1
|
Wang Y, Qin Y, Zhang J, Wu A, Qin X, Du L, Zhang H, Guo X, Zhang S. Sodium-Glucose Cotransporter-2 Inhibitors and Diabetic-Ketoacidosis in T2DM Patients: An Updated Meta-Analysis and a Mendelian Randomization Analysis. Clin Pharmacol Ther 2025; 117:1661-1669. [PMID: 40070044 DOI: 10.1002/cpt.3615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 02/10/2025] [Indexed: 05/20/2025]
Abstract
To evaluate the association of sodium-glucose cotransporter 2 inhibitors (SGLT2i) with diabetic ketoacidosis (DKA) in type 2 diabetes mellitus (T2DM) patients across different subgroups, we searched randomized controlled trials (RCTs) comparing SGLT2i with the control groups among T2DM patients and including DKA as a safety outcome. Pooled risk ratios (RRs) were calculated using random or fixed-effects models, as appropriate. An inverse-variance-weighted Mendelian randomization (MR) analysis was performed to estimate the genetic correlation. Twenty-two trials involving 80,235 patients were included. SGLT2i increased the risk of DKA compared to the control groups (RR 2.32, 95% CI 1.64-3.27). The risk was significantly increased in patients with higher HbA1c levels (> 7.9%) (RR 2.24, 95% CI 1.59-3.14), but not in those with lower HbA1c levels (≤ 7.9%) (RR 1.05, 95% CI 0.49-2.26; interaction P = 0.034). SGLT2i increased DKA risk in chronic kidney disease (CKD) (RR 2.70, 95% CI 1.55-4.71) and high atherosclerotic cardiovascular disease (ASCVD) risk trials (RR 2.46, 95% CI 1.47-4.11) but not significantly in heart failure (HF) trials (RR 1.23, 95% CI 0.51-2.96). Moreover, in the HF trials, SGLT2i consistently did not increase the risk of DKA in any clinical subgroups. Nevertheless, MR analysis still confirmed a genetic association between SGLT2i and the risk of DKA among overall T2DM patients. SGLT2i may increase the risk of DKA in T2DM patients, particularly in patients with higher levels of HbA1c and those with comorbid CKD or at high-risk ASCVD. However, the increased risk was not significant in patients with HF.
Collapse
Affiliation(s)
- Yufei Wang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuhan Qin
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Anhu Wu
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaohan Qin
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Le Du
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Huabing Zhang
- Department of Endocrinology, Key Laboratory of Endocrinology of the National Health Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoxiao Guo
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuyang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
2
|
Hu Y, Chen X, Zhao Q, Li G, Zhang H, Ma Z, Yu H, Zeng Q, Zhang H, Xu D. Berberine improves cardiac insufficiency through AMPK/PGC-1α signaling-mediated mitochondrial homeostasis and apoptosis in HFpEF mice. Int Immunopharmacol 2025; 155:114613. [PMID: 40222275 DOI: 10.1016/j.intimp.2025.114613] [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: 02/11/2025] [Revised: 03/23/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Heart failure (HF) with preserved ejection fraction (HFpEF) accounts for approximately half of cases of HF and is frequently clinically underdiagnosed. Although new therapies continue to emerge, determining optimal treatment strategies persists as a key clinical dilemma. Berberine(BBR), an isoquinoline alkaloid, is known to attenuate HF with reduced ejection fraction. PURPOSE In this study, we explored the cardiovascular benefits of BBR in diastolic dysfunction associated with HFpEF, both in vitro and in vivo. METHODS In vivo, adult male mice were fed with chow or a high-fat diet (60 % calories from lard) with L-NAME (0.5 g/L in drinking water) for 15 weeks. During the last 4 weeks, BBR (100 mg/Kg/d and 200 mg/Kg/d) was administered orally. Rat cardiac myoblast H9C2 cells were pretreated with BBR for 2 h, followed by exposure to palmitic acid (PA, 100 μM) for 24 h. RESULTS Exposure to a high-fat stimulation led to p-AMPK and PGC-1α downregulation, apoptotic cascade activation, elevated mt-ROS production, and disruption of mitochondrial homeostasis both in vivo and in vitro. Notably, BBR intervention elevated the expressions of p-AMPK and PGC-1α, inhibited apoptotic reaction, reduced mt-ROS, ameliorated TFAM/NRF1-mediated mitochondrial biogenesis disorder, alleviated mitochondrial impairment, and improved cardiac function. On the other hand, AMPK knockdown abolished the beneficial impact of BBR. Collectively, our findings underscored the cardioprotective role of BBR in maintaining mitochondrial homeostasis and preventing apoptosis, achieved through the modulation of the AMPK/PGC-1α pathway. CONCLUSIONS In summary, BBR possesses protective activity against cardiac insufficiency in HFpEF by maintaining mitochondrial homeostasis and inhibiting apoptosis.
Collapse
Affiliation(s)
- Yingchun Hu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Xiaoyu Chen
- Department of Nephrology, Rheumatism and Immunology, Chongqing Jiulongpo People's Hospital, Chongqing 400050, China
| | - Qiming Zhao
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Guohao Li
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Hao Zhang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Zhuang Ma
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Hao Yu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Qingchun Zeng
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Hanping Zhang
- Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Dingli Xu
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China.
| |
Collapse
|
3
|
Cinti F, Laborante R, Cappannoli L, Morciano C, Gugliandolo S, Pontecorvi A, Burzotta F, Donniacuo M, Cappetta D, Patti G, Giaccari A, D'Amario D. The effects of SGLT2i on cardiac metabolism in patients with HFpEF: Fact or fiction? Cardiovasc Diabetol 2025; 24:208. [PMID: 40369599 PMCID: PMC12079913 DOI: 10.1186/s12933-025-02767-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/29/2025] [Indexed: 05/16/2025] Open
Abstract
The rising prevalence of Type 2 diabetes (T2D) has been closely associated with an increased incidence of cardiovascular diseases, particularly heart failure with preserved ejection fraction (HFpEF). Cardiometabolic disturbances in T2D, such as insulin resistance, hyperglycemia, and dyslipidemia, contribute to both microvascular and macrovascular complications, thereby intensifying the risk of heart failure. Sodium-glucose cotransporter-2 inhibitors (SGLT2i), initially developed as glucose-lowering agents for T2D, have demonstrated promising cardiovascular benefits in patients with heart failure, including those with preserved ejection fraction (HFpEF), regardless of T2D status. These benefits include reduced heart failure hospitalization rates and improvements in various metabolic parameters. This review aims to critically examine the effects of SGLT2i on cardiac metabolism in HFpEF, evaluating whether the observed benefits can truly be attributed to their impact on myocardial energy regulation or whether they represent other, potentially confounding, mechanisms. We will focus on the key metabolic processes possibly modulated by SGLT2i, including myocardial glucose utilization, fatty acid oxidation, and mitochondrial function, and explore their effects on heart failure pathophysiology. Additionally, we will address the role of SGLT2i in other pathogenetic factors involved in HFpEF, such as sodium and fluid balance, inflammation, and fibrosis, and question the extent to which these mechanisms contribute to the observed clinical benefits. By synthesizing the current evidence, this review will provide an in-depth analysis of the mechanisms through which SGLT2i may influence cardiac metabolism in HFpEF, assessing whether their effects are supported by robust scientific data or remain speculative. We will also discuss the potential for personalized treatment strategies, based on individual patient characteristics, to optimize the therapeutic benefits of SGLT2i in managing both T2D and cardiovascular risk. This review seeks to clarify the true clinical utility of SGLT2i in the management of cardiometabolic diseases and HFpEF, offering insights into their role in improving long-term cardiovascular outcomes.
Collapse
Affiliation(s)
- Francesca Cinti
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Dipartimento di Medicina e Chirurgia Traslazionale, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Renzo Laborante
- Dipartimento di Scienze Cardiovascolari- CUORE, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Luigi Cappannoli
- Dipartimento di Scienze Cardiovascolari- CUORE, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Cassandra Morciano
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Dipartimento di Medicina e Chirurgia Traslazionale, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Shawn Gugliandolo
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Dipartimento di Medicina e Chirurgia Traslazionale, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Alfredo Pontecorvi
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Dipartimento di Medicina e Chirurgia Traslazionale, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Francesco Burzotta
- Dipartimento di Scienze Cardiovascolari- CUORE, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Maria Donniacuo
- Dipartimento di Medicina Sperimentale, Università del Salento, Lecce, Italy
| | - Donato Cappetta
- Dipartimento di Medicina Sperimentale, Università del Salento, Lecce, Italy
| | - Giuseppe Patti
- Dipartimento di Medicina Traslazionale (DiMET), Università del Piemonte Orientale, Novara, Italy
| | - Andrea Giaccari
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Dipartimento di Medicina e Chirurgia Traslazionale, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy.
| | - Domenico D'Amario
- Dipartimento di Medicina Traslazionale (DiMET), Università del Piemonte Orientale, Novara, Italy.
| |
Collapse
|
4
|
Tabatabaei Dakhili SA, Yang K, Stenlund MJ, Ussher JR. The multifaceted roles of ketones in physiology. Exp Physiol 2025. [PMID: 40349316 DOI: 10.1113/ep092243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/16/2025] [Indexed: 05/14/2025]
Abstract
The production of ketones, referred to as ketogenesis, plays an essential role in maintaining energy homeostasis during prolonged fasting/starvation, which primarily stems from its ability to serve as a fuel source to support neuronal ATP production, thereby limiting muscle wasting. Over the years, the field has come to appreciate that ketones are much more than just a fuel source supporting neuronal metabolism, as many other oxidative organs, such as the heart and skeletal muscle, are capable of metabolizing ketones. Furthermore, ketones appear to be an important fuel source for exercising muscle. Beyond supporting ATP production, it is also becoming widely recognized that ketones are powerful signalling molecules, as they serve as ligands for G-protein coupled receptors and can even modify gene expression via regulating DNA post-translational modifications. As they play a key role in supporting whole-body physiology, it is not surprising that perturbations in ketone metabolism can contribute to various pathologies, particularly in relation to cardiometabolic diseases. Some of the strongest evidence supporting the aforementioned statement is seen for both heart failure and type 2 diabetes. Accordingly, we will review herein the multifaceted roles of ketones in supporting whole-body physiology, while interrogating the evidence to suggest whether modifying ketone metabolism may have a therapeutic role in the management of heart failure and type 2 diabetes.
Collapse
Affiliation(s)
- Seyed Amirhossein Tabatabaei Dakhili
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Kunyan Yang
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Magnus J Stenlund
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
5
|
Schwegel N, Strohhofer C, Kolesnik E, Oltean S, Hüttmair A, Pipp C, Benedikt M, Verheyen N, Gollmer J, Ablasser K, Wallner M, Santner V, Tripolt N, Pferschy P, Zechner P, Alber H, Siller-Matula JM, Kopp K, Zirlik A, Aziz F, Sourij H, von Lewinski D. Impact of empagliflozin on cardiac structure and function assessed by echocardiography after myocardial infarction: a post-hoc sub-analysis of the emmy trial. Clin Res Cardiol 2025; 114:629-639. [PMID: 39297940 PMCID: PMC12058928 DOI: 10.1007/s00392-024-02523-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 08/14/2024] [Indexed: 09/21/2024]
Abstract
BACKGROUND Empagliflozin administered after acute myocardial infarction proofed to improve cardiometabolic parameters and biomarkers, but the impact on cardiac function is still largely unknown. The aim of this post-hoc echocardiographic sub-analysis of the EMMY trial was to provide in-depth echocardiographic analysis on the effects of empagliflozin versus placebo on standard and novel echocardiographic structural and functional parameters after acute myocardial infarction. METHODS In this post-hoc analysis of the EMMY trial a subset of 313 patients (157 empagliflozin vs. 156 placebo) was enrolled for post-processing analysis of echocardiographic structural and functional parameters. On top of two-dimensional and Doppler parameters, myocardial deformation analyses were performed to assess ventricular and atrial strain values. RESULTS Left ventricular volumes showed significant differences in favor of empagliflozin over the course of the trial (change in left ventricular end-diastolic volume median [interquartile range] 8 [-3;19]% versus 13 [0;29]%, p = 0.048; left ventricular end-systolic volume -3 [-15;12]% versus 4 [-12;18]%, p = 0.044). This effect persisted after adjusting for baseline values, age, and sex. Left ventricular systolic and diastolic function overall improved over the course of the trial and parameters for diastolic function showed a distinct trend between groups but did not meet statistical significance in this cohort. CONCLUSION In this post-hoc analysis among patients with acute myocardial infarction, treatment with empagliflozin resulted in a significant beneficial effect on left ventricular end-diastolic and end-systolic volume, without significantly improving left ventricular or right ventricular functional parameters compared to placebo after 26 weeks. CLINICALTRIALS GOV REGISTRATION NCT03087773.
Collapse
Affiliation(s)
- Nora Schwegel
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Christoph Strohhofer
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Ewald Kolesnik
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria.
| | - Sabrina Oltean
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Alexander Hüttmair
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Christian Pipp
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Martin Benedikt
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Nicolas Verheyen
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Johannes Gollmer
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Klemens Ablasser
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Markus Wallner
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Viktoria Santner
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Norbert Tripolt
- Trials Unit for Interdisciplinary Metabolic Medicine, Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Peter Pferschy
- Trials Unit for Interdisciplinary Metabolic Medicine, Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Peter Zechner
- Department of Cardiology and Intensive Care Medicine, Hospital Graz II, West Location, Graz, Austria
| | - Hannes Alber
- Department of Cardiology, Public Hospital Klagenfurt Am Woerthersee, Klagenfurt Am Woerthersee, Austria
| | | | - Kristen Kopp
- Division of Cardiology and Internal Intensive Care Medicine, Department of Internal Medicine II, Paracelsus Medical Private University of Salzburg, Salzburg, Austria
| | - Andreas Zirlik
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Faisal Aziz
- Trials Unit for Interdisciplinary Metabolic Medicine, Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Harald Sourij
- Trials Unit for Interdisciplinary Metabolic Medicine, Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Dirk von Lewinski
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| |
Collapse
|
6
|
Liu X, Zhao H, Liu S, Wen S, Fan W, Xie Q, Cui B, Zhou L, Peng J, Pan H, Zheng Z, Zhang Q. Comparison of the effects of metformin and empagliflozin on cardiac function in heart failure with preserved ejection fraction mice. Front Cardiovasc Med 2025; 12:1533820. [PMID: 40364820 PMCID: PMC12069290 DOI: 10.3389/fcvm.2025.1533820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/02/2025] [Indexed: 05/15/2025] Open
Abstract
Purpose Recent evidence suggests that empagliflozin (EMPA) and metformin (MET) may improve prognosis in heart failure with preserved ejection fraction (HFpEF) patients. This study aims to compare their effects on cardiac structure and function in HFpEF. Methods Male C57BL/6J mice were fed a high-fat diet with L-NAME for 8 weeks to induce HFpEF, followed by 4 weeks of MET or EMPA treatment. Cardiac structure and function were assessed. Network pharmacology and bioinformatics identified key targets, validated by RT-qPCR and WB. Results EMPA-treated mice lost weight, unlike MET-treated ones. MET reduced systolic blood pressure significantly. Both treatments improved glucose tolerance; MET enhanced insulin sensitivity. EMPA increased exercise tolerance by extending exhaustion distance. Both treatments improved diastolic function, reduced heart weight, and attenuated myocardial fibrosis and hypertrophy. Plasma NT-proBNP levels were slightly elevated but not significant. EMPA downregulated HSP90 mRNA and protein expression; both drugs downregulated TGFβ. Conclusion MET and EMPA improve cardiac fibrosis, diastolic function, and pulmonary congestion in HFpEF mice. MET acts by downregulating TGFβ, while EMPA affects collagen metabolism and downregulates HSP90 and TGFβ. These findings offer insights into HFpEF treatment.
