1
|
Gao Z, Qian XH, Ke JY, Lin K, Zhu JH, Zhou X, Zhou H, Wang LG. Angiography-derived index of microcirculatory resistance as a novel tool to assess coronary microvascular dysfunction in patients with diabetic cardiomyopathy. Int J Cardiol 2025; 431:133220. [PMID: 40188960 DOI: 10.1016/j.ijcard.2025.133220] [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: 10/01/2024] [Revised: 03/05/2025] [Accepted: 03/28/2025] [Indexed: 04/10/2025]
Abstract
BACKGROUNDS Coronary microvascular dysfunction (CMD) has been proposed as a major mechanism and a potential therapeutic target for diabetic cardiomyopathy (DbCM); however, it has not been fully investigated in a clinical setting. The angiography-derived index of microcirculatory resistance (AMR) is a novel non-invasive measurement of CMD that exhibits promising clinical applications. METHODS AMR was measured in hospitalized patients with DbCM and in control patients. The incidence, clinical characteristics, risk factors, and effects of pharmacological interventions on CMD were investigated. RESULTS AMR was significantly higher in patients who met the DbCM-2B diagnostic criteria. The independent risk factors for abnormal AMR included diabetes, body mass index (BMI), and N-terminal pro-brain natriuretic peptide (NT-pro-BNP). Patients with elevated NT-pro-BNP had high AMR, and those grouped by medication indicated that ACEI/ARB, sacubitril/valsartan, and trimetazidine might lower AMR in patients with elevated NT-pro-BNP. CONCLUSIONS The DbCM-2B diagnostic criteria demonstrated a strong correlation with CMD. ACEI/ARB, sacubitril/valsartan, and trimetazidine might improve CMD in patients with DbCM.
Collapse
Affiliation(s)
- Zhan Gao
- The Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Xue Hua Qian
- The Department of Information, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Jia Yu Ke
- The Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Ken Lin
- The Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Jian Han Zhu
- The Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Xi Zhou
- The Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Hao Zhou
- The Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Liang Guo Wang
- The Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China.
| |
Collapse
|
2
|
Kessler EL, Dal Canto E, Diez-Benavente E, van Ommen AM, Kapteijn D, Glade MC, Bekkering S, Haitjema S, Valstar G, Cramer MJ, Rutten FH, Teske AJ, Menken R, Hofstra L, den Ruijter HM, Riksen NP, de Jager SCA. Non-classical monocytes are associated with functional markers of left ventricular diastolic dysfunction and heart failure with preserved ejection fraction. Int J Cardiol 2025; 429:133161. [PMID: 40088950 DOI: 10.1016/j.ijcard.2025.133161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/13/2025] [Accepted: 03/12/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND Inflammatory conditions such as obesity and diabetes are linked to intermediate/non-classical monocyte activation, contributing to left ventricular diastolic dysfunction (LVDD) and heart failure with preserved ejection fraction (HFpEF). OBJECTIVE To investigate whether circulating monocyte subtypes and their activity are associated with the presence LVDD and HFpEF. METHODS We analyzed peripheral blood mononuclear cells (PBMCs) from 73 patients with or without LVDD/HFpEF. Cytokine secretion was measured post-stimulation, and gene expression was assessed via RNA sequencing. Monocyte subtypes were characterized using flow cytometry, and macrophage polarization was evaluated. We also examined the relationship between immunological markers and echocardiographic indicators of LVDD. RESULTS Among the participants, 24 were controls, 23 had LVDD, and 26 had HFpEF. PBMCs from LVDD patients secreted significantly less Interleukin-6 compared to controls (2053 ± 708 pg/mL vs 12,273 ± 3357 pg/mL, p = 0.008). RNA sequencing indicated increased estrogen receptor pathway activity in LVDD. HFpEF patients exhibited a 1.5-fold increase in intermediate monocytes and a significant rise in non-classical monocytes compared to controls. Stimulated macrophages from LVDD and HFpEF patients showed less CD206/CD80 expression, indicating a shift towards M2-macrophage polarization. Notably, higher non-classical monocyte counts correlated with lower E' septal velocity, suggesting an association with diastolic impairment (β = -0.15, p = 0.041). CONCLUSIONS LVDD and HFpEF are associated with a shift towards non-classical monocyte subtypes and higher numbers of non-classical monocytes are associated with signs of diastolic impairment. These findings highlight the importance of the inflammatory component in LVDD and HFpEF.
Collapse
Affiliation(s)
- Elise L Kessler
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands; Laboratory for Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University, Utrecht, the Netherlands
| | - Elisa Dal Canto
- Laboratory for Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands; Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Ernest Diez-Benavente
- Laboratory for Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Anne-Mar van Ommen
- Laboratory for Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Daniek Kapteijn
- Laboratory for Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marleen C Glade
- Laboratory for Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Siroon Bekkering
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Saskia Haitjema
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Gideon Valstar
- Laboratory for Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Maarten J Cramer
- Clinical Cardiology Department, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Frans H Rutten
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Arco J Teske
- Clinical Cardiology Department, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Roxana Menken
- Cardiology Centers of the Netherlands, the Netherlands
| | | | - Hester M den Ruijter
- Laboratory for Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Saskia C A de Jager
- Laboratory for Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University, Utrecht, the Netherlands.
| |
Collapse
|
3
|
Narimani-Javid R, Mahalleh M, Behboodi K, Izadpanahi K, Arzhangzadeh A, Nikfar R, Hosseini SA, Amini-Salehi E, Shafiei S, Vahidi H, Hosseini K, Soleimani H. Prognostic significance of right ventricular dysfunction in heart failure with preserved ejection fraction: a meta-analysis of reconstructed time-to-event data. Echo Res Pract 2025; 12:13. [PMID: 40437644 PMCID: PMC12121155 DOI: 10.1186/s44156-025-00080-5] [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: 01/22/2025] [Accepted: 05/12/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND The prognosis of Heart failure with preserved ejection fraction (HFpEF) is significantly impacted by the existence and severity of comorbidities. Recent studies highlight the right ventricle (RV) as a crucial player in heart failure pathophysiology. However, there are still gaps in understanding how right ventricular dysfunction (RVD) affects long-term outcomes in patients with heart failure with preserved ejection fraction (HFpEF). MATERIALS AND METHODS In this systematic review and meta-analysis, a comprehensive search was conducted to identify studies investigating RVD as the predictor of the composite outcome of All-cause death, cardiac death, and hospitalization for HF in patients with HFpEF published until October 2024. RVD was defined as the deviation of at least one measurement of RV function from the recommended normal range based on modality and the normal ranges established in each study. Time and survival probability were extracted for each Group (HFpEF patients with and without RVD) in each of the Kaplan-Meier curves. Individual patient data were reconstructed by processing the extracted time points, survival probabilities, and the number of patients at risk in a two-stage approach. The restricted mean survival time (RMST) was also calculated as the area under the survival curve for each group. RESULTS Seven studies met the inclusion criteria, comprising 1936 individuals, of which 555 patients had RVD. The pooled prevalence of RVD among HFpEF was 41.2% (95% CI: 36.5; 45.9). Patients with RVD had a significantly higher risk of adverse outcomes compared to those without RVD, with an HR of 2.28 (95% CI, 1.95; 2.68, p-value < 0.001) in the eight-year follow-up after the RVD diagnosis. The one-year landmark analysis revealed that the majority of the event-free survival disparity between patients with RVD and those without arises from the first year after an RVD diagnosis. Patients with RVD also had shorter event-free survival. (ΔRMST = -2.127 years, 95% CI, -2.383; -1.872, p-value < 0.001). CONCLUSION The development of RVD in HFpEF is linked to significantly increased composite outcomes of all-cause death and HF hospitalization and shorter event-free survival.
Collapse
Affiliation(s)
- Roozbeh Narimani-Javid
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Mahalleh
- Cardiovascular Diseases Research Institute, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Kiyarash Behboodi
- Cardiovascular Diseases Research Institute, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kasra Izadpanahi
- Cardiovascular Diseases Research Institute, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Reza Nikfar
- Cardiovascular Diseases Research Institute, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Hosseini
- School of Medicine, Babol University of Medical Sciences, Mazandaran, Iran
| | - Ehsan Amini-Salehi
- Department of Internal Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Sasan Shafiei
- Department of Cardiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamed Vahidi
- Department of Cardiology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Kaveh Hosseini
- Cardiovascular Diseases Research Institute, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamidreza Soleimani
- Department of Cardiology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Uchmanowicz I, Lisiak M, Ponikowski P, Jankowska EA, Mysiak A, Wleklik M. A two-phase approach to identifying HFpEF in heart failure patients: Risk score evaluation and decision tree development. ESC Heart Fail 2025. [PMID: 40433849 DOI: 10.1002/ehf2.15333] [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: 01/09/2025] [Revised: 04/19/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
AIMS Heart failure (HF) with preserved ejection fraction (HFpEF) poses significant diagnostic challenges due to its complex aetiology and overlapping symptoms with other HF types. The heterogeneity of HFpEF, compounded by frequent comorbidities, complicates diagnosis. This study aimed to enhance HFpEF prediction through a two-phase approach: a simplified risk score and a decision tree model. METHODS AND RESULTS In Phase 1, an 8-point risk score based on accessible clinical parameters was developed. In Phase 2, we conducted comprehensive predictive modelling using decision tree analysis. Data from 560 HF patients were analysed. It achieved an accuracy of 63.13% (sensitivity: 62.87%, specificity: 54.24%). In Phase 2, a decision tree model using broader clinical variables improved accuracy to 73.04% (sensitivity: 53.89%, specificity: 81.17%). CONCLUSIONS This dual framework provides tools for both quick screening and detailed risk stratification in various clinical settings.
