system.
Collapse
Affiliation(s)
- Alex Ali Sayour
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary.
| | - Csilla Celeng
- University Medical Center Utrecht, Utrecht, the Netherlands
| | - Attila Oláh
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Mihály Ruppert
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Tamás Radovits
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| |
Collapse
|