Collapse
Affiliation(s)
- Xiehong Liu
- Department of Cardiology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Institute of Emergency Medicine, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Department of Emergency, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Huiqi Zhao
- Department of Cardiology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha, Hunan, China
| | - Sisi Liu
- Department of Cardiology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha, Hunan, China
| | - Siao Wen
- Department of Cardiology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha, Hunan, China
| | - Wenjuan Fan
- Department of Cardiology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha, Hunan, China
| | - Qiong Xie
- Department of Cardiology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha, Hunan, China
| | - Bo Cui
- Department of Cardiology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha, Hunan, China
| | - Lin Zhou
- College of Clinical Laboratory, Changsha Medical University, Changsha, Hunan, China
| | - Jianqiang Peng
- Department of Cardiology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha, Hunan, China
| | - Hongwei Pan
- Department of Cardiology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha, Hunan, China
| | - Zhaofen Zheng
- Department of Cardiology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha, Hunan, China
| | - Qinghai Zhang
- Department of Cardiology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha, Hunan, China
| |
Collapse
|
7
|
Angermann CE, Sehner S, Gerhardt LMS, Santos-Gallego CG, Requena-Ibanez JA, Zeller T, Maack C, Sanz J, Frantz S, Ertl G, Badimon JJ. Anaemia predicts iron homoeostasis dysregulation and modulates the response to empagliflozin in heart failure with reduced ejection fraction: the EMPATROPISM-FE trial. Eur Heart J 2025; 46:1507-1523. [PMID: 39907687 PMCID: PMC12011522 DOI: 10.1093/eurheartj/ehae917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/18/2024] [Accepted: 09/27/2024] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND AND AIMS Sodium-glucose cotransporter 2 inhibitors (SGLT2i) impact iron metabolism in patients with heart failure but mechanisms are incompletely understood. This post hoc analysis explored interrelations between iron homeostasis, cardiac structure/function, exercise capacity, haematopoiesis, and sympathetic activity at baseline, and the effects of 6-month treatment with empagliflozin vs. placebo by anaemia status in EMPATROPISM-FE study participants. METHODS Myocardial iron content (MIC, estimated by cardiac magnetic resonance T2* imaging), left ventricular (LV) volumes and LV ejection fraction (LVEF), exercise capacity, laboratory iron markers (LIM), haemoglobin/haematocrit, erythropoietin, and plasma norepinephrine were determined at baseline and 6 months. RESULTS At baseline, 24/80 participants (30%) had anaemia (haemoglobin < 13/<12 mg/dL in men/women). Patients with vs. without anaemia had higher T2* (indicating lower MIC, P < .001), lower peak oxygen consumption (VO2max, P = .024) and hepcidin (P = .017), and higher erythropoietin (P = .040) and norepinephrine (P = .016). Across subgroups, lower MIC correlated with higher LV volumes (P < .01) and norepinephrine (P < .001), and lower LVEF (P < .01), VO2max (P < .001) and haemoglobin/haematocrit (P < .001). Associations with LIM were poor (all P > .10). Empagliflozin increased MIC (P < .012), improved exercise capacity, and activated haematopoiesis. Changes in LIM and norepinephrine suggested progressive systemic iron depletion and sympatholysis. LV reverse remodelling was greater in individuals with anaemia. CONCLUSIONS Dysregulated cellular iron uptake/availability may be a shared mechanism in myocardial structural/functional impairment, reduced exercise capacity, and restricted haematopoiesis in heart failure, which are worse in patients with anaemia, and improve with empagliflozin. Empagliflozin increases MIC and decreases norepinephrine. Given this inverse association, sympatholysis may help explain the diverse cardiac and systemic benefits from SGLT2i therapy. CLINICAL TRIAL REGISTRATION NCT03485222 (www.clinicaltrials.gov).
Collapse
Affiliation(s)
- Christiane E Angermann
- Comprehensive Heart Failure Center Würzburg, University and University Hospital Würzburg, Am Schwarzenberg 15, 97078 Würzburg, Germany
- Department of Medicine 1, University Hospital Würzburg, Oberdürrbacherstraße 6, 97080 Würzburg, Germany
| | - Susanne Sehner
- Institute of Medical Biometry and Epidemiology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Louisa M S Gerhardt
- Fifth Department of Medicine, University Medical Center Mannheim, Faculty of Medicine Mannheim of the University of Heidelberg, Mannheim, Germany
| | | | | | - Tanja Zeller
- University Center of Cardiovascular Science, University Heart and Vascular Center, University Hospital Hamburg-Eppendorf, and German Center of Cardiovascular Research, Partner Site Hamburg–Kiel–Lübeck, Hamburg, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center Würzburg, University and University Hospital Würzburg, Am Schwarzenberg 15, 97078 Würzburg, Germany
| | - Javier Sanz
- Icahn School of Medicine at Mount Sinai, Cardiovascular Institute, New York, NY, USA
| | - Stefan Frantz
- Comprehensive Heart Failure Center Würzburg, University and University Hospital Würzburg, Am Schwarzenberg 15, 97078 Würzburg, Germany
- Department of Medicine 1, University Hospital Würzburg, Oberdürrbacherstraße 6, 97080 Würzburg, Germany
| | - Georg Ertl
- Comprehensive Heart Failure Center Würzburg, University and University Hospital Würzburg, Am Schwarzenberg 15, 97078 Würzburg, Germany
- Department of Medicine 1, University Hospital Würzburg, Oberdürrbacherstraße 6, 97080 Würzburg, Germany
| | - Juan J Badimon
- Icahn School of Medicine at Mount Sinai, Cardiovascular Institute, New York, NY, USA
| |
Collapse
|
8
|
Lakhal-Littleton S. Anaemia, neurohormonal activation, and myocardial iron depletion in heart failure: can this vicious circle be broken? Eur Heart J 2025; 46:1524-1527. [PMID: 39907696 DOI: 10.1093/eurheartj/ehae798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2025] Open
Affiliation(s)
- Samira Lakhal-Littleton
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| |
Collapse
|
9
|
de Souza MDM, Defante MLR, de Athayde de Hollanda Morais BA, Muniz J, Mendes BX, Martins OC, Prizão VM, Silva MMF. Effects of SGLT2 inhibitors on health-related quality of life and functional capacity in patients with heart failure with and without diabetes: a meta-analysis of randomized controlled trials. Int J Clin Pharm 2025:10.1007/s11096-025-01908-0. [PMID: 40232664 DOI: 10.1007/s11096-025-01908-0] [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: 10/07/2024] [Accepted: 03/21/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND Sodium-glucose cotransporter 2 (SGLT2) inhibitors reduce major cardiovascular events among individuals with heart failure regardless the ejection fraction. The effect of SGLT2 inhibitors on health-related quality of life (HRQoL) and physical capacity are still unclear. AIM To investigate the effects of SGLT2 inhibitors on HRQoL and physical capacity in patients with heart failure. METHOD We systematically searched PubMed, Embase, and Cochrane Central databases for randomized controlled trials (RCTs) comparing SGLT2 inhibitors to placebo in this population. The outcomes analyzed were mean changes in Kansas City Cardiomyopathy Questionnaire (KCCQ) score and its domains: total symptoms score (TSS), physical limitations score (PLS), clinical summary score (CSS) and overall summary score (OSS), as well as the six-minute walk test (6MWT) and peak oxygen uptake (peak VO2). RESULTS Eighteen RCTs with 23,848 participants were included. There was a statistical significant improvement in KCCQ TSS (MD: 3.33; 95% CI 1.84 to 4.81; p < 0.001) in HFrEF, HFpEF and non-diabetic subgroups. Consistent findings were found in KCCQ PLS, KCCQ CSS, and KCCQ OSS. The distance covered in the 6MWT was significant higher (MD: 12.8; 95% CI 1.06 to 24.54; p = 0.03) and peak VO2 was increased (MD: 1.06; 95% CI 0.57 to 1.55; p < 0.001). CONCLUSION SGLT2 inhibitors improve HRQoL and functional capacity in patients with HF regardless the ejection fraction and co-diagnosis of diabetes.
Collapse
Affiliation(s)
- Mariana de Moura de Souza
- Department of Internal Medicine, Federal University of Paraná, R. Gen. Carneiro, 181 - Alto da Glória, Curitiba, PR, 80060-900, Brazil.
| | | | | | - Juliana Muniz
- Department of Internal Medicine, Schmieder Klinik Heidelber, Heidelberg, Germany
| | | | | | | | | |
Collapse
|
10
|
Weng J, Dong W, Liao R, Zheng Y, Fang X, You J, Wang Z, Zuo Y, Chen X, Peng X. High triglyceride-to-high-density lipoprotein cholesterol ratio predicts poor prognosis in new-onset heart failure: a retrospective study. BMC Cardiovasc Disord 2025; 25:251. [PMID: 40175907 PMCID: PMC11963554 DOI: 10.1186/s12872-025-04706-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/25/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND There is limited research on the relationship between the triglyceride-to-high-density lipoprotein cholesterol (TG/HDL-C) ratio and outcomes in new-onset heart failure (HF). Therefore, this study aimed to explore the association between TG/HDL-C ratio and clinical outcomes in these patients. METHODS A retrospective cohort of 614 adults with new-onset HF hospitalized at The First Affiliated Hospital of Nanchang University between July 2021 and December 2022 was analyzed. The primary endpoint was major adverse cardiovascular events (MACE), defined as cardiovascular (CV) death and HF rehospitalizations within 12 months after discharge. Kaplan-Meier (K-M) curves, restricted cubic spline (RCS) analysis, and Cox regression evaluated the association between TG/HDL-C ratio and MACE risk. RESULTS Patients were divided into four quartiles (Quartile 1, 2,3 and 4) based on their TG/HDL-C ratios. The mean age was 68.94 ± 14.34 years, with 59.12% male. The mean left ventricular ejection fraction (LVEF) was 46.59 ± 10.89%, with 45.11% having an LVEF ≤ 40%. During the 12-month follow-up, 156 patients experienced MACE, comprising 18 CV deaths and 138 HF rehospitalizations. The Quartile 4 group had the highest MACE risk incidence compared to other groups (P < 0.001). K-M analysis confirmed that the Quartile 4 group was associated with an increased cumulative incidence of MACE, HF rehospitalization, and CV death (all P < 0.001). RCS analysis revealed a positive nonlinear relationship between the TG/HDL-C ratio and MACE risk (P for nonlinear = 0.026), with a sharp risk increase above a ratio of 1.08. After adjustment, TG/HDL-C ratio was independently associated with MACE (HR: 1.44, 95% CI: 1.29-1.60). Compared to Quartile 1, adjusted HRs were significantly higher in Quartiles 2, 3, and 4 (all P < 0.005). CONCLUSIONS The TG/HDL-C ratio is independently associated with 12-month MACE risk in new-onset HF patients. It may serve as a simple, cost-effective marker to improve early risk stratification and guide closer monitoring and tailored management in this high-risk population.
Collapse
Affiliation(s)
- Junfei Weng
- The 1 st affiliated hospital, Jiangxi Medical College, Nanchang University, No. 461, Bayi Avenue, Nanchang City, 330006, Jiangxi Province, People's Republic of China
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Donghu District, Nanchang City, 330006, Jiangxi Province, People's Republic of China
| | - Wei Dong
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Donghu District, Nanchang City, 330006, Jiangxi Province, People's Republic of China
| | - Ruichun Liao
- The 1 st affiliated hospital, Jiangxi Medical College, Nanchang University, No. 461, Bayi Avenue, Nanchang City, 330006, Jiangxi Province, People's Republic of China
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Donghu District, Nanchang City, 330006, Jiangxi Province, People's Republic of China
| | - Yaofu Zheng
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Donghu District, Nanchang City, 330006, Jiangxi Province, People's Republic of China
| | - Xu Fang
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Donghu District, Nanchang City, 330006, Jiangxi Province, People's Republic of China
| | - Jiaxiang You
- The 1 st affiliated hospital, Jiangxi Medical College, Nanchang University, No. 461, Bayi Avenue, Nanchang City, 330006, Jiangxi Province, People's Republic of China
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Donghu District, Nanchang City, 330006, Jiangxi Province, People's Republic of China
| | - Zhichao Wang
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Donghu District, Nanchang City, 330006, Jiangxi Province, People's Republic of China
| | - Yingbing Zuo
- The 1 st affiliated hospital, Jiangxi Medical College, Nanchang University, No. 461, Bayi Avenue, Nanchang City, 330006, Jiangxi Province, People's Republic of China
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Donghu District, Nanchang City, 330006, Jiangxi Province, People's Republic of China
| | - Xuanying Chen
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Donghu District, Nanchang City, 330006, Jiangxi Province, People's Republic of China
| | - Xiaoping Peng
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Donghu District, Nanchang City, 330006, Jiangxi Province, People's Republic of China.
| |
Collapse
|
11
|
De Masi De Luca G, Palama Z, Longo S, Barba F, Robles AG, Nesti M, Scara A, Coluccia G, Colopi M, De Masi De Luca G, Minardi S, Fusco L, Palmisano P, Accogli M, Sciarra L, Romano S. Effect of Dapagliflozin on Ventricular Arrhythmic Events in Heart Failure Patients With an Implantable Cardioverter Defibrillator. Cardiol Res 2025; 16:140-152. [PMID: 40051671 PMCID: PMC11882230 DOI: 10.14740/cr2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 01/21/2025] [Indexed: 03/09/2025] Open
Abstract
Background The aim of our study was to evaluate the effects of dapagliflozin on the ventricular arrhythmia burden (VAb) in patients with heart failure with reduced ejection fraction (HFrEF) and an implantable cardioverter defibrillator (ICD), correlating the possible reduction in arrhythmic events and ICD therapies with the basal functional capacity, as well as the remodeling parameters induced by treatment. Methods A total of 117 outpatient ICD patients with a known diagnosis of HFrEF who underwent treatment with dapagliflozin were evaluated according to a prospective observational protocol. VAb (including sustained ventricular tachycardia, non-sustained ventricular tachycardia, ventricular fibrillation, and total ventricular events) and specific ICD therapies (anti-tachycardia pacing (ATP) and ICD shocks) were extrapolated from the devices' memory (events per patient per month) by comparing events in the observation period before and after the introduction of dapagliflozin. Results The VAb was significantly reduced after dapagliflozin introduction (2.9 ± 1.8 vs. 4.5 ± 2.0, P = 0.01). The burden of appropriate ATPs was significantly reduced (0.57 ± 0.80 vs. 0.65 ± 0.91, P = 0.03), but not for ICD shocks. In patients with a more advanced functional class, a greater reduction in VAb was observed than in patients with a better initial functional capacity (2.2 ± 0.8 vs. 5.5 ± 1.8, P = 0.001 in the New York Heart Association (NYHA) III/IV group; 3.5 ± 2.1 vs. 4.5 ± 2.2, P = 0.02 in the NYHA I/II group). Considering two independent groups according to reverse remodeling (Δleft ventricular ejection fraction (LVEF) > 15%), a significant reduction in VAb was observed only in those patients who presented significant reverse remodeling (2.5 ± 1.1 vs. 5.1 ± 1.6, P = 0.01). A statistically significant interaction between the variation of total ventricular arrhythmias (VTA) and the basal NYHA class (F(1,115) = 142.25, P < 0.0001, partial η2 = 0.553), as well as between the variation of VTA and the ΔLVEF (F(1,115) = 107.678, P < 0.0001, partial η2 = 0.484) has been demonstrated using a two-way analysis of variance (ANOVA) test. Conclusions In ICD outpatients with HFrEF, dapagliflozin treatment produces a reduction in arrhythmic ventricular events. This improvement is more evident in patients who have a worse functional class and thus a more precarious hemodynamic state, and in patients who present with significant ventricular reverse remodeling. Therefore, we can hypothesize that the hemodynamic and structural improvements induced by treatment represent, at least in the short-medium term, some of the principal elements justifying the significant reduction in VAb.