Collapse
Affiliation(s)
- Izabella Uchmanowicz
- Department of Nursing, Division of Research Methodology, Faculty of Nursing and Midwifery, Wroclaw Medical University, Wroclaw, Poland
- Centre for Cardiovascular Health, Edinburgh Napier University, Edinburgh, UK
| | - Magdalena Lisiak
- Department of Nursing, Division of Research Methodology, Faculty of Nursing and Midwifery, Wroclaw Medical University, Wroclaw, Poland
| | - Piotr Ponikowski
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Ewa A Jankowska
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Andrzej Mysiak
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Marta Wleklik
- Department of Nursing, Division of Research Methodology, Faculty of Nursing and Midwifery, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
5
|
Yoo TT, Baek IH, Stoletniy L, Hilliard A, Sakr A, Doycheva D. Impact of sodium-glucose transport protein-2 (SGLT2) inhibitors on the inflammasome pathway in acute myocardial infarction in type 2 diabetes mellitus: a comprehensive review. Cardiovasc Diabetol 2025; 24:227. [PMID: 40420176 PMCID: PMC12105141 DOI: 10.1186/s12933-025-02777-7] [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: 03/06/2025] [Accepted: 05/06/2025] [Indexed: 05/28/2025] Open
Abstract
Sodium-glucose transport protein-2 (SGLT2) inhibitors, initially developed for glycemic control in type 2 diabetes mellitus (T2DM), have emerged as potential cardioprotective agents, reducing cardiovascular mortality and improving heart failure outcomes. Recent evidence suggests that SGLT2 inhibitors exert anti-inflammatory effects, particularly through modulating the inflammasome pathway. This review explores the role of the inflammasome in acute myocardial infarction (AMI) in T2DM and discusses the mechanisms by which SGLT2 inhibitors influence this pathway. We evaluate current studies on the impact of SGLT2 inhibitors on key inflammatory mediators, particularly the NLRP3 inflammasome, and discuss their potential therapeutic implications for reducing inflammation and myocardial injury in patients with T2DM experiencing AMI. In summary, the key novelties in this review lie in its focused mechanistic approach on the inflammasome pathway, its integration of diabetes and cardiovascular research, and its potential to influence future therapeutic strategies for AMI in T2DM patients. It offers a novel angle by tying together molecular mechanisms of inflammation with clinical implications in a specific patient population that faces high cardiovascular risk.
Collapse
Affiliation(s)
- Thomas T Yoo
- Department of Internal Medicine, Loma Linda University Medical Center, Loma Linda, CA, 92354, USA
| | - In Hae Baek
- Department of Internal Medicine, Loma Linda University Medical Center, Loma Linda, CA, 92354, USA
| | - Liset Stoletniy
- Division of Cardiology, School of Medicine, Loma Linda University, 11234 Anderson St, Loma Linda, CA, 92354, USA
- Department of Internal Medicine, Loma Linda University Medical Center, Loma Linda, CA, 92354, USA
| | - Anthony Hilliard
- Division of Cardiology, School of Medicine, Loma Linda University, 11234 Anderson St, Loma Linda, CA, 92354, USA
- Department of Internal Medicine, Loma Linda University Medical Center, Loma Linda, CA, 92354, USA
| | - Antoine Sakr
- Division of Cardiology, School of Medicine, Loma Linda University, 11234 Anderson St, Loma Linda, CA, 92354, USA
- Department of Internal Medicine, Loma Linda University Medical Center, Loma Linda, CA, 92354, USA
| | - Desislava Doycheva
- Division of Cardiology, School of Medicine, Loma Linda University, 11234 Anderson St, Loma Linda, CA, 92354, USA.
- Department of Physiology and Pharmacology, Loma Linda University, 11175 Campus St, Loma Linda, CA, 92354, USA.
| |
Collapse
|
6
|
Huang D, Zou C, Wu H. Relationship between the ratio of erythrocyte distribution width to albumin level and mortality in hypertensive population: Mediating role of inflammatory markers. PLoS One 2025; 20:e0324027. [PMID: 40408377 PMCID: PMC12101704 DOI: 10.1371/journal.pone.0324027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 04/18/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND This study examined the ratio of erythrocyte distribution width (RDW) to albumin concentration (RAR) and all-cause, cardiovascular disease (CVD), and cancer mortality in the hypertension population, focusing on the role of inflammatory markers as mediators. PATIENTS AND METHODS Data from NHANES (1999-2018) were analyzed, linking National Death Index (NDI) records to mortality outcomes through December 31, 2019. A weighted sampling design categorized participants into three RAR groups. Cox regression models adjusted for demographic and clinical variables assessed the association between RAR and mortality outcomes. Mediation analyses explored the mediating role of the systemic Inflammatory response index (SIRI) and neutrophil-to-lymphocyte ratio (NLR). RESULTS Among 26,935 participants with a median follow-up of 102 months and 6,007 deaths, elevated RAR was associated with increased risks of all-cause mortality (HR = 1.83, 95% CI: 1.76-1.90), cardiovascular disease mortality (HR = 1.81, 95% CI: 1.68-1.95), and cancer mortality (HR = 1.70, 95% CI: 1.55-1.86). Segmented regression showed a nonlinear relationship between RAR and all-cause mortality, cardiovascular mortality, and cancer mortality, and the threshold effect results showed a fold value of 4.10, with a greater HR when RAR < 4.10. Mediation analysis revealed that SIRI and NLR mediated the relationship between RAR and all-cause mortality by 8.12% and 6.00%, respectively. CONCLUSION In hypertensive populations, higher RAR values are associated with increased all-cause mortality, cardiovascular mortality, and cancer mortality. Inflammation partially mediates the relationship between RAR and all-cause mortality.
Collapse
Affiliation(s)
- Dongli Huang
- Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Chun Zou
- Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Hang Wu
- Bishan Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
7
|
Lee CJM, Kosyakovsky LB, Khan MS, Wu F, Chen G, Hill JA, Ho JE, Foo RSY, Zannad F. Cardiovascular, Kidney, Liver, and Metabolic Interactions in Heart Failure: Breaking Down Silos. Circ Res 2025; 136:1170-1207. [PMID: 40403106 PMCID: PMC12125648 DOI: 10.1161/circresaha.125.325602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/30/2025] [Accepted: 04/07/2025] [Indexed: 05/24/2025]
Abstract
Over the past few decades, the rising burden of metabolic disease, including type 2 diabetes, prediabetes, obesity, and metabolic dysfunction-associated steatotic liver disease, has corresponded with fundamental shifts in the landscape of heart failure (HF) epidemiology, including the rising prevalence of HF with preserved ejection fraction. It has become increasingly important to understand the role of extracardiac contributors and interorgan communication in the pathophysiology and phenotypic heterogeneity of HF. Whereas traditional epidemiological strategies have separately examined individual contributions of specific comorbidities to HF risk, these approaches may not capture the shared mechanisms and more complex, bidirectional relationships between cardiac and noncardiac comorbidities. In this review, we highlight the cardiac, kidney, liver, and metabolism multiorgan interactions and pathways that complicate HF development and progression and propose research strategies to further understand HF in the context of multiple organ disease. This includes evolving epidemiological approaches such as multiomics and machine learning which may better capture common underlying mechanisms and interorgan crosstalk. We review existing preclinical models of HF and how they have enhanced our understanding of the role of multiorgan disease in the development of HF subtypes. We suggest recommendations as to how clinical practice across multiple specialties should screen for and manage multiorgan involvement in HF. Finally, recognizing the advent of novel combinatorial therapeutic agents that may have multiple indications across the cardiac-kidney-liver metabolism continuum, we review the current clinical trials landscape. We specifically highlight a pressing need for the design of more inclusive trials that examine the contributions of multimorbidity and incorporate multiorgan end points, which we propose may lead to outcomes that are evermore clinically relevant today.
Collapse
Affiliation(s)
- Chang Jie Mick Lee
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health System, Centre for Translational Medicine, 14 Medical Drive, Singapore 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research (A*STaR), 61 Biopolis Drive, Singapore 138673, Singapore
| | - Leah B. Kosyakovsky
- Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Muhammad Shahzeb Khan
- Baylor Scott and White Research Institute, Dallas, TX, USA
- The Heart Hospital, Plano, TX, USA
| | - Feng Wu
- Division of Cardiology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Guo Chen
- Division of Cardiology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Joseph A. Hill
- Division of Cardiology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Jennifer E. Ho
- Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Roger S-Y. Foo
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health System, Centre for Translational Medicine, 14 Medical Drive, Singapore 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research (A*STaR), 61 Biopolis Drive, Singapore 138673, Singapore
| | - Faiez Zannad
- Université de Lorraine, CHRU, Inserm Clinical Investigation Center 1433, Nancy, France
| |
Collapse
|
8
|
Capone F, Vacca A, Bidault G, Sarver D, Kaminska D, Strocchi S, Vidal-Puig A, Greco CM, Lusis AJ, Schiattarella GG. Decoding the Liver-Heart Axis in Cardiometabolic Diseases. Circ Res 2025; 136:1335-1362. [PMID: 40403112 DOI: 10.1161/circresaha.125.325492] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Abstract
The liver and heart are closely interconnected organs, and their bidirectional interaction plays a central role in cardiometabolic disease. In this review, we summarize current evidence linking liver dysfunction-particularly metabolic dysfunction-associated steatotic liver disease, alcohol-associated liver disease, and cirrhosis-with an increased risk of heart failure and other cardiovascular diseases. We discuss how these liver conditions contribute to cardiac remodeling, systemic inflammation, and hemodynamic stress and how cardiac dysfunction in turn impairs liver perfusion and promotes hepatic injury. Particular attention is given to the molecular mediators of liver-heart communication, including hepatokines and cardiokines, as well as the emerging role of advanced research methodologies, including omics integration, proximity labeling, and organ-on-chip platforms, that are redefining our understanding of interorgan cross talk. By integrating mechanistic insights with translational tools, this review aims to support the development of multiorgan therapeutic strategies for cardiometabolic disease.
Collapse
Affiliation(s)
- Federico Capone
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (F.C., A.V., S.S., G.G.S.)
- Department of Medicine, Unit of Internal Medicine III, Padua University Hospital, University of Padua, Padova, Italy (F.C.)
- Department of Biomedical Sciences, University of Padova, Italy (F.C.)
| | - Antonio Vacca
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (F.C., A.V., S.S., G.G.S.)
- Clinica Medica, Department of Medicine, University of Udine, Italy (A.V.)
| | - Guillaume Bidault
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, United Kingdom (G.B., A.V.-P.)
| | - Dylan Sarver
- Division of Cardiology, Department of Medicine (D.S., D.K., A.J.L.), University of California, Los Angeles
- Department of Microbiology, Immunology and Molecular Genetics (D.S., A.J.L.), University of California, Los Angeles
- Department of Human Genetics (D.S., A.J.L.), University of California, Los Angeles
| | - Dorota Kaminska
- Division of Cardiology, Department of Medicine (D.S., D.K., A.J.L.), University of California, Los Angeles
| | - Stefano Strocchi
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (F.C., A.V., S.S., G.G.S.)
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Deutsches Herzzentrum der Charité, Charité-Universitätsmedizin Berlin, Germany (S.S., G.G.S.)
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, United Kingdom (G.B., A.V.-P.)
- Centro de Investigacion Principe Felipe, Valencia, Spain (A.V.-P.)
| | - Carolina M Greco
- Department of Biomedical Sciences, Humanitas University, Milan, Italy (C.M.G.)
- IRCCS Humanitas Research Hospital, Milan, Italy (C.M.G.)
| | - Aldons J Lusis
- Division of Cardiology, Department of Medicine (D.S., D.K., A.J.L.), University of California, Los Angeles
- Department of Microbiology, Immunology and Molecular Genetics (D.S., A.J.L.), University of California, Los Angeles
- Department of Human Genetics (D.S., A.J.L.), University of California, Los Angeles
| | - Gabriele G Schiattarella
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (F.C., A.V., S.S., G.G.S.)
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Deutsches Herzzentrum der Charité, Charité-Universitätsmedizin Berlin, Germany (S.S., G.G.S.)
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany (G.G.S.)
- Friede Springer Cardiovascular Prevention Center at Charité-Universitätsmedizin Berlin, Germany (G.G.S.)
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (G.G.S.)
| |
Collapse
|
9
|
Mohanta SK, Heron C, Klaus-Bergmann A, Horstmann H, Brakenhielm E, Giannarelli C, Habenicht AJR, Gerhardt H, Weber C. Metabolic and Immune Crosstalk in Cardiovascular Disease. Circ Res 2025; 136:1433-1453. [PMID: 40403115 DOI: 10.1161/circresaha.125.325496] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Abstract
Cardiovascular diseases including atherosclerosis and heart failure, arise from the intricate interplay of metabolic, immune, and neural dysregulation within vascular and cardiac tissues: This review focuses on integrating recent advances in metabolic and immune crosstalk of the cardiac vasculature that affects cardiometabolic health and disease progression. Coronary and lymphatic endothelial cells regulate cardiac metabolism, and their dysfunction is linked to cardiovascular diseases. Lymphatics maintain tissue homeostasis, including clearing metabolic waste, lipids, and immune cells, and their maladaptation in metabolic diseases worsens outcomes. Altered vascular endothelial metabolism in heart failure drives immune-mediated inflammation, fibrosis, and adverse cardiac remodeling. Concurrently, artery tertiary lymphoid organs formed in the adventitia of advanced atherosclerotic arteries, serve as pivotal neuroimmune hubs, coordinating local immunity through T and B cell activation and neurovascular signaling via artery-brain circuits. T cells within plaques and artery tertiary lymphoid organs undergo clonal expansion as a result of peripheral tolerance breakdown, with proinflammatory CD4+ and CD8+ subsets amplifying atherosclerosis, effects further shaped by systemic immune activation. Therapeutic strategies targeting endothelial cell metabolism, lymphatic dysfunction, neuroimmune crosstalk, and T cell plasticity hold promise for integrated cardiovascular disease management.
Collapse
Affiliation(s)
- Sarajo K Mohanta
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU), LMU University Hospital, Munich, Germany (S.K.M., A.J.R.H., C.W.)
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany (S.K.M., A.J.R.H., C.W.)
| | - Coraline Heron
- UnivRouen Normandie, INSERM EnVI, UMR 1096, Rouen, France (C.H., E.B.)
| | - Alexandra Klaus-Bergmann
- Integrative Vascular Biology Laboratory, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (A.K.-B., H.G.)
- German Centre for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany (A.K.-B., H.G.)
| | - Hauke Horstmann
- Cardiology and Angiology, Medical Center (H.H.), University of Freiburg, Freiburg, Germany
- Faculty of Medicine (H.H.), University of Freiburg, Freiburg, Germany
- Department of Medicine, Division of Cardiology (H.H., C.G.), NYU Grossman School of Medicine, New York, NY
| | - Ebba Brakenhielm
- UnivRouen Normandie, INSERM EnVI, UMR 1096, Rouen, France (C.H., E.B.)
| | - Chiara Giannarelli
- Department of Medicine, Division of Cardiology (H.H., C.G.), NYU Grossman School of Medicine, New York, NY
- Department of Pathology (C.G.), NYU Grossman School of Medicine, New York, NY
| | - Andreas J R Habenicht
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU), LMU University Hospital, Munich, Germany (S.K.M., A.J.R.H., C.W.)
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany (S.K.M., A.J.R.H., C.W.)
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China (A.J.R.H.)
| | - Holger Gerhardt
- Integrative Vascular Biology Laboratory, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (A.K.-B., H.G.)
- German Centre for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany (A.K.-B., H.G.)
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU), LMU University Hospital, Munich, Germany (S.K.M., A.J.R.H., C.W.)
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany (S.K.M., A.J.R.H., C.W.)
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands (C.W.)
| |
Collapse
|
10
|
Fließer E, Jandl K, Chen SH, Wang MT, Schupp JC, Kuebler WM, Baker AH, Kwapiszewska G. Transcriptional signatures of endothelial cells shape immune responses in cardiopulmonary health and disease. JCI Insight 2025; 10:e191059. [PMID: 40401523 DOI: 10.1172/jci.insight.191059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025] Open
Abstract
The cardiopulmonary vasculature and its associated endothelial cells (ECs) play an essential role in sustaining life by ensuring the delivery of oxygen and nutrients. Beyond these foundational functions, ECs serve as key regulators of immune responses. Recent advances in single-cell RNA sequencing have revealed that the cardiopulmonary vasculature is composed of diverse EC subpopulations, some of which exhibit specialized immunomodulatory properties. Evidence for immunomodulation includes distinct expression profiles associated with antigen presentation, cytokine secretion, immune cell recruitment, translocation, and clearance - functions critical for maintaining homeostasis in the heart and lungs. In cardiopulmonary diseases, ECs undergo substantial transcriptional reprogramming, leading to a shift from homeostasis to an activated state marked by heightened immunomodulatory activity. This transformation has highlighted the critical role for ECs in disease pathogenesis and their potential as future therapy targets. This Review emphasizes the diverse functions of ECs in the heart and lungs, particularly adaptive and maladaptive immunoregulatory roles in cardiopulmonary health and disease.
Collapse
Affiliation(s)
- Elisabeth Fließer
- Otto Loewi Research Center, Lung Research Cluster, Medical University of Graz, Graz, Austria
- Institute for Lung Health, Cardiopulmonary Institute, Member of German Lung Center, Justus-Liebig University, Giessen, Germany
| | - Katharina Jandl
- Otto Loewi Research Center, Lung Research Cluster, Medical University of Graz, Graz, Austria
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Shiau-Haln Chen
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Mei-Tzu Wang
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
- Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jonas C Schupp
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pulmonary and Infectious Diseases, Hannover Medical School, Hannover, Germany
- Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), German Center for Lung Research BREATH, Hannover, Germany
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin, Berlin, Germany
- German Center for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
- German Center for Lung Research, Associated Partner Site Berlin, Berlin, Germany
- Department of Surgery and
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Centre, St Michael's Hospital, Toronto, Ontario, Canada
| | - Andrew H Baker
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
- Department of Pathology, Cardiovascular Research Institute Maastricht, School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
| | - Grazyna Kwapiszewska
- Otto Loewi Research Center, Lung Research Cluster, Medical University of Graz, Graz, Austria
- Institute for Lung Health, Cardiopulmonary Institute, Member of German Lung Center, Justus-Liebig University, Giessen, Germany
| |
Collapse
|
11
|
Siddiqui HF, Waqas SA, Batool RM, Salim H, Minhas AMK, Hasni SF, Alsaid A, Sannino A, Afzal AM, Khan MS. The effect of GLP-1 receptor agonists on cardiac remodeling in heart failure patients with preserved and reduced ejection fraction: a systematic review and meta-analysis. Heart Fail Rev 2025:10.1007/s10741-025-10523-0. [PMID: 40399552 DOI: 10.1007/s10741-025-10523-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/30/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Glucagon-like peptide-1 receptor agonists (GLP-1RA) have shown promising effects on heart failure (HF) outcomes, particularly in phenotype-specific populations. However, their impact on cardiac structure and function in HF with preserved ejection fraction (HFpEF) and reduced ejection fraction (HFrEF) remains unclear. METHODS Medline, Cochrane Library, and Scopus were queried through December 2024 for primary and secondary analyses of randomized controlled trials comparing GLP-1RA with placebo in HF patients. Outcomes included changes in left ventricular ejection fraction (LVEF), end-diastolic volume (LVEDV), end-systolic volume (LVESV), global longitudinal strain (GLS), left ventricular mass, left atrial volume (LAV), and NT-proBNP levels. Random-effects models were used to calculate weighted mean differences (WMDs) or hazard ratios (HRs). RESULTS Six trials (n = 1,195) were included, with three each evaluating HFpEF and HFrEF populations. In patients with HFpEF, GLP-1RA significantly reduced the LV mass (WMD: -8.6 g; 95% CI: -14.6, -2.6; p = 0.005) and LAV (WMD: -5.4 ml; 95% CI: -8.8, -2.0; p = 0.002) and lowered NT-proBNP concentration throughout (HR: 0.85; 95% CI: 0.8, 0.9; p < 0.001). A decrease in LAV was observed in the HFrEF population (WMD: -5.4 ml [95% CI: -8.8, -2.0]; p = 0.002). However, no significant improvements were observed in LVEF, LVEDV, LVESV, or GLS. There were significant differences between HFpEF and HFrEF for LVEDV (p = 0.01) and LVESV (p = 0.04). CONCLUSIONS GLP-1RA demonstrated phenotype-specific benefits, improving structural remodeling in HFpEF but showing limited effects in HFrEF. These findings highlight the importance of targeted therapeutic strategies based on HF phenotypes. Further research is warranted to elucidate underlying mechanisms and optimize patient selection.