Collapse
Affiliation(s)
- Gabriele De Masi De Luca
- Department of Life, Health and Environmental Science, University of L’Aquila, L’Aquila, Italy
- Cardiology Unit, Card. “G. Panico” Hospital, Tricase, Italy
- Cardiomed Medical Center, Maglie, Italy
| | - Zefferino Palama
- Department of Life, Health and Environmental Science, University of L’Aquila, L’Aquila, Italy
- Cardiology Unit, “Villa Verde” Hospital, Taranto, Italy
| | | | | | - Antonio Gianluca Robles
- Department of Life, Health and Environmental Science, University of L’Aquila, L’Aquila, Italy
- Cardiology Department, Ospedale “L. Bonomo”, Andria, Italy
| | - Martina Nesti
- Cardiology Unit, CNR Fondazione Toscana “Gabriele Monasterio”, Pisa, Italy
| | - Antonio Scara
- GVM Care and Research, “San Carlo di Nancy” Hospital, Rome, Italy
| | | | - Marzia Colopi
- Cardiology Unit, Card. “G. Panico” Hospital, Tricase, Italy
| | | | - Simona Minardi
- Department of Life, Health and Environmental Science, University of L’Aquila, L’Aquila, Italy
| | - Liuba Fusco
- Department of Life, Health and Environmental Science, University of L’Aquila, L’Aquila, Italy
| | | | | | - Luigi Sciarra
- Department of Life, Health and Environmental Science, University of L’Aquila, L’Aquila, Italy
| | - Silvio Romano
- Department of Life, Health and Environmental Science, University of L’Aquila, L’Aquila, Italy
| |
Collapse
|
12
|
Ghazal R, Wang M, Liu D, Tschumperlin DJ, Pereira NL. Cardiac Fibrosis in the Multi-Omics Era: Implications for Heart Failure. Circ Res 2025; 136:773-802. [PMID: 40146800 PMCID: PMC11949229 DOI: 10.1161/circresaha.124.325402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Cardiac fibrosis, a hallmark of heart failure and various cardiomyopathies, represents a complex pathological process that has long challenged therapeutic intervention. High-throughput omics technologies have begun revolutionizing our understanding of the molecular mechanisms driving cardiac fibrosis and are providing unprecedented insights into its heterogeneity and progression. This review provides a comprehensive analysis of how techniques-encompassing genomics, epigenomics, transcriptomics, proteomics, and metabolomics-are providing insight into our understanding of cardiac fibrosis. Genomic studies have identified novel genetic variants and regulatory networks associated with fibrosis susceptibility and progression, and single-cell transcriptomics has unveiled distinct cardiac fibroblast subpopulations with unique molecular signatures. Epigenomic profiling has revealed dynamic chromatin modifications controlling fibroblast activation states, and proteomic analyses have identified novel biomarkers and potential therapeutic targets. Metabolomic studies have uncovered important alterations in cardiac energetics and substrate utilization during fibrotic remodeling. The integration of these multi-omic data sets has led to the identification of previously unrecognized pathogenic mechanisms and potential therapeutic targets, including cell-type-specific interventions and metabolic modulators. We discuss how these advances are driving the development of precision medicine approaches for cardiac fibrosis while highlighting current challenges and future directions in translating multi-omic insights into effective therapeutic strategies. This review provides a systems-level perspective on cardiac fibrosis that may inform the development of more effective, personalized therapeutic approaches for heart failure and related cardiovascular diseases.
Collapse
Affiliation(s)
- Rachad Ghazal
- Departments of Cardiovascular Diseases (R.G., N.L.P.), Mayo Clinic, Rochester, MN
| | - Min Wang
- Molecular Pharmacology and Experimental Therapeutics (M.W., D.L., N.L.P.), Mayo Clinic, Rochester, MN
| | - Duan Liu
- Molecular Pharmacology and Experimental Therapeutics (M.W., D.L., N.L.P.), Mayo Clinic, Rochester, MN
| | | | - Naveen L. Pereira
- Departments of Cardiovascular Diseases (R.G., N.L.P.), Mayo Clinic, Rochester, MN
- Molecular Pharmacology and Experimental Therapeutics (M.W., D.L., N.L.P.), Mayo Clinic, Rochester, MN
| |
Collapse
|
13
|
Feng Q, Wu M, Mai Z. Emerging horizons: clinical applications and multifaceted benefits of SGLT-2 inhibitors beyond diabetes. Front Cardiovasc Med 2025; 12:1482918. [PMID: 40182430 PMCID: PMC11965600 DOI: 10.3389/fcvm.2025.1482918] [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: 01/19/2025] [Accepted: 03/10/2025] [Indexed: 04/05/2025] Open
Abstract
SGLT-2 inhibitors, initially developed for type 2 diabetes, demonstrate profound cardiorenal and metabolic benefits. This review synthesizes evidence from clinical trials and mechanistic studies to elucidate their roles in cardiovascular diseases, chronic kidney disease, and non-alcoholic fatty liver disease. Key findings include a notable reduction in cardiovascular death/heart failure hospitalization, a marked decrease in heart failure hospitalization risk, and significant improvements in renal and hepatic outcomes. Emerging mechanisms, such as autophagy induction, ketone utilization, and anti-inflammatory effects, underpin these benefits. Ongoing trials explore their potential in non-diabetic populations, positioning SGLT-2 inhibitors as transformative agents in multisystem disease management.
Collapse
Affiliation(s)
- Qing Feng
- Department of Cardiology, Kaiping Central Hospital, Kaiping, China
| | - Miaoqiong Wu
- Department of Endocrinology, Kaiping Central Hospital, Kaiping, China
| | - Zizhao Mai
- School of Stomatology, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
14
|
Fan Q, Wang Y, An Q, Ling Y. Right ventricular dysfunction following surgical repair of tetralogy of Fallot: Molecular pathways and therapeutic prospects. Biomed Pharmacother 2025; 184:117924. [PMID: 39983432 DOI: 10.1016/j.biopha.2025.117924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/11/2025] [Accepted: 02/15/2025] [Indexed: 02/23/2025] Open
Abstract
Tetralogy of Fallot (TOF) is the most common cyanotic congenital heart disease (CHD). Although surgical correction of TOF is possible, patients often face challenges related to right ventricle dysfunction (RVD) post-surgery, which can significantly impact their long-term survival. The causes of RVD in TOF patients are complex, involving both the unique structural characteristics of the TOF heart and damage resulting from surgical interventions. Residual anatomical issues following TOF repair are often unavoidable, placing the RV under stress and leading to the activation of multiple molecular pathways. This review comprehensively outlines the causes of RVD in patients after TOF surgery, particularly focusing the molecular pathways that contribute to RVD, including established signaling pathways as well as emerging pathways identified through transcriptomic analysis of RV myocardium in TOF patients. We also highlight the features of these molecular pathways concerning RVD, as well as the influence of gender disparities on these molecular pathways. By interpreting the causes and molecular mechanisms underlying RVD after TOF surgery, this review provides new insights for managing RVD in repaired TOF, potentially paving the way for targeted therapies aimed at improving long-term outcomes for those affected by RVD.
Collapse
Affiliation(s)
- Qiang Fan
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| | - Yabo Wang
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| | - Qi An
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| | - Yunfei Ling
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| |
Collapse
|
15
|
Ramos-Roman MA. Comparison Between SGLT2 Inhibitors and Lactation: Implications for Cardiometabolic Health in Parous Women. Metab Syndr Relat Disord 2025; 23:77-85. [PMID: 39431925 PMCID: PMC12021787 DOI: 10.1089/met.2024.0182] [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: 10/22/2024] Open
Abstract
Sodium-glucose cotransporter-2 (SGLT2) inhibition and lactation result in the excretion of large amounts of glucose in urine or milk and are associated with a lower risk of cardiovascular events. The respective mechanisms behind this association with cardiovascular protection are not clear. This review compares the contribution of noninsulin-mediated glucose transport during pharmacologic inhibition of SGLT2 with noninsulin-mediated glucose transport during lactation in terms of the implications for the cardiometabolic health of parous women. The search topics used to obtain information on SGLT2 inhibitors included mechanisms of action, atherosclerosis, and heart failure. The search topics used to obtain information on lactation included cardiovascular health and milk composition. Subsequent reference searches of retrieved articles were also used. Active treatment with SGLT2 inhibitors affects glucose and sodium transport in the kidneys and predominantly protects against hospitalization for heart failure soon after the onset of therapy. Active lactation stimulates glucose transport into the mammary gland and improves subclinical and clinical atherosclerotic vascular disease years after delivery. Both SGLT2 inhibitors and lactation have effects on a variety of glucose transporters. Several mechanisms have been proposed to explain the cardiometabolic benefits of SGLT2 inhibition and lactation. Learning from the similarities and differences between both processes will advance our understanding of cardiometabolic health for all people.
Collapse
Affiliation(s)
- Maria A. Ramos-Roman
- Department of Internal Medicine, Division of Endocrinology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
16
|
Tan W, Wang Y, Cheng S, Liu Z, Xie M, Song L, Qiu Q, Wang X, Li Z, Liu T, Guo F, Wang J, Zhou X. AdipoRon ameliorates the progression of heart failure with preserved ejection fraction via mitigating lipid accumulation and fibrosis. J Adv Res 2025; 68:299-315. [PMID: 38382593 PMCID: PMC11785573 DOI: 10.1016/j.jare.2024.02.015] [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: 12/12/2023] [Revised: 02/17/2024] [Accepted: 02/18/2024] [Indexed: 02/23/2024] Open
Abstract
INTRODUCTION Obesity and imbalance in lipid homeostasis contribute greatly to heart failure with preserved ejection fraction (HFpEF), the dominant form of heart failure. Few effective therapies exist to control metabolic alterations and lipid homeostasis. OBJECTIVES We aimed to investigate the cardioprotective roles of AdipoRon, the adiponectin receptor agonist, in regulating lipid accumulation in the two-hit HFpEF model. METHODS HFpEF mouse model was induced using 60 % high-fat diet plus L-NAME drinking water. Then, AdipoRon (50 mg/kg) or vehicle were administered by gavage to the two-hit HFpEF mouse model once daily for 4 weeks. Cardiac function was evaluated using echocardiography, and Postmortem analysis included RNA-sequencing, untargeted metabolomics, transmission electron microscopy and molecular biology methods. RESULTS Our study presents the pioneering evidence that AdipoR was downregulated and impaired fatty acid oxidation in the myocardia of HFpEF mice, which was associated with lipid metabolism as indicated by untargeted metabolomics. AdipoRon, orally active synthetic adiponectin receptor agonist, could upregulate AdipoR1/2 (independently of adiponectin) and reduce lipid droplet accumulation, and alleviate fibrosis to restore HFpEF phenotypes. Finally, AdipoRon primarily exerted its effects through restoring the balance of myocardial fatty acid intake, transport, and oxidation via the downstream AMPKα or PPARα signaling pathways. The protective effects of AdipoRon in HFpEF mice were reversed by compound C and GW6471, inhibitors of AMPKα and PPARα, respectively. CONCLUSIONS AdipoRon ameliorated the HFpEF phenotype by promoting myocardial fatty acid oxidation, decreasing fatty acid transport, and inhibiting fibrosis via the upregulation of AdipoR and the activation of AdipoR1/AMPKα and AdipoR2/PPARα-related downstream pathways. These findings underscore the therapeutic potential of AdipoRon in HFpEF. Importantly, all these parameters get restored in the context of continued mechanical and metabolic stressors associated with HFpEF.
Collapse
Affiliation(s)
- Wuping Tan
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Yijun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Siyi Cheng
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Zhihao Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Mengjie Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Lingpeng Song
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Qinfang Qiu
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Xiaofei Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Zeyan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Tianyuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Fuding Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China.
| | - Jun Wang
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China.
| | - Xiaoya Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China.
| |
Collapse
|
17
|
Hopkins S, Baqai F, Gajagowni S, Hickey G. Direct Cardiac Mechanisms of the Sodium Glucose Co-Transporter 2 Inhibitor Class. J Cardiovasc Pharmacol Ther 2025; 30:10742484251323428. [PMID: 40221961 DOI: 10.1177/10742484251323428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
BackgroundSodium-glucose co-transporter 2 (SGLT2) inhibitors have demonstrated significant cardiovascular benefits in clinical trial. While their role in reducing heart failure hospitalizations and cardiovascular mortality is well established, the precise mechanisms underlying their direct cardiac effects remain unclear. This literature review aims to synthesize current knowledge on the molecular and physiological pathways by which SGLT2 inhibitors may exert effects on cardiac tissue, independent of glycemic control.MethodsA comprehensive review of peer-reviewed articles, randomized controlled trials, meta-analyses, and mechanistic studies published in PubMed and related databases was conducted. The search focused on studies examining the direct impact of SGLT2 inhibitors on cardiac function, remodeling, metabolism, and intracellular signaling pathways. Only studies evaluating the cardiac effects separate from their glucose-lowering action were included in the analysis.ResultsThis review identified several key mechanisms by which SGLT2 inhibitors may benefit the heart directly, including reductions in oxidative stress, inflammation, and myocardial fibrosis. Emerging evidence suggests that these drugs modulate key pathways such as sodium-hydrogen exchange (NHE) inhibition, improvement of mitochondrial function, and promotion of ketone body utilization in cardiomyocytes.ConclusionsSGLT2 inhibitors appear to confer direct cardioprotective effects. These include anti-inflammatory, anti-fibrotic, and energy efficiency improvements in the myocardium. The findings highlight new potential therapeutic mechanisms and provide a foundation for further research into the non-diabetic use of SGLT2 inhibitors in heart failure and other cardiac conditions. Understanding these direct effects could lead to optimized treatment strategies for patients with and without diabetes.
Collapse
Affiliation(s)
| | - Faiz Baqai
- Department of Internal Medicine, Baylor College of Medicine, Houston, USA
| | | | - Gavin Hickey
- Heart and Vascular Institute, UPMC, Pittsburgh, USA
| |
Collapse
|
18
|
Elliott J, Oyama MA. Sodium glucose transporter 2 inhibitors: Will these drugs benefit non-diabetic veterinary patients with cardiac and kidney diseases? J Vet Pharmacol Ther 2025; 48 Suppl 1:1-18. [PMID: 39001645 PMCID: PMC11737021 DOI: 10.1111/jvp.13472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 06/14/2024] [Accepted: 07/03/2024] [Indexed: 01/18/2025]
Abstract
Sodium glucose transporter type 2 (SGLT2) inhibitors have been introduced into human medicine where their beneficial effects go beyond the expected improvement in blood glucose control. These drugs appear to prevent progression of both cardiovascular and kidney diseases, not only in diabetic but also in non-diabetic human patients. As these drugs have received conditional approval for use in diabetic cats and are being used in other veterinary species, the intriguing question as to whether they will have similar cardioprotective and nephroprotective effects in dogs and cats is being asked. The primary mechanism(s) by which SGLT2 inhibitors are cardio- and nephroprotective remain to be fully characterized. This paper reviews these suggested mechanisms in the context of the pathophysiology of progressive cardiovascular and kidney diseases in dogs and cats with the goal of predicting which categories of non-diabetic veterinary patients these drugs might be of most benefit.