Collapse
Affiliation(s)
| | - Saad Ahmed Waqas
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | | | - Hussain Salim
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | | | | | - Amro Alsaid
- Baylor Scott and White Heart Hospital, Plano, TX, USA
| | - Anna Sannino
- Baylor Scott and White Research Institute Cardiac Imaging Core Laboratory, Plano, TX, USA
| | - Aasim M Afzal
- Baylor Scott and White Heart Hospital, Plano, TX, USA
| | - Muhammad Shahzeb Khan
- Baylor Scott and White Heart Hospital, Plano, TX, USA.
- Baylor Scott and White Research Institute Cardiac Imaging Core Laboratory, Plano, TX, USA.
- Baylor College of Medicine, Temple, TX, USA.
- Baylor Scott and White Research Institute, Baylor Scott and White Health, Dallas, TX, USA.
| |
Collapse
|
12
|
Velmeden D, Söhne J, Schuch A, Zeid S, Schulz A, Troebs SO, Müller F, Heidorn MW, Buch G, Belanger N, Dinh W, Mondritzki T, Lackner KJ, Gori T, Münzel T, Wild PS, Prochaska JH. Role of Heart Rate Recovery in Chronic Heart Failure: Results From the MyoVasc Study. J Am Heart Assoc 2025; 14:e039792. [PMID: 40371587 DOI: 10.1161/jaha.124.039792] [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: 10/31/2024] [Accepted: 03/11/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Cardiac autonomic dysfunction is associated with heart failure (HF). Reduced heart rate recovery (HRR) indicates impaired parasympathetic reactivation after physical activity. Heart rate recovery 60 seconds after peak effort (HRR60) is linked to autonomic dysfunction, but data on its relevance across HF phenotypes are scarce. This study aimed to identify clinical determinants of HRR60 in an HF cohort and assess its relationship with clinical outcomes. METHODS Data from the MyoVasc study (NCT04064450; N=3289) were analyzed. Participants underwent standardized clinical phenotyping including cardiopulmonary exercise testing. HRR60 was defined as the heart rate decline 60 seconds after exercise termination. Clinical determinants of HRR60 were evaluated using multivariate regression, whereas Cox regression analyses assessed all-cause death and worsening of HF. RESULTS The analysis sample comprised 1289 individuals (median age, 66.0 [interquartile range {IQR}, 58.0-73.0] years, 30.4% women) ranging from stage B to stage C/D according to the universal definition of HF. Age, sex, smoking, obesity, peripheral artery disease, and chronic kidney disease were identified as determinants of HRR60. HRR60 showed a strong association with all-cause death (hazard ratio [HR]HRR60 [10 bpm], 1.56 [95% CI, 1.32-1.85]; P<0.0001) and worsening of HF (HRHRR60 [10 bpm], 1.36 [95% CI, 1.10-1.69]; P=0.0052) independent of age, sex, and clinical profile. Sensitivity analysis showed a stronger association with worsening HF in HF with preserved left ventricular ejection fraction (Pinteraction=0.027). CONCLUSIONS HRR60 was associated with clinical outcome in chronic HF. Because it showed a stronger association with outcomes in HF with preserved ejection fraction, future research should consider phenotype-specific differences.
Collapse
Affiliation(s)
- David Velmeden
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
- Department of Cardiology - Cardiology I University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
| | - Jakob Söhne
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
- Department of Cardiology - Cardiology I University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
| | - Alexander Schuch
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
- Department of Cardiology - Cardiology I University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
| | - Silav Zeid
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
| | - Andreas Schulz
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
| | - Sven-Oliver Troebs
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
| | - Felix Müller
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
- Department of Cardiology - Cardiology I University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
| | - Marc W Heidorn
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
- Department of Cardiology - Cardiology I University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
| | - Gregor Buch
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI) University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
| | - Noémie Belanger
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
| | - Wilfried Dinh
- Bayer AG, Research and Development, Translational Clinical Medicine, Experimental Medicine 1 Wuppertal Germany
- School of Medicine University Witten/Herdecke Witten Germany
| | - Thomas Mondritzki
- Research & Early Development, Clinical Experimentation CV, BAYER AG Wuppertal Germany
| | - Karl J Lackner
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
- Institute for Clinical Chemistry and Laboratory Medicine University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
| | - Tommaso Gori
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
- Department of Cardiology - Cardiology I University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
| | - Thomas Münzel
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
- Department of Cardiology - Cardiology I University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
| | - Philipp S Wild
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
- Clinical Epidemiology and Systems Medicine, Center for Thrombosis and Hemostasis (CTH) University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- Institute for Molecular Biology (IMB), Mainz, Working Group Systems Medicine Mainz Germany
| | - Jürgen H Prochaska
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
- Clinical Epidemiology and Systems Medicine, Center for Thrombosis and Hemostasis (CTH) University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
| |
Collapse
|
13
|
Mu Y, Geng J, Liu C, Jiang S, Han Y, Jiang J, Wang Y. Exploring the Multi-Faceted Effects of Berberine in Ameliorating Diastolic Dysfunction in Rats with Heart Failure with Preserved Ejection Fraction. Int J Mol Sci 2025; 26:4847. [PMID: 40429987 PMCID: PMC12112712 DOI: 10.3390/ijms26104847] [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: 03/11/2025] [Revised: 05/01/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF), marked by cardiac diastolic dysfunction, contributes to half of all heart failure cases globally and poses a significant public health challenge. Effective therapies for HFpEF are rare, largely due to its complex and heterogeneous pathophysiology, which often involves multiple comorbidities. Berberine (BBR), an isoquinoline alkaloid, has demonstrated beneficial effects on multiple metabolic and cardiovascular disorders; however, its impact on cardiac diastolic dysfunction in HFpEF remains poorly understood. In this study, we utilized a rat model of HFpEF induced by a sustained high-fat/high-sucrose (HFHS) diet to explore the impact and mechanisms of BBR on diastolic dysfunction. The results revealed that BBR administration effectively alleviated cardiac diastolic dysfunction and alleviated extracardiac comorbidities, including increased weight, impaired glucose tolerance, hypercholesterolemia and hypertension, in rats fed an HFHS diet. Furthermore, BBR mitigated myocardial inflammation, oxidative stress, microvascular endothelial dysfunction, and notably restored the disturbed NO-cGMP-PKG pathway. Additionally, BBR reduced myocardial fibrosis and inhibited the abnormally activated TGF-β/Smads signaling. Moreover, BBR attenuated the systemic inflammation and corrected immune dysregulation in an HFHS diet-fed rats. Our study suggests that BBR exhibits multi-beneficial effects in the prevention and management of HFpEF, demonstrating its potential as a holistic therapeutic candidate for HFpEF.
Collapse
Affiliation(s)
- Yu Mu
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jing Geng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Chilu Liu
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shuang Jiang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yanxing Han
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jiandong Jiang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yuhong Wang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
14
|
Patel R, Kokori E, Olatunji G, Adejumo FA, Ukah JD, Babalola AE, Ndakotsu A, Abraham IC, Aderinto N. Therapeutic Potential of GLP-1 Receptor Agonists in Heart Failure with Preserved Ejection Fraction (HFpEF) in Obese Patients. Curr Heart Fail Rep 2025; 22:17. [PMID: 40366488 DOI: 10.1007/s11897-025-00704-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/02/2025] [Indexed: 05/15/2025]
Abstract
PURPOSE OF REVIEW Heart failure with preserved ejection fraction (HFpEF) is increasingly prevalent among individuals with obesity, primarily due to metabolic dysfunction and structural cardiac remodeling. This review explores the emerging therapeutic role of glucagon-like peptide-1 receptor agonists (GLP-1 RAs) in managing HFpEF in obese populations. RECENT FINDINGS Recent clinical trials, including the STEP-HFpEF and SUMMIT studies, have shown that GLP-1 RAs such as semaglutide and tirzepatide significantly reduce body weight (13.3% and 13.9%, respectively), enhance exercise capacity (increases of 21.5m and 26m in 6-minute walk distance), and improve quality of life (19.5 and 16.6-point increases in KCCQ-CSS scores). Additionally, both agents demonstrated marked reductions in systemic inflammation, with C-reactive protein levels decreasing by 38.8% and 43.5%, respectively. GLP-1 RAs represent a promising class of agents targeting the cardiometabolic axis in HFpEF, offering meaningful improvements in functional capacity and symptom burden among obese patients. However, current evidence is limited by short trial durations, lack of population diversity, and insufficient long-term data. Future research should focus on more inclusive cohorts and extended outcomes such as hospitalization rates and cardiovascular events to fully define the long-term safety and efficacy of GLP-1 RAs in HFpEF management.