Collapse
Affiliation(s)
- Jonathan Elliott
- Department of Comparative Biomedical SciencesRoyal Veterinary College, University of LondonLondonUK
| | - Mark A. Oyama
- Department of Clinical Sciences & Advanced MedicineUniversity of Pennsylvania School of Veterinary MedicinePhiladelphiaPennsylvaniaUSA
| |
Collapse
|
19
|
Lazareva L, Shubrook JH, Dhond M. Comprehensive review of the heart failure management guidelines presented by the American College of Cardiology and the current supporting evidence. J Osteopath Med 2025; 125:1-8. [PMID: 39054773 DOI: 10.1515/jom-2024-0071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 06/19/2024] [Indexed: 07/27/2024]
Abstract
CONTEXT Heart failure (HF) is a chronic condition that affects the heart's functional capacity, resulting in symptoms such as fatigue, edema, and dyspnea. It affects millions of adults in the United States and presents challenges in optimizing treatment and coordinating care among clinicians. Additionally, the various classifications for HF and limited research on treatment outcomes in heart failure with midrange ejection fraction (HFmrEF) and heart failure with preserved ejection fraction (HFpEF) further complicate the pharmacological management of patients with this disease. OBJECTIVES The objectives of this article are to review the pharmacotherapy guidelines for HF provided by the American College of Cardiology (ACC) and offer an update on the current trials conducted on these agents. METHODS The paper includes a post hoc analysis of established randomized controlled trials (RCTs), current RCTs, analysis of HF registries, and the guidelines published by the ACC. The gathering of research began in June 2023 and completed in August 2023. PubMed was utilized with the following search items: "treatment for HFrEF" (heart failure with reduced ejection fraction), "treatment for HFmrEF," and "treatment for HFpEF." The screening process was completed by one author. The automation tools utilized were "clinical trials," "randomized control trials," and "five years". Meta-analyses, systematic reviews, and case reports were excluded from the screening process. This review does not include research regarding medical devices, interventional therapies, and lifestyle modifications. Finally, research regarding additional comorbidities, nonpharmacological focused research, and agents not recommended by the ACC are not included in this paper. RESULTS The search began with 6,561 records identified from PubMed, with 407 records screened after automation tools were utilized to filter for "clinical trials," "randomized control trials," "one year," and "five years". A total of 22 duplicates were reviewed, 318 were sought for screening after trials from 2019 were removed, and 31 studies were ultimately included in the review. A detailed summary of the most recent recommendations by the ACC are provided. The discussion includes indications, mechanisms of action, side effects, and contraindications for the selected agents. Additionally, recent clinical trials are included to provide evidence on the efficacy of the recommended classes of drugs. CONCLUSIONS The current guidelines for managing HFrEF have been consistent, but there is limited consensus on treating HFmrEF and HFpEF. Large RCTs have provided compelling evidence supporting the use of the recommended pharmacological agents. However, despite the new effective treatment protocols, slow clinical inertia and underoptimization of HF management persist. Thus, it is crucial to synchronize care among clinicians involved in managing patients with this disease.
Collapse
Affiliation(s)
- Lia Lazareva
- 59431 College of Osteopathic Medicine, Touro University California , Vallejo, CA, USA
| | - Jay H Shubrook
- 59431 College of Osteopathic Medicine, Touro University California , Vallejo, CA, USA
| | - Milind Dhond
- Medical Director, Cardiovascular Medicine, North Bay Medical Center, Fairfield, CA, USA
| |
Collapse
|
20
|
Lau EW, Bonnemeier H, Baldauf B. Left bundle branch block-Innocent bystander, silent menace, or both. Heart Rhythm 2024:S1547-5271(24)03711-1. [PMID: 39742988 DOI: 10.1016/j.hrthm.2024.12.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/06/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025]
Abstract
Left bundle branch block (LBBB) causes immediate electrical and mechanical dyssynchrony of the left ventricle (LV) and gradual structural damages in the Purkinje cells and myocardium. Mechanical dyssynchrony reduces the LV ejection fraction (EF) instantly, but only to ≈55% in an otherwise normal heart. Because of the heart's in-built functional redundancy, a patient with LBBB does not always notice the heart's reduced efficiency straightaway. After a variable period of time (which could be from days to decades), the patient may become symptomatic with heart failure (HF), which classifies as HF with preserved EF ≥50% (HFpEF). The LVEF drops further because of continuous adverse remodeling and inefficient cardiac contraction. The patient transits to HF with moderately reduced EF 35%-50% (HFmrEF) and then reduced EF ≤35% (HFrEF) over 5-21 years. Cardiac resynchronization therapy (CRT) is currently only indicated in guidelines for HFrEF and LBBB. LBBB shortens the median survival of patients with HFmrEF by 5.5 years. Randomized controlled trials have shown that CRT improves echocardiographic indices for HFmrEF with LBBB. CRT in HFpEF with LBBB is a promising but underexplored/underused therapy. There have been anecdotal reports that CRT produced symptom relief in patients debilitated by HFpEF with LBBB, who constitute ≈6% of all patients with HF and an adequate pool of potential randomized controlled trial participants. Conduction system pacing in the form of left bundle branch area pacing is an emerging pacing strategy that might reverse and forestall the deleterious effects of LBBB.
Collapse
Affiliation(s)
- Ernest W Lau
- Department of Cardiology, Royal Victoria Hospital, Belfast, United Kingdom
| | - Hendrik Bonnemeier
- Department of Cardiology, University Rostock, Rostock, Germany; Medical Faculty, Christian-Albrechts-University, Kiel, Germany; Division of Life Sciences, University of Applied Sciences, Bremerhaven, Germany
| | - Benito Baldauf
- Medical Faculty, Christian-Albrechts-University, Kiel, Germany; Division of Life Sciences, University of Applied Sciences, Bremerhaven, Germany.
| |
Collapse
|
21
|
Ng YH, Koay YC, Marques FZ, Kaye DM, O’Sullivan JF. Leveraging metabolism for better outcomes in heart failure. Cardiovasc Res 2024; 120:1835-1850. [PMID: 39351766 PMCID: PMC11630082 DOI: 10.1093/cvr/cvae216] [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: 03/09/2024] [Revised: 06/26/2024] [Accepted: 08/07/2024] [Indexed: 12/11/2024] Open
Abstract
Whilst metabolic inflexibility and substrate constraint have been observed in heart failure for many years, their exact causal role remains controversial. In parallel, many of our fundamental assumptions about cardiac fuel use are now being challenged like never before. For example, the emergence of sodium-glucose cotransporter 2 inhibitor therapy as one of the four 'pillars' of heart failure therapy is causing a revisit of metabolism as a key mechanism and therapeutic target in heart failure. Improvements in the field of cardiac metabolomics will lead to a far more granular understanding of the mechanisms underpinning normal and abnormal human cardiac fuel use, an appreciation of drug action, and novel therapeutic strategies. Technological advances and expanding biorepositories offer exciting opportunities to elucidate the novel aspects of these metabolic mechanisms. Methodologic advances include comprehensive and accurate substrate quantitation such as metabolomics and stable-isotope fluxomics, improved access to arterio-venous blood samples across the heart to determine fuel consumption and energy conversion, high quality cardiac tissue biopsies, biochemical analytics, and informatics. Pairing these technologies with recent discoveries in epigenetic regulation, mitochondrial dynamics, and organ-microbiome metabolic crosstalk will garner critical mechanistic insights in heart failure. In this state-of-the-art review, we focus on new metabolic insights, with an eye on emerging metabolic strategies for heart failure. Our synthesis of the field will be valuable for a diverse audience with an interest in cardiac metabolism.
Collapse
Affiliation(s)
- Yann Huey Ng
- Cardiometabolic Medicine, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Room 3E71 D17, Camperdown, NSW 2006, Australia
- Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Office 3E72, Camperdown, NSW 2006, Australia
| | - Yen Chin Koay
- Cardiometabolic Medicine, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Room 3E71 D17, Camperdown, NSW 2006, Australia
- Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Office 3E72, Camperdown, NSW 2006, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, VIC 3800, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC 3800, Australia
- Victorian Heart Institute, Monash University, Melbourne, VIC 3800, Australia
| | - David M Kaye
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC 3800, Australia
- Department of Cardiology, Alfred Hospital, Melbourne, VIC 3004, Australia
- Monash-Alfred-Baker Centre for Cardiovascular Research, Monash University, Melbourne, VIC 3800, Australia
| | - John F O’Sullivan
- Cardiometabolic Medicine, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Room 3E71 D17, Camperdown, NSW 2006, Australia
- Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Office 3E72, Camperdown, NSW 2006, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
- Department of Medicine, Technische Univeristat Dresden, 01062 Dresden, Germany
| |
Collapse
|
22
|
Gupta R, Umeh C, Mohta T, Vaidya A, Wolfson A, Nattiv J, Bhatia H, Kaur G, Dhawan R, Darji P, Eghreriniovo B, Sanwo E, Hotwani P, Mahdavian P, Kumar S, Tiwari B. Representation of women and racial minorities in SGLT2 inhibitors and heart failure clinical trials. IJC HEART & VASCULATURE 2024; 55:101539. [PMID: 39502337 PMCID: PMC11535350 DOI: 10.1016/j.ijcha.2024.101539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/12/2024] [Accepted: 10/14/2024] [Indexed: 11/08/2024]
Abstract
Background Inadequate representation of women and racial minorities in heart failure (HF) clinical trials continues to limit the generalizability of the results. This could create a disparity in treatment for future heart failure therapies and devices. The study aims to assess the representation of women and racial minorities in recent heart failure studies involving sodium-glucose cotransporter-2 (SGLT-2) inhibitors. Methods PubMed was used to search randomized controlled trials (RCTs) looking at SGLT-2 inhibitors and heart failure, which were published from inception to August 2024. Results A total of 43 RCTs with 27,703 participants were identified. The studies were published between 2018 and 2024. Seven studies (41 %) were multi-country, with 45 countries represented. The overall proportion of women enrolled in the studies was 35.6 %. The proportion of women was 24.06 % in studies that recruited only patients with HFrEF, 44.33 % in those that recruited only patients with HFpEF, and 41.4 % in those that recruited both HFrEF and HFpEF. Data on race was partially reported in 25 studies (58 %). 76 % of the pharmaceutical industry-funded studies reported race data. However, only 33.3 % of the unfunded or non-industry-funded studies reported race data. In the studies that reported race data, 72.91 % were Caucasians, 15.48 % were Asians, 5.62 % were African-American and 4.1 % were mixed race or others.In the bivariate analysis, race was more likely to be reported in studies done in the US (p < 0.001), multi-country studies (p = 0.013), and studies sponsored by pharmaceutical companies. More than a third of the study participants were more likely to be women in more recently published studies than older studies (p < 0.001). Additionally, more than a third of the study participants were more likely to be women in studies done in the US (p = 0.055). The multivariate analysis showed an increased odds of having more than a third of the study participants being women in more recently published studies (OR 1.83, 95 % CI 1.06-3.17, p = 0.031) and in studies done in the US (OR 7.69, 95 % CI 1.53-38.59, p = 0.013). Conclusion Our study found that women and racial minority individuals have remained underrepresented in recent heart failure studies. Although some progress has been made over the years, more work is needed to improve data reporting and address barriers to enrollment for women and racial minority individuals in clinical trials.
Collapse
Affiliation(s)
| | | | | | - Ajay Vaidya
- University of Southern California, Los Angeles, CA 90033, USA
| | - Aaron Wolfson
- University of Southern California, Los Angeles, CA 90033, USA
| | - Jonathan Nattiv
- University of Southern California, Los Angeles, CA 90033, USA
| | | | - Gagan Kaur
- Hemet Global Medical Center, CA 92543, USA
| | | | - Puja Darji
- Hemet Global Medical Center, CA 92543, USA
| | | | | | | | | | - Sabina Kumar
- McLaren Macomb Hospital/Michigan State University, Mt. Clements, MI 48043, USA
| | | |
Collapse
|
23
|
Yuan X, Pan L, Zhang C, Zhu Q, Huang Z, Qin Y, Zhang G, Feng Z, Yang C, Hou N. Empagliflozin improves pressure-overload-induced cardiac hypertrophy by inhibiting the canonical Wnt/β-catenin signaling pathway. Front Pharmacol 2024; 15:1499542. [PMID: 39664517 PMCID: PMC11631586 DOI: 10.3389/fphar.2024.1499542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 10/30/2024] [Indexed: 12/13/2024] Open
Abstract
Background Empagliflozin (EMPA) is an SGLT-2 inhibitor that can control hyperglycemia. Clinical trials have indicated its cardio-protective effects against cardiac remodeling in diabetes or non-diabetes patients. However, the underlying molecular mechanisms of EMPA's cardio-protective effects remain elusive. Methods We evaluated whether the EMPA attenuated the pressure-overload-induced cardiac hypertrophy by inhibiting the Wnt/β-catenin pathway. Furthermore, the effects of the EMPA on a mouse model of transverse aortic constriction (TAC) induced cardiac hypertrophy was also evaluated. Mice were administrated with 0.5% CMC-Na as a vehicle or EMPA (10 mg/kg/day, daily, throughout the study) by intragastric gavage. Results The in vivo echocardiography and histologic morphological analyses revealed that EMPA attenuated TAC-induced cardiac hypertrophy. Moreover, it also ameliorated TAC-induced cardiac fibrosis and decreased the cell size of the cardiomyocytes in isolated adult cardiomyocytes. Molecular mechanism analysis revealed that the EMPA reduced the TAC-induced enhanced expression of the Wnt/β-catenin pathway in vivo. For in vitro assessments, isolated neonatal rat cardiomyocytes (NRCMs) were treated with Angiotensin II (AngII) and EMPA; the results showed that in the absence of EMPA, the expression of the Wnt/β-catenin pathway was enhanced. In the trans-genetic heterozygous β-catenin deletion mice, EMPA attenuated TAC-induced cardiac remodeling by reducing the Wnt/β-catenin pathway. In addition, molecular docking analysis indicated that EMPA interacts with FZD4 to inhibit the TAC and AngII induced Wnt/β-catenin pathway in cardiomyocytes. Conclusion Our study illustrated that EMPA might directly interact with FZD4 to inhibit the TAC and AngII-induced activation of the Wnt/β-catenin pathway to attenuate the adverse cardiac remodeling.
Collapse
Affiliation(s)
- Xun Yuan
- The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, China
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Li Pan
- Department of Physiology, School of Basic Medicine Sciences, Guangzhou Health Science College, Guangzhou, China
| | - Chi Zhang
- School of Stomatology, Guangzhou Medical University, Guangzhou, China
| | - Qiulian Zhu
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Zexin Huang
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Yuan Qin
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Guiping Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Zhimei Feng
- The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, China
| | - Caixian Yang
- The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, China
| | - Ning Hou
- The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, China
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
24
|
Fu K, Yang Z, Wang N, Dong Y, Wang Z, Chen W, Lu H. Recurrence of left ventricular systolic dysfunction and its risk factors in heart failure with improved ejection fraction patients receiving guideline-directed medical therapy: A trajectory analysis based on echocardiography. Int J Cardiol 2024; 415:132370. [PMID: 39029560 DOI: 10.1016/j.ijcard.2024.132370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/10/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND Despite the better prognosis of heart failure (HF) with improved ejection fraction (HFimpEF), remnant cardiovascular risks, including cardiovascular death, rehospitalization, and future deterioration of left ventricular (LV) systolic function, remain in HFimpEF. However, for HFimpEF patients, especially for those receiving guideline-directed medical therapy (GDMT), the recurrent LV systolic dysfunction and its risk factors is still unclear. METHODS A total of 1098 HF patients under HF follow-up management system were initially screened. Echocardiography was re-evaluated at 3-, 6-, and 12-month follow-up. After exclusion, a total of 203 HFimpEF patients on GDMT were enrolled in our final analysis. Cox regression analysis was conducted to select risk factors. RESULTS During the 1-year follow-up, a total of 28 (13.8%) patients had recurrent LV systolic dysfunction. The trajectory analysis of echocardiographic parameters illustrated that persistent decline of left ventricular ejection fraction (LVEF) and worsening LV remodeling was observed in patients with recurrent LV systolic dysfunction. Multivariable Cox regression analysis identified that ischemic cardiomyopathy, atrial fibrillation, higher left ventricular end-diastolic diameter index (LVEDDI), elevated serum potassium, and a lack of sodium-glucose co-transporter-2 inhibitors (SGLT2i) treatment were confirmed as independent risk factors for recurrent LV systolic dysfunction. Recurrent LV systolic dysfunction was associated with higher rehospitalization rate. CONCLUSION In our longitudinal cohort study, almost 14% HFimpEF receiving GDMT suffered recurrent LV systolic dysfunction. Ischemic cardiomyopathy, atrial fibrillation, higher LVEDDI, higher serum potassium, and a lack of SGLT2i therapy were tightly associated with recurrence of LV systolic dysfunction. Relapse of LV systolic dysfunction correlated with poor prognosis.
Collapse
Affiliation(s)
- Kang 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
| | - Zhuohao Yang
- 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
| | - Ning Wang
- 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
| | - Youran Dong
- 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
| | - Zhiyuan Wang
- 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
| | - Wenqiang Chen
- 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.
| | - Huixia Lu
- 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
|
25
|
Li W, Shen X, Zhang M, Tan W, Jiang X, Wen H, Shen Y. Meta-analysis of the efficacy and impact on cardiac function of sodium-glucose cotransporter 2 inhibitor Empagliflozin in heart failure patients. Medicine (Baltimore) 2024; 103:e40409. [PMID: 39533603 PMCID: PMC11557010 DOI: 10.1097/md.0000000000040409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Currently, there is no comprehensive systematic review available to comprehensively assess the efficacy and safety of Empagliflozin and other sodium-glucose cotransporter 2 inhibitors in the treatment of heart failure (HF). This study employed a meta-analysis approach to systematically evaluate the therapeutic effects of Empagliflozin in HF patients and its impact on cardiac function. METHOD The keywords including "heart failure," "HF," "cardiac failure," "cardiac disease," "Empagliflozin," and "sodium-glucose cotransporter 2 inhibitors" were utilized to search for relevant clinical studies on Empagliflozin in the treatment of HF in various databases, such as China National Knowledge Infrastructure, Wanfang, VIP Chinese Medical Journal Database, PubMed, MEDLINE, Embase, Cochrane Library, Springer, and Science Direct. The studies included patients with HF who received drug treatment. Data on baseline characteristics and posttreatment outcomes, including HF hospitalization (HHF), cardiovascular mortality, all-cause mortality, estimated glomerular filtration rate changes, Kansas City Cardiomyopathy Questionnaire quality of life (QoL) scores, N-terminal pro-B-type natriuretic peptide, left ventricular ejection fraction, hematocrit, and other relevant indicators were collected. Meta-analysis was conducted using RevMan5.3 to analyze the extracted data. RESULTS A total of 15 studies were included in the final analysis, comprising 36,917 patients with HF. Among them, 18,486 patients were in Empagliflozin group, and 18,431 patients were in control (Ctrl) group. The results of the meta-analysis demonstrated that, relative to Ctrl group, Empagliflozin group showed a substantially lower HHF rate, a substantial improvement in estimated glomerular filtration rate changes, a reduced cardiovascular mortality rate, a higher Kansas City Cardiomyopathy Questionnaire QoL score, increased hematocrit values, reduced N-terminal pro-B-type natriuretic peptide changes, and enhanced left ventricular ejection fraction changes. These findings suggest that remarkable improvements in various outcomes compared to the Ctrl group. CONCLUSION The sodium-glucose cotransporter 2 inhibitor Empagliflozin markedly reduces the HHF rate and cardiovascular mortality in HF patients. It also improves patients' QoL, enhances renal function, and increases cardiac function while reducing both, the preload and afterload.
Collapse
Affiliation(s)
- Weidong Li
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xuanyang Shen
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Meiqi Zhang
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Wentao Tan
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiaolu Jiang
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Hongfu Wen
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yuan Shen
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
26
|
Yerra VG, Connelly KA. Extrarenal Benefits of SGLT2 Inhibitors in the Treatment of Cardiomyopathies. Physiology (Bethesda) 2024; 39:0. [PMID: 38888433 DOI: 10.1152/physiol.00008.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/05/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors have emerged as pivotal medications for heart failure, demonstrating remarkable cardiovascular benefits extending beyond their glucose-lowering effects. The unexpected cardiovascular advantages have intrigued and prompted the scientific community to delve into the mechanistic underpinnings of these novel actions. Preclinical studies have generated many mechanistic theories, ranging from their renal and extrarenal effects to potential direct actions on cardiac muscle cells, to elucidate the mechanisms linking these drugs to clinical cardiovascular outcomes. Despite the strengths and limitations of each theory, many await validation in human studies. Furthermore, whether SGLT2 inhibitors confer therapeutic benefits in specific subsets of cardiomyopathies akin to their efficacy in other heart failure populations remains unclear. By examining the shared pathological features between heart failure resulting from vascular diseases and other causes of cardiomyopathy, certain specific molecular actions of SGLT2 inhibitors (particularly those targeting cardiomyocytes) would support the concept that these medications will yield therapeutic benefits across a broad range of cardiomyopathies. This article aims to discuss the important mechanisms of SGLT2 inhibitors and their implications in hypertrophic and dilated cardiomyopathies. Furthermore, we offer insights into future research directions for SGLT2 inhibitor studies, which hold the potential to further elucidate the proposed biological mechanisms in greater detail.
Collapse
Affiliation(s)
- Veera Ganesh Yerra
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada
| | - Kim A Connelly
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada
| |
Collapse
|
27
|
Fibbi G, Sato R, Vatic M, Genreith FP, von Haehling S. Pharmacological management of heart failure: a patient-centred approach. Expert Opin Pharmacother 2024; 25:2151-2165. [PMID: 39434709 DOI: 10.1080/14656566.2024.2418414] [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: 06/29/2024] [Revised: 10/02/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024]
Abstract
INTRODUCTION Heart failure (HF) is a global health challenge that requires a multidisciplinary approach. Despite recent advances in pharmacological and interventional therapy, morbidity and mortality in these patients remain high. For this reason, and because of its interplay with other cardiovascular and non-cardiovascular diseases, HF represents a major area of research, with new trials being published every year and international guidelines constantly updated. AREAS COVERED The authors review the current status and possible future developments in HF pharmacotherapy. EXPERT OPINION The treatment of HF has made significant advances in recent years, and the current recommendations are based on large outcome trials. This has led to significant reductions in both mortality and morbidity, but the death rate remains unacceptably high. In this context, a patient-centered approach that considers comorbidities and specific clinical scenarios when dosing HF medication is essential. Prevention of hospital admissions for cardiac decompensation is of utmost importance in patients with HF as is the enablement of activities of daily living, an endpoint which has only recently been incorporated into major HF trials.
Collapse
Affiliation(s)
- Guglielmo Fibbi
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
- Department of Geriatrics, University Medical Center Göttingen, Göttingen, Germany
| | - Ryosuke Sato
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Mirela Vatic
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Frederik Pascal Genreith
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| |
Collapse
|
28
|
Selvaraj S, Patel S, Sauer AJ, McGarrah RW, Jones P, Kwee LC, Windsor SL, Ilkayeva O, Muehlbauer MJ, Newgard CB, Borlaug BA, Kitzman DW, Shah SJ, Margulies KB, Husain M, Inzucchi SE, McGuire DK, Lanfear DE, Javaheri A, Umpierrez G, Mentz RJ, Sharma K, Kosiborod MN, Shah SH. Metabolic Effects of the SGLT2 Inhibitor Dapagliflozin in Heart Failure Across the Spectrum of Ejection Fraction. Circ Heart Fail 2024; 17:e011980. [PMID: 39421941 PMCID: PMC11634023 DOI: 10.1161/circheartfailure.124.011980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/27/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Mechanisms of benefit with SGLT2is (sodium-glucose cotransporter-2 inhibitors) in heart failure (HF) remain incompletely characterized. Dapagliflozin alters ketone and fatty acid metabolism in HF with reduced ejection fraction though similar effects have not been observed in HF with preserved ejection fraction. We explore whether metabolic effects of SGLT2is vary across the left ventricular ejection fraction spectrum and their relationship with cardiometabolic end points in 2 randomized trials of dapagliflozin in HF. METHODS Metabolomic profiling of 61 metabolites was performed in 527 participants from DEFINE-HF (Dapagliflozin Effects on Biomarkers, Symptoms and Functional Status in Patients With HF With Reduced Ejection Fraction) and PRESERVED-HF (Dapagliflozin in PRESERVED Ejection Fraction HF; 12-week, placebo-controlled trials of dapagliflozin in HF with reduced ejection fraction and HF with preserved ejection fraction, respectively). Linear regression was used to assess changes in principal components analysis-defined metabolite factors with treatment from baseline to 12 weeks, as well as the relationship between changes in metabolite clusters and HF-related end points. RESULTS The mean age was 66±11 years, 43% were female, and 33% were self-identified as Black. Two principal components analysis-derived metabolite factors (which were comprised of ketone and short-/medium-chain acylcarnitines) increased with dapagliflozin compared with placebo. Ketosis (defined as 3-hydroxybutyrate >500 μM) was achieved in 4.5% with dapagliflozin versus 1.2% with placebo (P=0.03). There were no appreciable treatment effects on amino acids, including branched-chain amino acids. Increases in several acylcarnitines were consistent across LVEF (Pinteraction>0.10), whereas the ketogenic effect diminished at higher LVEF (Pinteraction=0.01 for 3-hydroxybutyrate). Increases in metabolites reflecting mitochondrial dysfunction (particularly long-chain acylcarnitines) and aromatic amino acids and decreases in branched-chain amino acids were associated with worse HF-related outcomes in the overall cohort, with consistency across treatment and LVEF. CONCLUSIONS SGLT2is demonstrate common (fatty acid) and distinct (ketogenic) metabolic signatures across the LVEF spectrum. Changes in key pathways related to fatty acid and amino acid metabolism are associated with HF-related end points and may serve as therapeutic targets across HF subtypes. REGISTRATION URL: https://www.clinicaltrials.gov; Unique Identifiers: NCT03030235 and NCT02653482.
Collapse
Affiliation(s)
- Senthil Selvaraj
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | - Shachi Patel
- Saint Luke’s Mid America Heart Institute, Kansas City, MO
| | - Andrew J. Sauer
- Saint Luke’s Mid America Heart Institute, Kansas City, MO
- University of Missouri-Kansas City, MO
| | - Robert W. McGarrah
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | - Philip Jones
- Saint Luke’s Mid America Heart Institute, Kansas City, MO
| | - Lydia Coulter Kwee
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | | | - Olga Ilkayeva
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
- Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | | | | | - Barry A. Borlaug
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Dalane W. Kitzman
- Department of Internal Medicine, Sections on Cardiovascular Medicine and Geriatrics, Wake Forest School of Medicine, Winston-Salem, NC
| | - Sanjiv J. Shah
- Division of Cardiology, Department of Medicine and Bluhm Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Kenneth B. Margulies
- Division of Cardiology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Mansoor Husain
- Ted Rogers Centre for Heart Research, University of Toronto, Canada
| | | | - Darren K. McGuire
- University of Texas Southwestern Medical Center and Parkland Health and Hospital System, Dallas, TX
| | - David E. Lanfear
- Center for Individual and Genomic Medicine Research and Division of Cardiovascular Medicine, Henry Ford Hospital, Detroit MI
| | - Ali Javaheri
- Washington University School of Medicine, St. Louis, MO; John Cochran VA, St. Louis, MO
| | | | - Robert J. Mentz
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Kavita Sharma
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Mikhail N. Kosiborod
- Saint Luke’s Mid America Heart Institute, Kansas City, MO
- University of Missouri-Kansas City, MO
| | - Svati H. Shah
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| |
Collapse
|
29
|
Harada T, Tada A, Borlaug BA. Imaging and mechanisms of heart failure with preserved ejection fraction: a state-of-the-art review. Eur Heart J Cardiovasc Imaging 2024; 25:1475-1490. [PMID: 38912836 DOI: 10.1093/ehjci/jeae152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 06/15/2024] [Indexed: 06/25/2024] Open
Abstract
Understanding of the pathophysiology of heart failure with preserved ejection fraction (HFpEF) has advanced rapidly over the past two decades. Currently, HFpEF is recognized as a heterogeneous syndrome, and there is a growing movement towards developing personalized treatments based on phenotype-guided strategies. Left ventricular dysfunction is a fundamental pathophysiological abnormality in HFpEF; however, recent evidence also highlights significant roles for the atria, right ventricle, pericardium, and extracardiac contributors. Imaging plays a central role in characterizing these complex and highly integrated domains of pathophysiology. This review focuses on established evidence, recent insights, and the challenges that need to be addressed concerning the pathophysiology of HFpEF, with a focus on imaging-based evaluations and opportunities for further research.
Collapse
Affiliation(s)
- Tomonari Harada
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Atsushi Tada
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Barry A Borlaug
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
30
|
Ipek R, Holland J, Cramer M, Rider O. CMR to characterize myocardial structure and function in heart failure with preserved left ventricular ejection fraction. Eur Heart J Cardiovasc Imaging 2024; 25:1491-1504. [PMID: 39205602 PMCID: PMC11522877 DOI: 10.1093/ehjci/jeae224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/21/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Despite remarkable progress in therapeutic drugs, morbidity, and mortality for heart failure (HF) remains high in developed countries. HF with preserved ejection fraction (HFpEF) now accounts for around half of all HF cases. It is a heterogeneous disease, with multiple aetiologies, and as such poses a significant diagnostic challenge. Cardiac magnetic resonance (CMR) has become a valuable non-invasive modality to assess cardiac morphology and function, but beyond that, the multi-parametric nature of CMR allows novel approaches to characterize haemodynamics and with magnetic resonance spectroscopy (MRS), the study of metabolism. Furthermore, exercise CMR, when combined with lung water imaging provides an in-depth understanding of the underlying pathophysiological and mechanistic processes in HFpEF. Thus, CMR provides a comprehensive phenotyping tool for HFpEF, which points towards a targeted and personalized therapy with improved diagnostics and prevention.