Collapse
Affiliation(s)
- Ravi Patel
- Methodist Health System Dallas, Dallas, TX, USA
| | - Emmanuel Kokori
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Gbolahan Olatunji
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | | | - Joan Dumebi Ukah
- National Agency for Food and Drug Administration and Control, Lagos, Nigeria
| | | | - Andrew Ndakotsu
- Department of Medicine, MedStar Union Memorial Hospital, Baltimore, MD, USA
| | | | - Nicholas Aderinto
- Department of Medicine and Surgery, Ladoke Akintola University of Technology, Ogbomoso, Nigeria.
| |
Collapse
|
15
|
Wang M, Preckel B, Zuurbier CJ, Weber NC. Effects of SGLT2 inhibitors on ion channels in heart failure: focus on the endothelium. Basic Res Cardiol 2025:10.1007/s00395-025-01115-y. [PMID: 40366385 DOI: 10.1007/s00395-025-01115-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 05/06/2025] [Accepted: 05/09/2025] [Indexed: 05/15/2025]
Abstract
Heart failure (HF) is a life-threatening cardiovascular disease associated with high mortality, diminished quality of life, and a significant economic burden on both patients and society. The pathogenesis of HF is closely related to the endothelium, where endothelial ion channels play an important role in regulating intracellular Ca2+ signals. These ion channels are essential to maintain vascular function, including endothelium-dependent vascular tone, inflammation response, and oxidative stress. Sodium-glucose cotransporter 2 inhibitors (SGLT2i) have shown promising cardiovascular benefits in HF patients, reducing mortality risk and hospitalization in several large clinical trials. Clinical and preclinical studies indicate that the cardioprotective effects of SGLT2i in HF are mediated by endothelial nitric oxide (NO) pathways, as well as by reducing inflammation and reactive oxygen species in cardiac endothelial cells. Additionally, SGLT2i may confer endothelial protection by lowering intracellular Ca2+ level through the inhibition of sodium-hydrogen exchanger 1 (NHE1) and sodium-calcium exchanger (NCX) in endothelial cells. In this review, we discuss present knowledge regarding the expression and role of Ca2+-related ion channels in endothelial cells in HF, focusing on the effects of SGLT2i on endothelial NHE1, NCX as well as on vascular tone.
Collapse
Affiliation(s)
- Mengnan Wang
- Department of Anesthesiology - Laboratory of Experimental Intensive Care and Anesthesiology-L.E.I.C.A, Amsterdam University Medical Centers, Amsterdam Cardiovascular Science, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Benedikt Preckel
- Department of Anesthesiology - Laboratory of Experimental Intensive Care and Anesthesiology-L.E.I.C.A, Amsterdam University Medical Centers, Amsterdam Cardiovascular Science, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Coert J Zuurbier
- Department of Anesthesiology - Laboratory of Experimental Intensive Care and Anesthesiology-L.E.I.C.A, Amsterdam University Medical Centers, Amsterdam Cardiovascular Science, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Nina C Weber
- Department of Anesthesiology - Laboratory of Experimental Intensive Care and Anesthesiology-L.E.I.C.A, Amsterdam University Medical Centers, Amsterdam Cardiovascular Science, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
16
|
Roman-Pepine D, Serban AM, Capras RD, Cismaru CM, Filip AG. A Comprehensive Review: Unraveling the Role of Inflammation in the Etiology of Heart Failure. Heart Fail Rev 2025:10.1007/s10741-025-10519-w. [PMID: 40360833 DOI: 10.1007/s10741-025-10519-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/27/2025] [Indexed: 05/15/2025]
Abstract
Heart failure (HF) remains a leading cause of morbidity and mortality worldwide, with inflammation playing a pivotal role in its pathogenesis. This comprehensive review aims to elucidate the intricate mechanisms by which inflammation contributes to the development and progression of HF. The review synthesizes current research on the involvement of both innate and adaptive immune responses in HF, highlighting the roles of cytokines, chemokines, and other inflammatory mediators. Recent studies have demonstrated that chronic inflammation, driven by factors such as oxidative stress, neurohormonal activation, and metabolic disturbances, leads to adverse cardiac remodeling and impaired myocardial function. The review explores how systemic inflammation, characterized by elevated levels of inflammatory biomarkers like C-reactive protein (CRP) and interleukin-6 (IL-6), correlates with HF severity and outcomes. Additionally, it discusses the impact of comorbid conditions such as diabetes, obesity, and hypertension on inflammatory pathways and HF risk. The review also delves into the therapeutic implications of targeting inflammation in HF. Despite mixed results from early clinical trials, emerging evidence suggests that anti-inflammatory therapies offer benefits in specific HF phenotypes. The potential of novel therapeutic strategies, including the use of biologics and small molecule inhibitors, is examined in the context of their ability to modulate inflammatory responses and improve clinical outcomes.
Collapse
Affiliation(s)
- Diana Roman-Pepine
- Department of Anatomy and Embryology, Iuliu Hatieganu University of Medicine and Pharmacy, 400348, Cluj-Napoca-Napoca, Romania.
- Cardiology Department, Heart Institute Niculae Stăncioiu, 19-21 Motilor Street, 400001, Cluj-Napoca- Napoca, Romania.
| | - Adela Mihaela Serban
- Cardiology Department, Heart Institute Niculae Stăncioiu, 19-21 Motilor Street, 400001, Cluj-Napoca- Napoca, Romania
- 5 Th Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400348, Cluj-Napoca-Napoca, Romania
| | - Roxana-Denisa Capras
- Department of Anatomy and Embryology, Iuliu Hatieganu University of Medicine and Pharmacy, 400348, Cluj-Napoca-Napoca, Romania
| | - Cristina Mihaela Cismaru
- Department of Infectious Diseases, Iuliu Hatieganu University of Medicine and Pharmacy, 400348, Cluj-Napoca-Napoca, Romania
| | - Adriana Gabriela Filip
- Department of Anatomy and Embryology, Iuliu Hatieganu University of Medicine and Pharmacy, 400348, Cluj-Napoca-Napoca, Romania
| |
Collapse
|
17
|
Packer M, Zile MR, Kramer CM, Murakami M, Ou Y, Borlaug BA. Interplay of Chronic Kidney Disease and the Effects of Tirzepatide in Patients With Heart Failure, Preserved Ejection Fraction, and Obesity: The SUMMIT Trial. J Am Coll Cardiol 2025; 85:1721-1735. [PMID: 40162940 DOI: 10.1016/j.jacc.2025.03.009] [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: 02/12/2025] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Obesity leads to both heart failure with a preserved ejection fraction (HFpEF) and to chronic kidney disease (CKD); CKD may both influence the clinical course of obesity-related HFpEF; and incretin-based drugs may influence renal function. OBJECTIVES This analysis had dual objectives: 1) to evaluate the influence of CKD on the clinical responses to tirzepatide in patients with obesity-related HFpEF; and 2) to investigate the complexity of tirzepatide-related changes in renal function. For both objectives, we focused on discrepancies between creatinine-based and cystatin C-based estimates of the estimated glomerular filtration rate (eGFR). METHODS The SUMMIT trial randomly assigned 731 patients with HFpEF and a body mass index ≥30 kg/m2, who were enriched for participants with CKD. Patients received either placebo or tirzepatide for a median of 104 weeks and were followed for cardiovascular death or worsening heart failure events and for changes in the Kansas City Cardiomyopathy Questionnaire Clinical Summary Score (KCCQ-CSS) after 52 weeks. Because of the confounding produced by obesity and changes in muscle mass, eGFR was assessed at randomization and after 12, 24, and 52 weeks by both creatinine-based and cystatin C-based formulae. RESULTS Patients with CKD (based on creatinine or cystatin C) had greater severity of heart failure, as reflected by: 1) worse functional class, KCCQ-CSS scores, and 6-minute walk distance; 2) higher levels of NT-proBNP and cardiac troponin T; and 3) a 2-fold increase in the risk of worsening heart failure events. CKD did not influence the effect of tirzepatide to reduce the relative risk of major adverse heart failure events and to improve KCCQ-CSS, quality of life, and functional capacity, but the absolute risk reduction in the primary events was numerically greater in patients with CKD. Regarding renal function assessments, baseline eGFR-cystatin C was consistently ≈9 mL/min/1.73 m2 lower than that eGFR-creatinine, with significant individual variance. Furthermore, tirzepatide increased eGFR at 52 weeks, assessed by both creatinine-based and cystatin C-based formulae, but with considerable discordance in individual patients. Tirzepatide produced a decline in eGFR at 12 weeks with eGFR-creatinine (but not eGFR-cystatin C), and it led to an improvement in eGFR at 52 weeks in all patients (when assessed by cystatin C), but only in patients with CKD (when assessed by eGFR-creatinine). CONCLUSIONS The triad of obesity, HFpEF, and CKD identifies patients with considerable functional impairment and an unfavorable prognosis, who nevertheless respond favorably to tirzepatide. Long-term tirzepatide improves renal function (both by cystatin C and creatinine), but the measurement of eGFR in patients with obesity receiving incretin-based drugs is likely to be skewed by the effects of fat and muscle mass (and by changes in body composition) on the synthesis of both cystatin C and creatinine. (A Study of Tirzepatide [LY3298176] in Participants With Heart Failure With Preserved Ejection Fraction [HFpEF] and Obesity: The SUMMIT Trial; NCT04847557).
Collapse
Affiliation(s)
- Milton Packer
- Baylor University Medical Center, Dallas Texas, USA; Imperial College, London, United Kingdom.
| | - Michael R Zile
- RHJ Department of Veterans Affairs, Medical Center and Medical University of South Carolina, Charleston, South Carolina, USA
| | - Christopher M Kramer
- Cardiovascular Division, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia, USA
| | | | - Yang Ou
- Eli Lilly & Company, Indianapolis, Indiana, USA
| | - Barry A Borlaug
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
18
|
Ter Maaten JM, Butler J. Tirzepatide, HFpEF, and the Kidney: A Triangular Relationship Requiring Measurement of GFR, Not Guesses. J Am Coll Cardiol 2025; 85:1736-1739. [PMID: 40183720 DOI: 10.1016/j.jacc.2025.03.508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 04/05/2025]
Affiliation(s)
- Jozine M Ter Maaten
- University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, Texas, USA; University of Mississippi, Jackson, Mississippi, USA.
| |
Collapse
|
19
|
Hobbach AJ, Brix TJ, Weyer-Elberich V, Varghese J, Reinecke H, Linke WA. Obesity and Comorbidities in HFpEF: A Retrospective Cohort Analysis in a University Hospital Setting. J Clin Med 2025; 14:3348. [PMID: 40429344 PMCID: PMC12112532 DOI: 10.3390/jcm14103348] [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: 03/26/2025] [Revised: 05/02/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Heart failure (HF) with preserved ejection fraction (HFpEF) poses a diagnostic challenge, as it lacks a definitive hallmark beyond preserved left ventricular (LV)EF. This retrospective study aims to analyze the demographic and clinical characteristics of HFpEF patients within a real-world hospital cohort, with a particular focus on obesity and associated comorbidities. Methods: A total of 4019 patients who underwent echocardiography in 2020-2021 at a university hospital were screened for HFpEF. After stringent manual verification, 219 patients fulfilled the European Society of Cardiology (ESC) criteria for HFpEF. Demographic, clinical, and comorbidity data were analyzed and stratified by body mass index (BMI) categories and sex distribution. Results: Among the 219 HFpEF patients, 71.3% were classified as pre-obese or obese. Hypertension (93.6%), atrial fibrillation (74.3%), and obesity were common in the cohort, while sex distribution was balanced. Edema was more prevalent among obese patients, though HFpEF severity, as reflected by New York Heart Association (NYHA) classification and natriuretic peptide levels, did not differ significantly across BMI groups. Medical treatment patterns varied with BMI, with obese patients more frequently receiving diuretics, sodium-glucose cotransporter-2 inhibitors (SGLT-2i), and angiotensin receptor blockers (ARB). No significant differences were observed between male and female patients in terms of HFpEF severity markers. Conclusions: Obesity is a predominant feature in HFpEF and is associated with a high burden of comorbidities. However, sex does not appear to influence HFpEF severity in this cohort. These findings underscore the need for targeted therapeutic strategies in obese HFpEF patients.
Collapse
Affiliation(s)
- Anastasia Janina Hobbach
- Department of Cardiology I, Coronary, Peripheral Vascular Disease and Heart Failure, University Hospital Münster, 48149 Münster, Germany;
- Institute of Physiology II, University of Münster, 48149 Münster, Germany
| | - Tobias Johannes Brix
- Institute of Medical Informatics, University of Münster, 48149 Münster, Germany; (T.J.B.); (J.V.)
| | - Veronika Weyer-Elberich
- Institute of Biostatistics and Clinical Research, University of Münster, 48149 Münster, Germany;
| | - Julian Varghese
- Institute of Medical Informatics, University of Münster, 48149 Münster, Germany; (T.J.B.); (J.V.)
| | - Holger Reinecke
- Department of Cardiology I, Coronary, Peripheral Vascular Disease and Heart Failure, University Hospital Münster, 48149 Münster, Germany;
| | | |
Collapse
|
20
|
Pepin ME, Konrad PJM, Nazir S, Bazgir F, Maack C, Nickel A, Gorman J, Hohl M, Schreiter F, Dewenter M, de Britto Chaves Filho A, Schulze A, Karlstaedt A, Frey N, Seidman C, Seidman J, Backs J. Mitochondrial NNT Promotes Diastolic Dysfunction in Cardiometabolic HFpEF. Circ Res 2025. [PMID: 40340422 DOI: 10.1161/circresaha.125.326154] [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: 01/10/2025] [Revised: 04/14/2025] [Accepted: 04/21/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Clinical management of heart failure with preserved ejection fraction (HFpEF) is hindered by a lack of disease-modifying therapies capable of altering its distinct pathophysiology. Despite the widespread implementation of a 2-hit model of cardiometabolic HFpEF to inform precision therapy, which utilizes ad libitum high-fat diet and 0.5% N(ω)-nitro-L-arginine methyl ester, we observe that C57BL6/J mice exhibit less cardiac diastolic dysfunction in response to high-fat diet and 0.5% N(ω)-nitro-L-arginine methyl ester. METHODS Genetic strain-specific single-nucleus transcriptomic analysis identified disease-relevant genes that enrich oxidative metabolic pathways within cardiomyocytes. Because C57BL/6J mice are known to harbor a loss-of-function mutation affecting the inner mitochondrial membrane protein Nnt (nicotinamide nucleotide transhydrogenase), we used an isogenic model of Nnt loss-of-function to determine whether intact NNT is necessary for the pathological cardiac manifestations of high-fat diet and 0.5% N(ω)-nitro-L-arginine methyl ester. Twelve-week-old mice cross-bred to isolate wild-type (Nnt+/+) or loss-of-function (Nnt-/-) Nnt in the C57BL/6N background were challenged with high-fat diet and 0.5% N(ω)-nitro-L-arginine methyl ester for 9 weeks (n=6-10). RESULTS Nnt+/+ mice exhibited impaired ventricular diastolic relaxation and pathological remodeling, as assessed via E/e' (42.8 versus 21.5, P=1.2×10-10), E/A (2.3 versus 1.4, P=4.1×10-2), diastolic stiffness (0.09 versus 0.04 mm Hg/μL, P=5.1×10-3), and myocardial fibrosis (P=2.3×10-2). Liquid chromatography and mass spectroscopy exposed a 40.0% reduction in NAD+ (P=8.4×10-3) and a 38.8% reduction in glutathione:GSSG (P=2.6×10-2) among Nnt+/+ mice after high-fat diet and 0.5% N(ω)-nitro-L-arginine methyl ester feeding. Using single-nucleus ligand-receptor analysis, we implicate Fgf1 (fibroblast growth factor 1) as a putative NNT-dependent mediator of cardiomyocyte-to-fibroblast signaling of myocardial fibrosis. CONCLUSIONS Together, these findings underscore the pivotal role of mitochondrial dysfunction in HFpEF pathogenesis, implicating both NNT and Fgf1 as novel therapeutic targets.
Collapse
Affiliation(s)
- Mark E Pepin
- Medical Faculty Heidelberg, Institute of Experimental Cardiology, Heidelberg University, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Department of Internal Medicine VIII, Heidelberg University Hospital, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
- Department of Genetics, Harvard Medical School, Boston, MA (M.E.P., J.G., C.S., J.S.)
- Broad Institute of Harvard and MIT, Boston, MA (M.E.P.)
- Division of Cardiovascular Medicine, Stanford University Hospital, CA (M.E.P.)
| | - Philipp J M Konrad
- Medical Faculty Heidelberg, Institute of Experimental Cardiology, Heidelberg University, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Department of Internal Medicine VIII, Heidelberg University Hospital, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Department of Internal Medicine III, Heidelberg University Hospital, Germany. (P.J.M.K., N.F.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
| | - Sumra Nazir
- Medical Faculty Heidelberg, Institute of Experimental Cardiology, Heidelberg University, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Department of Internal Medicine VIII, Heidelberg University Hospital, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
| | - Farhad Bazgir
- Medical Faculty Heidelberg, Institute of Experimental Cardiology, Heidelberg University, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Department of Internal Medicine VIII, Heidelberg University Hospital, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
| | - Christoph Maack
- Comprehensive Heart Failure Center, University Clinic Würzburg, Germany (C.M., A.N.)
- Medical Clinic I, University Clinic Würzburg, Germany (C.M.)
| | - Alexander Nickel
- Comprehensive Heart Failure Center, University Clinic Würzburg, Germany (C.M., A.N.)
| | - Joshua Gorman
- Department of Genetics, Harvard Medical School, Boston, MA (M.E.P., J.G., C.S., J.S.)
| | - Mathias Hohl
- Department of Internal Medicine III, Saarland University Hospital and Saarland University, Homburg/Saar, Germany (M.H.)
| | - Friederike Schreiter
- Medical Faculty Heidelberg, Institute of Experimental Cardiology, Heidelberg University, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Department of Internal Medicine VIII, Heidelberg University Hospital, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
| | - Matthias Dewenter
- Medical Faculty Heidelberg, Institute of Experimental Cardiology, Heidelberg University, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Department of Internal Medicine VIII, Heidelberg University Hospital, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory (EMBL), Germany and Heidelberg University, Germany (M.D., J.B.)
| | | | - Almut Schulze
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center, Heidelberg (A.d.B.C.F., A.S.)
| | - Anja Karlstaedt
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (A.K.)
| | - Norbert Frey
- Department of Internal Medicine III, Heidelberg University Hospital, Germany. (P.J.M.K., N.F.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
| | - Christine Seidman
- Department of Genetics, Harvard Medical School, Boston, MA (M.E.P., J.G., C.S., J.S.)
| | - Jonathan Seidman
- Department of Genetics, Harvard Medical School, Boston, MA (M.E.P., J.G., C.S., J.S.)
| | - Johannes Backs
- Medical Faculty Heidelberg, Institute of Experimental Cardiology, Heidelberg University, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Helmholtz-Institute for Translational AngioCardioScience (HI-TAC) of the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Heidelberg University, Germany. (J.B.)
- Department of Internal Medicine VIII, Heidelberg University Hospital, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory (EMBL), Germany and Heidelberg University, Germany (M.D., J.B.)
| |
Collapse
|
21
|
Taylor EB, Hall JE, Mouton AJ. Current anti-inflammatory strategies for treatment of heart failure: From innate to adaptive immunity. Pharmacol Res 2025; 216:107761. [PMID: 40348101 DOI: 10.1016/j.phrs.2025.107761] [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: 02/23/2025] [Revised: 04/18/2025] [Accepted: 04/30/2025] [Indexed: 05/14/2025]
Abstract
Cardiovascular disease (CVD) is the leading cause of death in developed countries worldwide, often manifesting in the form of heart failure (HF). Recent successful clinical outcomes of anti-inflammatory therapies in HF patients have greatly boosted interest in basic and translational research on the role of inflammation in development of HF. In this review, we discuss recent and ongoing therapies targeting inflammation in CVD/HF, including broad-spectrum anti-inflammatory drugs, supplements, and biologicals such as canakinumab and anakinra. We also discuss the growing body of literature supporting off-target/anti-inflammatory actions of mainline CVD/HF agents, including guideline-directed medical therapy (GDMT) drugs that target the neurohormonal axis, and statins. We discuss therapeutics that target autoimmune mechanisms, and their implications for treating patients with autoimmune diseases with HF or at-risk of developing HF. We also discuss recent evidence for vaccines in modulating the immune response during HF. We conclude that despite the wealth of knowledge gained in the past decade, the therapeutic efficacy of anti-inflammatory therapy is driven by many biological and logistical factors that vary from patient to patient. Furthermore, more studies are needed to understand the adaptive/autoimmune component of HF, particularly in women and patients with pre-existing autoimmune disease. As the number of patients with HF patients who suffer from obesity, diabetes, or autoimmune disease continues to grow, our understanding of inflammation must continue to evolve to reflect these underlying co-morbidities.