Collapse
Affiliation(s)
- Rojda Ipek
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research (OCMR), John Radcliffe Hospital, Level 0, University of Oxford, Oxford, OX3 9DU, UK
- Divison of Cardiology, Pulmonary Disease and Vascular Medicine, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Jennifer Holland
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research (OCMR), John Radcliffe Hospital, Level 0, University of Oxford, Oxford, OX3 9DU, UK
| | - Mareike Cramer
- Divison of Cardiology, Pulmonary Disease and Vascular Medicine, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Oliver Rider
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research (OCMR), John Radcliffe Hospital, Level 0, University of Oxford, Oxford, OX3 9DU, UK
| |
Collapse
|
31
|
Bhandari M, Pradhan A, Vishwakarma P, Singh A, Sethi R. Sodium glucose cotransporter 2 inhibitors in the management of heart failure: Veni, Vidi, and Vici. World J Cardiol 2024; 16:550-563. [PMID: 39492976 PMCID: PMC11525799 DOI: 10.4330/wjc.v16.i10.550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 08/25/2024] [Accepted: 09/06/2024] [Indexed: 10/17/2024] Open
Abstract
Heart failure (HF) is a chronic disease associated with high morbidity and mortality rates. Renin-angiotensin-aldosterone system blockers (including angiotensin receptor/neprilysin inhibitors), beta-blockers, and mineralocorticoid receptor blockers remain the mainstay of pharmacotherapy for HF with reduced ejection fraction (HFrEF). However, despite the use of guideline-directed medical therapy, the mortality from HFrEF remains high. HF with preserved ejection fraction (HFpEF) comprises approximately half of the total incident HF cases; however, unlike HFrEF, there are no proven therapies for this condition. Sodium glucose cotransporter-2 inhibitors (SGLT-2is) represent a new class of pharmacological agents approved for diabetes mellitus (DM) that inhibit SGLT-2 receptors in the kidney. A serendipitous finding from seminal trials of SGLT-2is in DM was the significant improvement in renal and cardiovascular (CV) outcomes. More importantly, the improvement in HF hospitalization (HHF) in the CV outcomes trials of SGLT-2is was striking. Multiple mechanisms have been proposed for the pleiotropic effects of SGLT-2is beyond their glycemic control. However, as patients with HF were not included in any of these trials, it can be considered as a primary intervention. Subsequently, two landmark studies of SGLT-2is in patients with HFrEF, namely, an empagliflozin outcome trial in patients with chronic HF and a reduced ejection fraction (EMPEROR-Reduced) and dapagliflozin and prevention of adverse outcomes in HF (DAPA-HF), demonstrated significant improvement in HHF and CV mortality regardless of the presence of DM. These impressive results pitchforked these drugs as class I indications in patients with HFrEF across major guidelines. Thereafter, empagliflozin outcome trial in patients with chronic HF with preserved ejection fraction (EMPEROR-Preserved) and dapagliflozin evaluation to improve the lives of patients with preserved ejection fraction HF (DELIVER) trials successively confirmed that SGLT-2is also benefit patients with HFpEF with or without DM. These results represent a watershed as they constitute the first clinically meaningful therapy for HFpEF in the past three decades of evolution of HF management. Emerging positive data for the use of SGLT-2is in acute HF and post-myocardial infarction scenarios have strengthened the pivotal role of these agents in the realm of HF. In a short span of time, these classes of drugs have captivated the entire scenario of HF.
Collapse
Affiliation(s)
- Monika Bhandari
- Department of Cardiology, King Georg's Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Akshyaya Pradhan
- Department of Cardiology, King Georg's Medical University, Lucknow 226003, Uttar Pradesh, India.
| | - Pravesh Vishwakarma
- Department of Cardiology, King Georg's Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Abhishek Singh
- Department of Cardiology, King Georg's Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Rishi Sethi
- Department of Cardiology, King Georg's Medical University, Lucknow 226003, Uttar Pradesh, India
| |
Collapse
|
32
|
Tiwari K, Deora S, Choudhary R, Kaushik A, Dwivedi P, Singh S, Ambwani S, Midha N, Shukla R, Sankanagoudar S, Shamim MA, Tiwari VK, Yadav I, Dodiya R, Varthya SB. Rationale and design of Dapagliflozin vErsus SacubiTrIl-valsartaN therapY in Heart Failure with reduced ejection fraction (DESTINY-HF): a pragmatic randomised controlled trial protocol. BMJ Open 2024; 14:e089562. [PMID: 39424390 PMCID: PMC11492940 DOI: 10.1136/bmjopen-2024-089562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/30/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Heart failure affects almost 64 million people, with more than half of it constituting heart failure with reduced ejection fraction (HFrEF). Angiotensin receptor-neprilysin inhibitors (ARNI) and sodium-glucose cotransporter-2 (SGLT2) inhibitors (SGLT2i) are in the first line for HFrEF, but no head-to-head trials are available. Moreover, growth differentiation factor-15 (GDF-15) has been demonstrated as a promising prognostic marker, specifically for HFrEF, but has not been explored much. METHODS This pragmatic randomised controlled trial recruits 100 patients with HFrEF (ejection fraction <40%) of New York Heart Association (NYHA) II-III and allocates them in a 1:1 ratio to the dapagliflozin and sacubitril/valsartan groups. The primary objective is to assess the difference in N-terminal pro-brain natriuretic peptide serum levels at the end of 16 weeks. The secondary efficacy objectives are to assess GDF-15, Kansas City Cardiomyopathy Questionnaire-overall summary score and estimated glomerular filtration rate. Patients will be assessed at baseline, fourth week and 16th week after randomisation. As health technology assessment practices widely differ in countries, cost assessment is a vital factor to consider. The cost needed to treat one cardiovascular event is also compared between both groups. The occurrence of safety events will also be evaluated at each follow-up point. CONCLUSION This pragmatic study aims to compare the efficacy, safety and cost-effectiveness of dapagliflozin versus sacubitril/valsartan in patients with HFrEF in real-world settings. The study aims to provide clinicians with data to make informed decisions regarding the preferred drug class. Additionally, examining the impact of ARNI and SGLT2i on GDF-15 levels could offer better insights into prognosis among patients with HFrEF. ETHICS AND DISSEMINATION This study involves human participants and was approved by Institutional Ethics Committee at AlIMS Jodhpur with reference number AIIMS/IEC/2023/5842 approved this study. Participants gave informed consent to participate in the study before taking part. The research findings will be disseminated via closed group discussions at the site of study, scientific conferences, peer-reviewed published manuscripts, and social media. TRIAL REGISTRATION NUMBER CTRI/2023/12/060772.
Collapse
Affiliation(s)
- Krishna Tiwari
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Surender Deora
- Department of Cardiology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Rahul Choudhary
- Department of Cardiology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Atul Kaushik
- Department of Cardiology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Pradeep Dwivedi
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Surjit Singh
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Sneha Ambwani
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Naresh Midha
- Department of General Medicine, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Ravindra Shukla
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences, New Delhi, India
| | | | - Muhammad Aaqib Shamim
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Vikas Kumar Tiwari
- Laboratory of Systems Neuroscience, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Physiology, JIET Medical College and Hospital, Jodhpur, Rajasthan, India
| | - Isha Yadav
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Rakesh Dodiya
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Shoban Babu Varthya
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| |
Collapse
|
33
|
Nilsson CN, Ersbøll MK, Gustafsson F. Haemodynamic Effects of Sodium-Glucose Cotransporter 2 Inhibitor Treatment in Chronic Heart Failure Patients. Card Fail Rev 2024; 10:e09. [PMID: 39309522 PMCID: PMC11413986 DOI: 10.15420/cfr.2023.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/07/2024] [Indexed: 09/25/2024] Open
Abstract
Sodium-glucose cotransporter 2 inhibitors (SGLT-2i) are now recommended in the current European Society of Cardiology/American College of Cardiology guidelines for the treatment of heart failure (HF) across the spectrum of left ventricular ejection fraction (LVEF) and several large trials have documented the beneficial effects of this drug class on cardiovascular outcomes. Although the clinical efficacy of SGLT-2 inhibition in HF is now well recognised, research is still ongoing to better understand the underlying mechanistic effects of this drug class. In this paper we assess the haemodynamic effects following SGLT-2i treatment in HF patients by reviewing the current literature. We focus our review on preload of the LV in terms of filling pressure and pulmonary artery pressure, cardiac output and afterload. We discuss these variables stratified according to HF with reduced LVEF (HFrEF) and HF with preserved LVEF (HFpEF). Finally, we examine the evidence of LV remodelling in the setting of SGLT-2i-related changes in haemodynamics.
Collapse
Affiliation(s)
- C Noah Nilsson
- Department of Cardiology, Copenhagen University Hospital Rigshospitalet Denmark
| | | | - Finn Gustafsson
- Department of Cardiology, Copenhagen University Hospital Rigshospitalet Denmark
| |
Collapse
|
34
|
von Haehling S, Assmus B, Bekfani T, Dworatzek E, Edelmann F, Hashemi D, Hellenkamp K, Kempf T, Raake P, Schütt KA, Wachter R, Schulze PC, Hasenfuss G, Böhm M, Bauersachs J. Heart failure with preserved ejection fraction: diagnosis, risk assessment, and treatment. Clin Res Cardiol 2024; 113:1287-1305. [PMID: 38602566 PMCID: PMC11371894 DOI: 10.1007/s00392-024-02396-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/02/2024] [Indexed: 04/12/2024]
Abstract
The aetiology of heart failure with preserved ejection fraction (HFpEF) is heterogenous and overlaps with that of several comorbidities like atrial fibrillation, diabetes mellitus, chronic kidney disease, valvular heart disease, iron deficiency, or sarcopenia. The diagnosis of HFpEF involves evaluating cardiac dysfunction through imaging techniques and assessing increased left ventricular filling pressure, which can be measured directly or estimated through various proxies including natriuretic peptides. To better narrow down the differential diagnosis of HFpEF, European and American heart failure guidelines advocate the use of different algorithms including comorbidities that require diagnosis and rigorous treatment during the evaluation process. Therapeutic recommendations differ between guidelines. Whilst sodium glucose transporter 2 inhibitors have a solid evidence base, the recommendations differ with regard to the use of inhibitors of the renin-angiotensin-aldosterone axis. Unless indicated for specific comorbidities, the use of beta-blockers should be discouraged in HFpEF. The aim of this article is to provide an overview of the current state of the art in HFpEF diagnosis, clinical evaluation, and treatment.
Collapse
Affiliation(s)
- Stephan von Haehling
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Strasse 40, 37075, Göttingen, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany.
| | - Birgit Assmus
- Department of Cardiology and Angiology, Universitätsklinikum Gießen und Marburg, Giessen, Germany
| | - Tarek Bekfani
- Department of Cardiology and Angiology, Universitätsklinikum Magdeburg, Magdeburg, Germany
| | - Elke Dworatzek
- Institute of Gender in Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Frank Edelmann
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Campus Virchow-Klinikum, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Djawid Hashemi
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Campus Virchow-Klinikum, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Digital Clinician Scientist Program, Berlin, Germany
| | - Kristian Hellenkamp
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
| | - Tibor Kempf
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Philipp Raake
- I. Medical Department, Cardiology, Pneumology, Endocrinology and Intensive Care Medicine, University Hospital Augsburg, University of Augsburg, Augsburg, Germany
| | - Katharina A Schütt
- Department of Internal Medicine I, University Hospital RWTH Aachen, Aachen, Germany
| | - Rolf Wachter
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig, Germany
| | - Paul Christian Schulze
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, FSU, Jena, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Michael Böhm
- Kardiologie, Angiologie und Internistische Intensivmedizin, Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, Saarland University, Homburg, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
35
|
Hahn VS, Selvaraj S, Sharma K, Shah SH. Towards Metabolomic-Based Precision Approaches for Classifying and Treating Heart Failure. JACC Basic Transl Sci 2024; 9:1144-1158. [PMID: 39444924 PMCID: PMC11494393 DOI: 10.1016/j.jacbts.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/15/2024] [Accepted: 04/05/2024] [Indexed: 10/25/2024]
Abstract
Both heart failure and cardiometabolic disease are on the rise, and abnormal cardiac and peripheral metabolism are central to the syndrome of heart failure. Advances in metabolomic profiling have improved our understanding of the heart's metabolic flexibility in patients with and without heart failure. Prior studies have noted patients with heart failure display metabolomic profiles associated with marked abnormalities in the metabolism of fatty acids, branched-chain amino acids, ketones, and glucose compared with control subjects. Metabolomics can highlight specific pathways that are dysregulated; however, other metabolites beyond those related to fuel metabolism may also play a role in precision-medicine approaches. Novel approaches include metabolic flux studies, spatial and single-cell analysis, serial monitoring of treatment response, and integration with other -omics data. The goal of these innovative approaches should be to harness metabolomic technologies to affect precision care for patients with heart failure.
Collapse
Affiliation(s)
- Virginia S. Hahn
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Senthil Selvaraj
- Division of Cardiology, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Kavita Sharma
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Svati H. Shah
- Division of Cardiology, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
36
|
Bray JJ, Coronelli M, Scott SG, Henry JA, Couch LS, Ahmad M, Ormerod J, Gamble J, Betts TR, Lewis A, Rider OJ, Green PG, Herring N. The effect of sodium-glucose co-transporter 2 inhibitors on outcomes after cardiac resynchronization therapy. ESC Heart Fail 2024; 11:2314-2322. [PMID: 38649305 PMCID: PMC11287366 DOI: 10.1002/ehf2.14784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/14/2024] [Accepted: 03/10/2024] [Indexed: 04/25/2024] Open
Abstract
AIMS The trials upon which recommendations for the use of cardiac resynchronization therapy (CRT) in heart failure used optimal medical therapy (OMT) before sodium-glucose co-transporter 2 inhibitors (SGLT2i). Moreover, the SGLT2i heart failure trials included only a small proportion of participants with CRT, and therefore, it remains uncertain whether SGLT2i should be considered part of OMT prior to CRT. METHODS AND RESULTS We compared electrocardiogram (ECG) and echocardiographic responses to CRT as well as hospitalization and mortality rates in consecutive patients undergoing implantation at a large tertiary centre between January 2019 to June 2022 with and without SGLT2i treatment. Three hundred seventy-four participants were included aged 74.0 ± 11.5 years (mean ± standard deviation), with a left ventricular ejection fraction (LVEF) of 31.8 ± 9.9% and QRS duration of 161 ± 29 ms. The majority had non-ischaemic cardiomyopathy (58%) and were in NYHA Class II/III (83.6%). These characteristics were similar between patients with (n = 66) and without (n = 308) prior SGLT2i treatment. Both groups demonstrated similar evidence of response to CRT in terms of QRS duration shortening, and improvements in LVEF, left ventricular end-diastolic inner-dimension (LVIDd) and diastolic function (E/A and e/e'). While there was no difference in rates of hospitalization (for heart failure or overall), mortality was significantly lower in patients treated with SGLT2i compared with those who were not (6.5 vs. 16.6%, P = 0.049). CONCLUSIONS We observed an improvement in mortality in patients undergoing CRT prescribed SGLT2i compared with those not prescribed SGLT2i, despite similar degrees of reverse remodelling. The authors recommend starting SGLT2i prior to CRT implantation, where it does not delay implantation.
Collapse
Affiliation(s)
| | - Marco Coronelli
- Oxford Heart CentreOxford University Hospitals, NHS TrustOxfordUK
| | - Sam G.C. Scott
- Oxford Heart CentreOxford University Hospitals, NHS TrustOxfordUK
| | - John A. Henry
- Oxford Heart CentreOxford University Hospitals, NHS TrustOxfordUK
| | - Liam S. Couch
- Oxford Heart CentreOxford University Hospitals, NHS TrustOxfordUK
| | | | - Julian Ormerod
- Oxford Heart CentreOxford University Hospitals, NHS TrustOxfordUK
| | - James Gamble
- Oxford Heart CentreOxford University Hospitals, NHS TrustOxfordUK
| | - Timothy R. Betts
- Oxford Heart CentreOxford University Hospitals, NHS TrustOxfordUK
- Department of Cardiovascular MedicineUniversity of OxfordOxfordUK
| | - Andrew Lewis
- Oxford Heart CentreOxford University Hospitals, NHS TrustOxfordUK
- Department of Cardiovascular MedicineUniversity of OxfordOxfordUK
| | - Oliver J. Rider
- Oxford Heart CentreOxford University Hospitals, NHS TrustOxfordUK
- Department of Cardiovascular MedicineUniversity of OxfordOxfordUK
| | | | - Neil Herring
- Oxford Heart CentreOxford University Hospitals, NHS TrustOxfordUK
- Department of Cardiovascular MedicineUniversity of OxfordOxfordUK
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| |
Collapse
|
37
|
Forelli N, Eaton D, Patel J, Bowman CE, Kawakami R, Kuznetsov IA, Li K, Brady C, Bedi K, Yang Y, Koya K, Megill E, Kanter DS, Smith LG, Bowman GR, Snyder N, Edwards J, Margulies K, Arany Z. SGLT2 inhibitors activate pantothenate kinase in the human heart. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.26.605401. [PMID: 39091820 PMCID: PMC11291109 DOI: 10.1101/2024.07.26.605401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Inhibitors of sodium glucose cotransporter-2 (SGLT2i) demonstrate strong symptomatic and mortality benefits in the treatment of heart failure but appear to do so independently of SGLT2. The relevant pharmacologic target of SGLT2i remains unclear. We show here that SGLT2i directly activate pantothenate kinase 1 (PANK1), the rate-limiting enzyme that initiates the conversion of pantothenate (vitamin B5) to coenzyme-A (CoA), an obligate co-factor for all major pathways of fuel use in the heart. Using stable-isotope infusion studies, we show that SGLT2i promote pantothenate consumption, activate CoA synthesis, rescue decreased levels of CoA in human failing hearts, and broadly stimulate fuel use in ex vivo perfused human cardiac blocks from patients with heart failure. Furthermore, we show that SGLT2i bind to PANK1 directly at physiological concentrations and promote PANK1 enzymatic activity in assays with purified components. Novel in silico dynamic modeling identified the site of SGLT2i binding on PANK1 and indicated a mechanism of activation involving prevention of allosteric inhibition of PANK1 by acyl-CoA species. Finally, we show that inhibition of PANK1 prevents SGLT2i-mediated increased contractility of isolated adult human cardiomyocytes. In summary, we demonstrate robust and specific off-target activation of PANK1 by SGLT2i, promoting CoA synthesis and efficient fuel use in human hearts, providing a likely explanation for the remarkable clinical benefits of SGLT2i.
Collapse
Affiliation(s)
- Nicholas Forelli
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Deborah Eaton
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jiten Patel
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Caitlyn E. Bowman
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ryo Kawakami
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ivan A. Kuznetsov
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Kristina Li
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Claire Brady
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Kenneth Bedi
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Yijun Yang
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Kaustubh Koya
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Emily Megill
- Aging & Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Daniel S. Kanter
- Aging & Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Louis G. Smith
- Departments of Biochemistry & Biophysics, and Bioengineering, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Gregory R. Bowman
- Departments of Biochemistry & Biophysics, and Bioengineering, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Nathaniel Snyder
- Aging & Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Jonathan Edwards
- Division of Cardiology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Kenneth Margulies
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Zoltan Arany
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
38
|
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
|
39
|
Bornstein MR, Tian R, Arany Z. Human cardiac metabolism. Cell Metab 2024; 36:1456-1481. [PMID: 38959861 PMCID: PMC11290709 DOI: 10.1016/j.cmet.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 04/12/2024] [Accepted: 06/05/2024] [Indexed: 07/05/2024]
Abstract
The heart is the most metabolically active organ in the human body, and cardiac metabolism has been studied for decades. However, the bulk of studies have focused on animal models. The objective of this review is to summarize specifically what is known about cardiac metabolism in humans. Techniques available to study human cardiac metabolism are first discussed, followed by a review of human cardiac metabolism in health and in heart failure. Mechanistic insights, where available, are reviewed, and the evidence for the contribution of metabolic insufficiency to heart failure, as well as past and current attempts at metabolism-based therapies, is also discussed.
Collapse
Affiliation(s)
- Marc R Bornstein
- Cardiovascular Institute Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rong Tian
- Mitochondria and Metabolism Center, Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, USA
| | - Zoltan Arany
- Cardiovascular Institute Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
40
|
Selvaraj S, Patel S, Sauer AJ, McGarrah RW, Jones P, Kwee LC, Windsor SL, Ilkayeva O, Muehlbauer MJ, Newgard CB, Borlaug BA, Kitzman DW, Shah SJ, Shah SH, Kosiborod MN. Targeted Metabolomic Profiling of Dapagliflozin in Heart Failure With Preserved Ejection Fraction: The PRESERVED-HF Trial. JACC. HEART FAILURE 2024; 12:999-1011. [PMID: 38639697 PMCID: PMC11153021 DOI: 10.1016/j.jchf.2024.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/12/2024] [Accepted: 02/14/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND Although sodium glucose co-transporter 2 inhibitors (SGLT2is) improve heart failure (HF)-related symptoms and outcomes in HF with preserved ejection fraction (HFpEF), underlying mechanisms remain unclear. In HF with reduced EF, dapagliflozin altered ketone and fatty acid metabolites vs placebo; however, metabolite signatures of SGLT2is have not been well elucidated in HFpEF. OBJECTIVES The goal of this study was to assess whether SGLT2i treatment altered systemic metabolic pathways and their relationship to outcomes in HFpEF. METHODS Targeted profiling of 64 metabolites was performed from 293 participants in PRESERVED-HF (Dapagliflozin in PRESERVED Ejection Fraction Heart Failure), a 12-week, placebo-controlled trial of dapagliflozin. Linear regression assessed changes in metabolite factors defined by principal components analysis (PCA) with dapagliflozin vs placebo. The relationship between changes in metabolite factors with changes in study endpoints was also assessed. RESULTS The mean age was 70 ± 11 years, 58% were female, and 29% were Black. There were no significant differences in 12 PCA-derived metabolite factors between treatment arms, including metabolites reflecting ketone, fatty acid, or branched-chain amino acid (BCAA) pathways. Combining treatment arms, changes in BCAAs and branched-chain ketoacids were negatively associated with changes in N-terminal pro-B-type natriuretic peptide; changes in medium-/long-chain acylcarnitines were positively associated with changes in N-terminal pro-B-type natriuretic peptide and negatively associated with changes in 6-minute walk test distance; and changes in ketones were negatively associated with changes in weight, without treatment interaction. CONCLUSIONS Leveraging targeted metabolomics in a placebo-controlled SGLT2i trial of HFpEF, dapagliflozin did not alter systemic metabolic as reflected by circulating metabolites, in contrast with reported effects in HF with reduced ejection fraction. Metabolite biomarkers reflecting BCAA, ketone, and fatty acid metabolism were associated with markers of disease severity, suggesting a role for potential novel treatment targets. (Dapagliflozin in PRESERVED Ejection Fraction Heart Failure [PRESERVED-HF]; NCT03030235).
Collapse
Affiliation(s)
- Senthil Selvaraj
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA; Duke Molecular Physiology Institute, Durham, North Carolina, USA
| | - Shachi Patel
- Saint Luke's Mid America Heart Institute, Kansas City, Missouri, USA
| | - Andrew J Sauer
- Saint Luke's Mid America Heart Institute, Kansas City, Missouri, USA; University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Robert W McGarrah
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA; Duke Molecular Physiology Institute, Durham, North Carolina, USA
| | - Philip Jones
- Saint Luke's Mid America Heart Institute, Kansas City, Missouri, USA
| | | | - Sheryl L Windsor
- Saint Luke's Mid America Heart Institute, Kansas City, Missouri, USA
| | - Olga Ilkayeva
- Duke Molecular Physiology Institute, Durham, North Carolina, USA; Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | | | | | - Barry A Borlaug
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Dalane W Kitzman
- Department of Internal Medicine, Sections on Cardiovascular Medicine and Geriatrics, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Sanjiv J Shah
- Division of Cardiology, Department of Medicine and Bluhm Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Svati H Shah
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA; Duke Molecular Physiology Institute, Durham, North Carolina, USA
| | - Mikhail N Kosiborod
- Saint Luke's Mid America Heart Institute, Kansas City, Missouri, USA; University of Missouri-Kansas City, Kansas City, Missouri, USA.
| |
Collapse
|
41
|
Arvidsson PM, Berg J, Carlsson M, Arheden H. Noninvasive Pressure-Volume Loops Predict Major Adverse Cardiac Events in Heart Failure With Reduced Ejection Fraction. JACC. ADVANCES 2024; 3:100946. [PMID: 38938852 PMCID: PMC11198266 DOI: 10.1016/j.jacadv.2024.100946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/05/2024] [Accepted: 03/06/2024] [Indexed: 06/29/2024]
Abstract
Background Heart failure with reduced ejection fraction (HFrEF) is characterized by ventricular remodeling and impaired myocardial energetics. Left ventricular pressure-volume (PV) loop analysis can be performed noninvasively using cardiovascular magnetic resonance (CMR) imaging to assess cardiac thermodynamic efficiency. Objectives The aim of the study was to investigate whether noninvasive PV loop parameters, derived from CMR, could predict major adverse cardiac events (MACE) in HFrEF patients. Methods PV loop parameters (stroke work, ventricular efficiency, external power, contractility, and energy per ejected volume) were computed from CMR cine images and brachial blood pressure. The primary end point was MACE (cardiovascular death, heart failure (HF) hospitalization, myocardial infarction, revascularization, ventricular tachycardia/fibrillation, heart transplantation, or left ventricular assist device implantation within 5 years). Associations between PV loop parameters and MACE were evaluated using multivariable Cox regression. Results One hundred and sixty-four HFrEF patients (left ventricular ejection fraction ≤40%, age 63 [IQR: 55-70] years, 79% male) who underwent clinical CMR examination between 2004 and 2014 were included. Eighty-eight patients (54%) experienced at least one MACE after an average of 2.8 years. Unadjusted models demonstrated a significant association between MACE and all PV loop parameters (P < 0.05 for all), HF etiology (P < 0.001), left ventricular ejection fraction (P = 0.003), global longitudinal strain (P < 0.001), and N-terminal prohormone of brain natriuretic peptide level (P = 0.001). In the multivariable Cox regression analysis adjusted for age, sex, hypertension, diabetes, and HF etiology, ventricular efficiency was associated with MACE (HR: 1.04 (95% CI: 1.01-1.08) per-% decrease, P = 0.01). Conclusions Ventricular efficiency, derived from noninvasive PV loop analysis from standard CMR scans, is associated with MACE in patients with HFrEF.
Collapse
Affiliation(s)
- Per M. Arvidsson
- Department of Clinical Sciences Lund, Clinical Physiology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Jonathan Berg
- Department of Clinical Sciences Lund, Clinical Physiology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Marcus Carlsson
- Department of Clinical Sciences Lund, Clinical Physiology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Håkan Arheden
- Department of Clinical Sciences Lund, Clinical Physiology, Skåne University Hospital, Lund University, Lund, Sweden
| |
Collapse
|
42
|
Tsuruta H, Yamahara K, Yasuda-Yamahara M, Kume S. Emerging Pathophysiological Roles of Ketone Bodies. Physiology (Bethesda) 2024; 39:0. [PMID: 38260943 DOI: 10.1152/physiol.00031.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 01/24/2024] Open
Abstract
The discovery of insulin approximately a century ago greatly improved the management of diabetes, including many of its life-threatening acute complications like ketoacidosis. This breakthrough saved many lives and extended the healthy lifespan of many patients with diabetes. However, there is still a negative perception of ketone bodies stemming from ketoacidosis. Originally, ketone bodies were thought of as a vital source of energy during fasting and exercise. Furthermore, in recent years, research on calorie restriction and its potential impact on extending healthy lifespans, as well as studies on ketone bodies, have gradually led to a reevaluation of the significance of ketone bodies in promoting longevity. Thus, in this review, we discuss the emerging and hidden roles of ketone bodies in various organs, including the heart, kidneys, skeletal muscles, and brain, as well as their potential impact on malignancies and lifespan.
Collapse
Affiliation(s)
- Hiroaki Tsuruta
- Department of Medicine, Shiga University of Medical Science, Seta, Otsu, Shiga, Japan
| | - Kosuke Yamahara
- Department of Medicine, Shiga University of Medical Science, Seta, Otsu, Shiga, Japan
| | - Mako Yasuda-Yamahara
- Department of Medicine, Shiga University of Medical Science, Seta, Otsu, Shiga, Japan
| | - Shinji Kume
- Department of Medicine, Shiga University of Medical Science, Seta, Otsu, Shiga, Japan
| |
Collapse
|
43
|
Gao M, Bhatia K, Kapoor A, Badimon J, Pinney SP, Mancini DM, Santos-Gallego CG, Lala A. SGLT2 Inhibitors, Functional Capacity, and Quality of Life in Patients With Heart Failure: A Systematic Review and Meta-Analysis. JAMA Netw Open 2024; 7:e245135. [PMID: 38573633 PMCID: PMC11192183 DOI: 10.1001/jamanetworkopen.2024.5135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/08/2024] [Indexed: 04/05/2024] Open
Abstract
Importance The associations of sodium glucose cotransporter-2 inhibitors (SGLT2is) with reduction in mortality and hospitalization rates in patients with heart failure (HF) are well established. However, their association with improving functional capacity and quality of life (QOL) has been variably studied and less reported. Objective To provide evidence on the extent to which SGLT2is are associated with improvement on objective measures of functional capacity and QOL in patients living with HF. Data Sources The MEDLINE, EMBASE, and Cochrane databases were systematically searched for relevant articles on July 31, 2023. Study Selection Randomized, placebo-controlled clinical trials reporting the effect of SGLT2i on functional outcomes of exercise capacity (peak oxygen consumption [peak VO2] or 6-minute walk distance [6MWD]) and/or QOL using validated questionnaires for patients with HF were included. Data Extraction and Synthesis Data were extracted by 2 authors following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses 2020 guidelines, and a meta-analysis using the restricted maximum likelihood random-effects model was conducted. Main Outcomes and Measures Outcomes of interest included changes in peak VO2, 6MWD, and Kansas City Cardiomyopathy Questionnaire-12 total symptom score (KCCQ-TSS), clinical summary score (KCCQ-CSS), and overall summary score (KCCQ-OSS). Results In this meta-analysis of 17 studies, 23 523 patients (mean [range] age, 69 [60-75] years) were followed over a period ranging from 12 to 52 weeks. Four studies included peak VO2 as an outcome, 7 studies included 6MWD, and 10 studies reported KCCQ scores. Mean (SD) left ventricular ejection fraction was 43.5% (12.4%). Compared with controls, patients receiving SGLT2i treatment experienced significant increases in peak VO2 (mean difference [MD], 1.61 mL/kg/min; 95% CI, 0.59-2.63 mL/kg/min; P = .002) and 6MWD (MD, 13.09 m; 95% CI, 1.20-24.97 m; P = .03). SGLT2i use was associated with increased KCCQ-TSS (MD, 2.28 points; 95% CI, 1.74-2.81 points; P < .001), KCCQ-CSS (MD, 2.14 points; 95% CI, 1.53-2.74 points; P < .001), and KCCQ-OSS (MD, 1.90 points; 95% CI, 1.41-2.39 points; P < .001) scores. Subgroup analysis and meta-regression demonstrated almost all improvements were consistent across ejection fraction, sex, and the presence of diabetes. Conclusions and Relevance These findings suggest that in addition to known clinical associations with mortality and hospitalization outcomes, SGLT2i use is associated with improvement in outcomes of interest to patients' everyday lives as measured by objective assessments of maximal exercise capacity and validated QOL questionnaires, regardless of sex or ejection fraction.