Collapse
Affiliation(s)
- Erin B Taylor
- Department of Physiology and Biophysics and Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - John E Hall
- Department of Physiology and Biophysics and Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - Alan J Mouton
- Department of Physiology and Biophysics and Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS 39216, United States.
| |
Collapse
|
22
|
Hao J, Wang M, Wu Q, Song T, Hao Y, Chang L, Hou Y, Jia Z. Deciphering the molecular mechanisms of QLQX capsules in heart failure: A multi-omics perspective. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156828. [PMID: 40378592 DOI: 10.1016/j.phymed.2025.156828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 04/26/2025] [Accepted: 05/02/2025] [Indexed: 05/19/2025]
Abstract
PURPOSE This study investigates the therapeutic mechanisms of Qiliqiangxin (QLQX) capsules in treating Heart Failure with Preserved Ejection Fraction (HFpEF). The study aims to understand how QLQX impacts cardiac function and underlying molecular pathways. METHODS HFpEF was induced in a rat model through unilateral nephrectomy, DOCA pellet implantation, and a high-salt diet. Cardiac function was assessed via M-mode imaging and Doppler flow measurements, focusing on key parameters like ejection fraction and diastolic function. A network pharmacology approach identified active QLQX components and potential targets, followed by comprehensive multi-omics analyses-including transcriptomics, proteomics, and metabolomics-to uncover the molecular mechanisms modulated by QLQX. Quantitative RT-PCR was employed to measure mRNA levels of key cardiac markers, providing further insights into QLQX's impact on cardiac remodeling. RESULTS QLQX treatment significantly improved cardiac function, with notable enhancements in ejection fraction and left ventricular diastolic function. Network pharmacology revealed 530 potential targets of QLQX, with 38 overlapping HFpEF targets. Key pathways identified include cGMP-PKG, adrenergic signaling, and calcium signaling. Transcriptomic analysis showed significant gene expression changes related to inflammation, energy metabolism, and myocardial remodeling, which were reversed by QLQX. Proteomic analysis identified 401 differentially expressed proteins, enriched in pathways such as cGMP-PKG and NF-κB signaling. Metabolomic profiling highlighted the role of lipid metabolism and adrenergic signaling in HFpEF, which were normalized by QLQX. In vivo validation confirmed the involvement of the cGMP-PKG pathway, with increased serum NO and cGMP levels, improved endothelial function, and reduced pro-fibrotic markers following QLQX treatment. CONCLUSION QLQX exerts multifaceted therapeutic effects on HFpEF by modulating gene expression, protein function, and metabolic pathways, particularly through the cGMP-PKG signaling pathway. These findings support QLQX as a promising therapeutic intervention for HFpEF, offering improvements in cardiac function and reversing pathological changes at multiple molecular levels.
Collapse
Affiliation(s)
- Jiameng Hao
- China Science and Technology Development Center for Chinese Medicine, Beijing, China
| | - Mingye Wang
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, No.326, Xinshi South Road, Shijiazhuang 050091, Hebei, China
| | - Qiulan Wu
- Hebei Medical University, No.361 Zhongshan East Road, Shijiazhuang 050011, Hebei, China
| | - Tao Song
- Shijiazhuang Yiling Pharmaceutical Co., Ltd, No.238, South of Tianshan Street, Shijiazhuang 050035, Hebei, China
| | - Yuanyuan Hao
- Shijiazhuang Yiling Pharmaceutical Co., Ltd, No.238, South of Tianshan Street, Shijiazhuang 050035, Hebei, China
| | - Liping Chang
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, No.326, Xinshi South Road, Shijiazhuang 050091, Hebei, China; Hebei Medical University, No.361 Zhongshan East Road, Shijiazhuang 050011, Hebei, China; State Key Laboratory for Innovation and Transformation of Luobing Theory, Hebei, China
| | - Yunlong Hou
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, No.326, Xinshi South Road, Shijiazhuang 050091, Hebei, China; Hebei Medical University, No.361 Zhongshan East Road, Shijiazhuang 050011, Hebei, China; State Key Laboratory for Innovation and Transformation of Luobing Theory, Hebei, China.
| | - Zhenhua Jia
- Shijiazhuang Yiling Pharmaceutical Co., Ltd, No.238, South of Tianshan Street, Shijiazhuang 050035, Hebei, China; State Key Laboratory for Innovation and Transformation of Luobing Theory, Hebei, China; Hebei Yiling Hospital, Shijiazhuang, 050035, Hebei, China.
| |
Collapse
|
23
|
Qian W, Lingli X, Dexue L, Yangwen C, Yongyan S, Weihua W. Influence of fluctuations in fasting blood glucose on left ventricular function in patients with type 2 diabetes mellitus and coronary microcirculation dysfunction: a prospective cohort study. Acta Diabetol 2025:10.1007/s00592-025-02514-2. [PMID: 40332563 DOI: 10.1007/s00592-025-02514-2] [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: 12/31/2024] [Accepted: 04/16/2025] [Indexed: 05/08/2025]
Abstract
AIMS To examine the effects of fluctuations in fasting blood glucose (FBG) levels on left ventricular function in patients with T2DM and coronary microcirculation dysfunction (CMD). METHODS A total of 290 patients with T2DM who received glucose-lowering therapy during hospitalization and were subsequently followed up for 18 months at the First Affiliated Hospital of Harbin Medical University, were enrolled in this study. 135 were diagnosed with CMD and were assigned to the CMD group, whereas 155 patients without CMD were allocated to the non-CMD group. The fasting blood glucose coefficient of variation (FBG-CV) was calculated for all participants. The CMD group was further stratified into three subgroups based on their FBG-CV values: CMD1 (FBG-CV > 25%), CMD2 (FBG-CV 15% ~ 25%), and CMD3 (FBG-CV < 15%). The left ventricular function, assessed by left ventricular ejection fraction (LVEF) and the E/e' ratio, was compared within each group before and after the follow-up period. This study was registered in the Chinese Clinical Trial Register, ChiCTR-ORC-16009800. RESULTS After the end of follow-up, the E/e' ratio in CMD1 was significantly higher than that in CMD2 and CMD3 (14.35 vs 8.57; p < 0.01; 14.35 vs 6.61; p < 0.01), and the E/e' ratio in CMD2 was significantly higher than that in CMD3 (8.57 vs 6.61; p < 0.01). Compared to the baseline measurements, the E/e' ratio in CMD1 showed a significant increase after an average 17.8 months of follow up (14.35 vs 8.44; p < 0.001). We found elevated E/e' ratio was associated with an increased FBG-CV level (odds ratio [OR]: 2.571; 95% CI 1.819-3.634; p < 0.001). In multivariate logistic analysis, course of diabetes (OR:1.062; 1.016-1.11; P = 0.007) and CMD (OR:2.231; 1.303-3.819; P = 0.003), were significantly associated with elevated E/e' ratio, while oral stains drugs (OR = 0.412 95% CI 0.237-0.715; P = 0.002) and insulin injections (OR = 0.536 95% CI 0.311-0.924; P = 0.025) behaved as a protective factor. CONCLUSIONS Our study clarified the association between FBG-CV levels and the E/e' ratio in a prospective cohort study. In T2DM patients with CMD, FBG-CV > 25% may adversely affect left ventricular diastolic function, whereas an optimal FBG-CV is considered to be less than 15%.
Collapse
Affiliation(s)
- Wang Qian
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Department of Endocrinology, Shenzhen Third People's Hospital, Shenzhen, 518112, Guangdong Province, China
| | - Xie Lingli
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Lu Dexue
- Department of Endocrinology, Shenzhen Third People's Hospital, Shenzhen, 518112, Guangdong Province, China
| | - Chen Yangwen
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Shan Yongyan
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Wu Weihua
- Department of Endocrinology, Shenzhen Third People's Hospital, Shenzhen, 518112, Guangdong Province, China.
| |
Collapse
|
24
|
Cotter G, Petrie MC, Butler J, Davison B, Chioncel O, Biegus J, Pagnesi M, Voors AA, Metra M, Ponikowski P, Mann D, Bhatt DL. Obesity and inflammation in chronic and acute heart failure. Heart Fail Rev 2025:10.1007/s10741-025-10518-x. [PMID: 40319221 DOI: 10.1007/s10741-025-10518-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2025] [Indexed: 05/07/2025]
Abstract
Obesity and inflammation have been associated with an increased incidence of heart failure (HF) and death. However, until recent years, no therapy directed towards reducing inflammation and reducing obesity has been shown to reduce those adverse outcomes. Over the past few years, a few small studies have suggested that improving obesity-and in even smaller studies, reducing inflammation-may help improve HF severity, congestion, quality of life, and possibly outcomes. Larger studies that are being planned and executed, which will report their results within the next 2-3 years, should help further clarify the effects of weight and inflammation reduction in patients with HF.
Collapse
Affiliation(s)
- Gad Cotter
- Université Paris Cité, INSERM UMR-S 942 (MASCOT), Paris, France.
- Momentum Research Inc, Durham, NC, USA.