Collapse
Affiliation(s)
- Michael Gao
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kirtipal Bhatia
- Mount Sinai Fuster Heart Hospital, Mount Sinai Morningside, New York, New York
| | - Arjun Kapoor
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Juan Badimon
- Department of Population Health Science, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sean P. Pinney
- Mount Sinai Fuster Heart Hospital, Mount Sinai Morningside, New York, New York
| | - Donna M. Mancini
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Carlos G. Santos-Gallego
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Anuradha Lala
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
44
|
Dabour MS, George MY, Daniel MR, Blaes AH, Zordoky BN. The Cardioprotective and Anticancer Effects of SGLT2 Inhibitors: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2024; 6:159-182. [PMID: 38774006 PMCID: PMC11103046 DOI: 10.1016/j.jaccao.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 05/24/2024] Open
Abstract
Sodium-glucose cotransporter-2 (SGLT2) inhibitors, originally approved for type 2 diabetes mellitus, have demonstrated efficacy in reducing cardiovascular events, particularly heart failure, in patients with and without diabetes. An intriguing research area involves exploring the potential application of SGLT2 inhibitors in cardio-oncology, aiming to mitigate the cardiovascular adverse events associated with anticancer treatments. These inhibitors present a unique dual nature, offering both cardioprotective effects and anticancer properties, conferring a double benefit for cardio-oncology patients. In this review, the authors first examine the established cardioprotective effects of SGLT2 inhibitors in heart failure and subsequently explore the existing body of evidence, including both preclinical and clinical studies, that supports the use of SGLT2 inhibitors in the context of cardio-oncology. The authors further discuss the mechanisms through which SGLT2 inhibitors protect against cardiovascular toxicity secondary to cancer treatment. Finally, they explore the potential anticancer effects of SGLT2 inhibitors along with their proposed mechanisms.
Collapse
Affiliation(s)
- Mohamed S. Dabour
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Mina Y. George
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mary R. Daniel
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Anne H. Blaes
- Division of Hematology/Oncology/Transplantation, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - Beshay N. Zordoky
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
45
|
Triposkiadis F, Xanthopoulos A, Drakos SG, Boudoulas KD, Briasoulis A, Skoularigis J, Tsioufis K, Boudoulas H, Starling RC. Back to the basics: The need for an etiological classification of chronic heart failure. Curr Probl Cardiol 2024; 49:102460. [PMID: 38346611 DOI: 10.1016/j.cpcardiol.2024.102460] [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: 02/09/2024] [Accepted: 02/09/2024] [Indexed: 02/23/2024]
Abstract
The left ventricular (LV) ejection fraction (LVEF), despite its severe limitations, has had an epicentral role in heart failure (HF) classification, management, and risk stratification for decades. The major argument favoring the LVEF based HF classification has been that it defines groups of patients in which treatment is effective. However, this reasoning has recently collapsed, since medical treatment with neurohormonal inhibitors, has proved beneficial in most HF patients regardless of the LVEF. In addition, there has been compelling evidence, that the LVEF provides poor guidance for device treatment of chronic HF (implantation of cardioverter defibrillator, cardiac resynchronization therapy) since sudden cardiac death may occur and cardiac dyssynchronization may be disastrous in all HF patients. The same holds true for LV assist device implantation, in which the LVEF has been used as a surrogate for LV size. In this review article we update the evidence questioning the use of LVEF-based HF classification and argue that guidance of chronic HF treatment should transition to more contemporary concepts. Specifically, we propose an etiologic chronic HF classification predominantly based on epidemiological data, which will be foundational for further higher resolution phenotyping in the emerging era of precision medicine.
Collapse
Affiliation(s)
- Filippos Triposkiadis
- School of Medicine, European University Cyprus, Nicosia 2404, Cyprus; Department of Cardiology, University Hospital of Larissa, Larissa 41110, Greece.
| | - Andrew Xanthopoulos
- Department of Cardiology, University Hospital of Larissa, Larissa 41110, Greece
| | - Stavros G Drakos
- University of Utah Health and School of Medicine and Salt Lake VA Medical Center, Salt Lake City, UT 84108, USA
| | | | - Alexandros Briasoulis
- Medical School of Athens, National and Kapodistrian University of Athens, Athens 15772, Greece
| | - John Skoularigis
- Department of Cardiology, University Hospital of Larissa, Larissa 41110, Greece
| | - Konstantinos Tsioufis
- First Department of Cardiology, Medical School, Hippokration Hospital, University of Athens, Athens 115 27, Greece
| | | | - Randall C Starling
- Department of Cardiovascular Medicine, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| |
Collapse
|
46
|
Cersosimo A, Salerno N, Sabatino J, Scatteia A, Bisaccia G, De Rosa S, Dellegrottaglie S, Bucciarelli-Ducci C, Torella D, Leo I. Underlying mechanisms and cardioprotective effects of SGLT2i and GLP-1Ra: insights from cardiovascular magnetic resonance. Cardiovasc Diabetol 2024; 23:94. [PMID: 38468245 PMCID: PMC10926589 DOI: 10.1186/s12933-024-02181-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 02/26/2024] [Indexed: 03/13/2024] Open
Abstract
Originally designed as anti-hyperglycemic drugs, Glucagon-Like Peptide-1 receptor agonists (GLP-1Ra) and Sodium-glucose cotransporter-2 inhibitors (SGLT2i) have demonstrated protective cardiovascular effects, with significant impact on cardiovascular morbidity and mortality. Despite several mechanisms have been proposed, the exact pathophysiology behind these effects is not yet fully understood. Cardiovascular imaging is key for the evaluation of diabetic patients, with an established role from the identification of early subclinical changes to long-term follow up and prognostic assessment. Among the different imaging modalities, CMR may have a key-role being the gold standard for volumes and function assessment and having the unique ability to provide tissue characterization. Novel techniques are also implementing the possibility to evaluate cardiac metabolism through CMR and thereby further increasing the potential role of the modality in this context. Aim of this paper is to provide a comprehensive review of changes in CMR parameters and novel CMR techniques applied in both pre-clinical and clinical studies evaluating the effects of SGLT2i and GLP-1Ra, and their potential role in better understanding the underlying CV mechanisms of these drugs.
Collapse
Affiliation(s)
- Angelica Cersosimo
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Nadia Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Jolanda Sabatino
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Alessandra Scatteia
- Advanced Cardiovascular Imaging Unit, Ospedale Medico-Chirurgico Accreditato Villa dei Fiori, Naples, Italy
| | - Giandomenico Bisaccia
- Department of Neuroscience, Imaging and Clinical Sciences, Institute for Advanced Biomedical Technologies "G. d'Annunzio", University of Chieti-Pescara, Chieti, Italy
| | - Salvatore De Rosa
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Santo Dellegrottaglie
- Advanced Cardiovascular Imaging Unit, Ospedale Medico-Chirurgico Accreditato Villa dei Fiori, Naples, Italy
| | - Chiara Bucciarelli-Ducci
- CMR Unit, Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, Kings College London, London, UK
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| | - Isabella Leo
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| |
Collapse
|
47
|
Henry JA, Couch LS, Rider OJ. Myocardial Metabolism in Heart Failure with Preserved Ejection Fraction. J Clin Med 2024; 13:1195. [PMID: 38592048 PMCID: PMC10931709 DOI: 10.3390/jcm13051195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/11/2024] [Accepted: 02/18/2024] [Indexed: 04/10/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is increasingly prevalent and now accounts for half of all heart failure cases. This rise is largely attributed to growing rates of obesity, hypertension, and diabetes. Despite its prevalence, the pathophysiological mechanisms of HFpEF are not fully understood. The heart, being the most energy-demanding organ, appears to have a compromised bioenergetic capacity in heart failure, affecting all phenotypes and aetiologies. While metabolic disturbances in heart failure with reduced ejection fraction (HFrEF) have been extensively studied, similar insights into HFpEF are limited. This review collates evidence from both animal and human studies, highlighting metabolic dysregulations associated with HFpEF and its risk factors, such as obesity, hypertension, and diabetes. We discuss how changes in substrate utilisation, oxidative phosphorylation, and energy transport contribute to HFpEF. By delving into these pathological shifts in myocardial energy production, we aim to reveal novel therapeutic opportunities. Potential strategies include modulating energy substrates, improving metabolic efficiency, and enhancing critical metabolic pathways. Understanding these aspects could be key to developing more effective treatments for HFpEF.
Collapse
Affiliation(s)
- John Aaron Henry
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK (O.J.R.)
- Department of Cardiology, Jersey General Hospital, Gloucester Street, St. Helier JE1 3QS, Jersey, UK
| | - Liam S. Couch
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK (O.J.R.)
| | - Oliver J. Rider
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK (O.J.R.)
| |
Collapse
|
48
|
Gao S, Liu XP, Li TT, Chen L, Feng YP, Wang YK, Yin YJ, Little PJ, Wu XQ, Xu SW, Jiang XD. Animal models of heart failure with preserved ejection fraction (HFpEF): from metabolic pathobiology to drug discovery. Acta Pharmacol Sin 2024; 45:23-35. [PMID: 37644131 PMCID: PMC10770177 DOI: 10.1038/s41401-023-01152-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 08/08/2023] [Indexed: 08/31/2023]
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) is currently a preeminent challenge for cardiovascular medicine. It has a poor prognosis, increasing mortality, and is escalating in prevalence worldwide. Despite accounting for over 50% of all HF patients, the mechanistic underpinnings driving HFpEF are poorly understood, thus impeding the discovery and development of mechanism-based therapies. HFpEF is a disease syndrome driven by diverse comorbidities, including hypertension, diabetes and obesity, pulmonary hypertension, aging, and atrial fibrillation. There is a lack of high-fidelity animal models that faithfully recapitulate the HFpEF phenotype, owing primarily to the disease heterogeneity, which has hampered our understanding of the complex pathophysiology of HFpEF. This review provides an updated overview of the currently available animal models of HFpEF and discusses their characteristics from the perspective of energy metabolism. Interventional strategies for efficiently utilizing energy substrates in preclinical HFpEF models are also discussed.
Collapse
Affiliation(s)
- Si Gao
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Xue-Ping Liu
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Ting-Ting Li
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Li Chen
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Yi-Ping Feng
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Yu-Kun Wang
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Yan-Jun Yin
- School of Pharmacy, Bengbu Medical College, Bengbu, 233000, China
| | - Peter J Little
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, 4102, Australia
| | - Xiao-Qian Wu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Suo-Wen Xu
- Department of Endocrinology, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| | - Xu-Dong Jiang
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China.
| |
Collapse
|
49
|
Barison A, Timoteo AT, Liga R, Borodzicz-Jazdzyk S, El Messaoudi S, Luong C, Mandoli GE, Moscatelli S, Ramkisoensing AA, Moharem-Elgamal S, Pontone G, Neglia D. Cardiovascular imaging research and innovation in 2023. EUROPEAN HEART JOURNAL. IMAGING METHODS AND PRACTICE 2024; 2:qyae029. [PMID: 39045198 PMCID: PMC11195771 DOI: 10.1093/ehjimp/qyae029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 03/28/2024] [Indexed: 07/25/2024]
Abstract
In 2023, cardiovascular imaging has made significant advancements, in terms of technology, pathophysiology, and clinical application. In this review, the most recent research findings in the field of cardiovascular imaging are discussed. Artificial intelligence and large population cohorts, together with several technical improvements, have had a crucial impact on the technological advancements of echocardiography, cardiovascular magnetic resonance, computed tomography (CT), and nuclear medicine. In the field of ischaemic heart disease, it has been demonstrated that appropriate non-invasive imaging strategies improve patients' management and reduce invasive procedures and the need for additional testing at follow-up. Moreover, improvements in plaque characterization with CT are an expanding field of research with relevant implications for the prediction of disease severity, evolution, and response to treatment. In the field of valvular heart disease, imaging techniques have advanced alongside improvements in transcatheter treatment for aortic stenosis, mitral, and tricuspid regurgitation. Finally, in the field of heart failure and cardiomyopathies, cardiovascular imaging has reinforced its crucial role in early diagnosis and risk evaluation, showcasing advanced techniques that outperform traditional methods in predicting adverse outcomes.
Collapse
Affiliation(s)
- Andrea Barison
- Cardiology and Cardiovascular Medicine Department, Fondazione Toscana Gabriele Monasterio, Via Moruzzi, 1, Pisa 56124, Italy
- Interdisciplinary Center for Health Science, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Ana Teresa Timoteo
- Cardiology Department, Santa Marta Hospital, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
- NOVA Medical School, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Riccardo Liga
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Italy
- Cardiology Division, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Sonia Borodzicz-Jazdzyk
- Department of Cardiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
- First Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Saloua El Messaoudi
- Department of Cardiology, Radboud university medical center, Nijmegen, The Netherlands
| | - Christina Luong
- Division of Cardiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Giulia Elena Mandoli
- Department of Medical Biotechnologies, Division of Cardiology, University of Siena, Siena, Italy
| | - Sara Moscatelli
- Inherited Cardiovascular Diseases, Great Ormond Street Hospital, Children NHS Foundation Trust, London, UK
- Institute of Cardiovascular Sciences, University College London, London, UK
| | | | | | - Gianluca Pontone
- Department of Perioperative Cardiology and Cardiovascular Imaging, Centro Cardiologico Monzino IRCCS, Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Danilo Neglia
- Cardiology and Cardiovascular Medicine Department, Fondazione Toscana Gabriele Monasterio, Via Moruzzi, 1, Pisa 56124, Italy
- Interdisciplinary Center for Health Science, Scuola Superiore Sant'Anna, Pisa, Italy
| |
Collapse
|
50
|
Wei J, Duan X, Chen J, Zhang D, Xu J, Zhuang J, Wang S. Metabolic adaptations in pressure overload hypertrophic heart. Heart Fail Rev 2024; 29:95-111. [PMID: 37768435 DOI: 10.1007/s10741-023-10353-y] [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] [Accepted: 09/19/2023] [Indexed: 09/29/2023]
Abstract
This review article offers a detailed examination of metabolic adaptations in pressure overload hypertrophic hearts, a condition that plays a pivotal role in the progression of heart failure with preserved ejection fraction (HFpEF) to heart failure with reduced ejection fraction (HFrEF). The paper delves into the complex interplay between various metabolic pathways, including glucose metabolism, fatty acid metabolism, branched-chain amino acid metabolism, and ketone body metabolism. In-depth insights into the shifts in substrate utilization, the role of different transporter proteins, and the potential impact of hypoxia-induced injuries are discussed. Furthermore, potential therapeutic targets and strategies that could minimize myocardial injury and promote cardiac recovery in the context of pressure overload hypertrophy (POH) are examined. This work aims to contribute to a better understanding of metabolic adaptations in POH, highlighting the need for further research on potential therapeutic applications.
Collapse
Affiliation(s)
- Jinfeng Wei
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Xuefei Duan
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jiaying Chen
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Dengwen Zhang
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jindong Xu
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jian Zhuang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.
| | - Sheng Wang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
- Linzhi People's Hospital, Linzhi, Tibet, China.
| |
Collapse
|