- , Heart Initiative, Durham, NC, USA.
| | - Mark C Petrie
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, Scotland, UK
| | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, TX, USA
- Department of Medicine, University of Mississippi, Jackson, MS, USA
| | - Beth Davison
- Université Paris Cité, INSERM UMR-S 942 (MASCOT), Paris, France
- Momentum Research Inc, Durham, NC, USA
- , Heart Initiative, Durham, NC, USA
| | - Ovidiu Chioncel
- Emergency Institute for Cardiovascular Diseases "Prof C C Iliescu", University of Medicine "Carol Davila", Bucharest, Romania
| | - Jan Biegus
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Matteo Pagnesi
- Cardiology, Cardiology, ASST Spedali Civili and Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy
| | - Adriaan A Voors
- Department of Cardiology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Marco Metra
- Cardiology, Cardiology, ASST Spedali Civili and Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy
| | - Piotr Ponikowski
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Douglas Mann
- Cardiovascular Division, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Deepak L Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
25
|
Nakashima M, Miyoshi T, Nishihara T, Miki T, Ejiri K, Hara S, Takaya Y, Nakayama R, Ichikawa K, Osawa K, Yuasa S. Prognostic Value of Pericoronary Fat Attenuation Index on Computed Tomography for Hospitalization for Heart Failure. JACC. ADVANCES 2025; 4:101685. [PMID: 40286356 PMCID: PMC12102526 DOI: 10.1016/j.jacadv.2025.101685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 02/15/2025] [Accepted: 02/24/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Pericoronary fat attenuation index (FAI) assessed on computed tomography is associated with the inflammation of the pericoronary artery. OBJECTIVES This study aimed to investigate whether pericoronary FAI predicts hospitalization for heart failure with preserved ejection fraction (HFpEF). METHODS This retrospective single-center study included 1,196 consecutive patients who underwent clinically indicated coronary computed tomography angiography (CCTA) and transthoracic echocardiography. We assessed the FAI of proximal 40-mm segments for each major epicardial coronary vessel. The primary outcome was the incidence of hospitalization for HFpEF. Patients were divided into groups based on the optimal cutoff value for predicting hospitalization for HFpEF by receiver operating characteristic curve analysis. RESULTS During a median follow-up of 4.3 years, 29 hospitalizations for HFpEF occurred. Multivariable Cox regression analysis revealed that a left anterior descending artery (LAD)-FAI ≥-63.4 HU and a left circumflex artery-FAI ≥-61.6 HU were significantly associated with hospitalization for HF after adjustment for age and sex (HR: 4.8; 95% CI: 2.1-10.8 and HR: 4.5; 95% CI: 2.1-9.4, respectively). The addition of LAD-FAI >-63.4 HU to a model incorporating other risk factors, including hypertension, estimated glomerular filtration rate <60 mL/min/1.73 m2, and significant stenosis on CCTA, increased the C-statistic for predicting hospitalization for HFpEF from 0.646 to 0.750 (P = 0.010). CONCLUSIONS LAD- and left circumflex artery-FAI can predict hospitalization for HFpEF in patients undergoing clinically indicated CCTA. Pericoronary inflammation may be useful for identifying patients at high risk of developing HFpEF.
Collapse
Affiliation(s)
- Mitsutaka Nakashima
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toru Miyoshi
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| | - Takahiro Nishihara
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takashi Miki
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kentaro Ejiri
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shohei Hara
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yoichi Takaya
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Rie Nakayama
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Keishi Ichikawa
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazuhiro Osawa
- Department of General Internal Medicine 3, Kawasaki Medical School General Medicine Centre, Okayama, Japan
| | - Shinsuke Yuasa
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
26
|
Seleng J, Celovska D, Procka P, Labuda M, Borik S. Camera-based evaluation of deep breathing effects on plantar foot microcirculation - A pilot study on young healthy. Comput Biol Med 2025; 189:109996. [PMID: 40068495 DOI: 10.1016/j.compbiomed.2025.109996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/26/2025] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Microvascular function, particularly of the plantar foot, reflects overall vascular health and is influenced by physiological oscillators such as heart rate, respiratory, myogenic, and neurogenic rhythms. Slow deep breathing modulates autonomic nervous system activity and affects peripheral microcirculation. This study investigates the effects of slow deep breathing on plantar foot perfusion using photoplethysmography imaging (PPGI). METHODS Twenty healthy young adults participated in a four-stage protocol: baseline, deep breathing test (DBT), and two recovery stages (REST1 and REST2). PPGI was used to measure changes in plantar foot perfusion, focusing on energy, amplitude, and phase synchronization within frequency bands corresponding to key physiological oscillators. Time-frequency analyses and advanced signal processing were applied to assess these parameters. RESULTS Significant increases in energy were observed in all frequency bands during DBT, with slow frequency oscillators (SFOs) maintaining elevated activity up to 5 min after DBT. Amplitude analysis revealed a significant decrease in the first and second harmonic components of the heart rate signals during DBT. Phase synchronization between medial and lateral foot regions improved for respiratory, myogenic, and neurogenic frequency bands during DBT, with myogenic synchronization persisting for up to 2.5 min after DBT. CONCLUSIONS Slow deep breathing enhances microvascular perfusion and synchronizes autonomic oscillators in healthy individuals. PPGI proved effective in capturing these dynamics, indicating its potential as a non-invasive tool for assessing autonomic and microvascular function. Future research should explore its applicability in detecting early autonomic or vascular dysfunction in clinical populations.
Collapse
Affiliation(s)
- Jan Seleng
- Dept. of Electromagnetic and Biomedical Engineering, Faculty of Electrical Engineering and Information Technology, University of Zilina, Zilina, Slovakia
| | - Denisa Celovska
- 1st Department of Internal Medicine, Faculty of Medicine, Comenius University, 813 69, Bratislava, Slovakia
| | - Patrik Procka
- Dept. of Electromagnetic and Biomedical Engineering, Faculty of Electrical Engineering and Information Technology, University of Zilina, Zilina, Slovakia
| | - Michal Labuda
- Dept. of Electromagnetic and Biomedical Engineering, Faculty of Electrical Engineering and Information Technology, University of Zilina, Zilina, Slovakia
| | - Stefan Borik
- Dept. of Electromagnetic and Biomedical Engineering, Faculty of Electrical Engineering and Information Technology, University of Zilina, Zilina, Slovakia.
| |
Collapse
|
27
|
Holm H, Magnusson M, Jujić A, Lagrange J, Bozec E, Lamiral Z, Bresso E, Huttin O, Baudry G, Monzo L, Rossignol P, Zannad F, Girerd N. Association of ventricular-arterial coupling with biomarkers involved in heart failure pathophysiology - the STANISLAS cohort. Eur J Heart Fail 2025; 27:860-871. [PMID: 39189882 DOI: 10.1002/ejhf.3411] [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: 01/15/2024] [Revised: 07/21/2024] [Accepted: 07/25/2024] [Indexed: 08/28/2024] Open
Abstract
AIMS Impaired left ventricular-arterial coupling (VAC) has been shown to correlate with worse prognosis in cardiac diseases and heart failure (HF). The extent of the relationship between VAC and circulating biomarkers associated with HF has been scarcely documented. We aimed to explore associations of VAC with proteins involved in HF pathophysiology within a large population-based cohort of middle-aged individuals. METHODS AND RESULTS In the forth visit of the STANISLAS family cohort, involving 1309 participants (mean age 48 ± 14 years; 48% male) from parent and children generations, we analysed the association of 32 HF-related proteins with non-invasively assessed VAC using pulse wave velocity (PWV)/global longitudinal strain (GLS) and arterial elastance (Ea)/ventricular end-systolic elastance (Ees). Among the 32 tested proteins, fatty acid-binding protein adipocyte 4, interleukin-6, growth differentiation factor 15, matrix metalloproteinase (MMP)-1, and MMP-9 and adrenomedullin were positively associated with PWV/GLS whereas transforming growth factor beta receptor type 3, MMP-2 and N-terminal pro-B-type natriuretic peptide (NT-proBNP) were negatively associated. In multivariable models, only MMP-2 and NT-proBNP were significantly and inversely associated with PWV/GLS in the whole population and in the parent generation. Higher levels of NT-proBNP were also negatively associated with Ea/Ees in the whole cohort but this association did not persist in the parent subgroup. CONCLUSION Elevated MMP-2 and NT-proBNP levels correlate with better VAC (lower PWV/GLS), possibly indicating a compensatory cardiovascular response to regulate left ventricular pressure amidst cardiac remodelling and overload.
Collapse
Affiliation(s)
- Hannes Holm
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
| | - Martin Magnusson
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa
| | - Amra Jujić
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
| | - Jérémy Lagrange
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Erwan Bozec
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Zohra Lamiral
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Emmanuel Bresso
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Olivier Huttin
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Guillaume Baudry
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Luca Monzo
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Patrick Rossignol
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Faiez Zannad
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Nicolas Girerd
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| |
Collapse
|
28
|
Fazzini L, Hubers SA, Cao JJ, Scott CG, McCully RB, Castrichini M, Figueiral M, Mohananey A, Wang L, Gulati R, Montisci R, Pellikka PA, Pereira NL. Exercise-Induced Reduction in Left Ventricular Ejection Fraction in the Absence of Coronary Artery Disease: Clinical Characteristics and Outcomes. J Am Soc Echocardiogr 2025; 38:421-430. [PMID: 39613116 DOI: 10.1016/j.echo.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND During exercise stress echocardiography (ESE), there are patients with normal left ventricular ejection fraction (LVEF) who paradoxically develop reduced LVEF during exercise despite absence of coronary artery disease (CAD) and a significant hypertensive response. This study sought to describe the clinical features and outcomes of this population. METHODS Among ESEs performed between 2003 and 2022, patients without CAD by angiogram within 90 days of ESE and resting LVEF ≥50% with a ≥5% LVEF decrease during ESE were included. Outcomes assessed were all-cause mortality, heart failure (HF) hospitalization, and atrial fibrillation (AF). Kaplan-Meier and Cox regression methods were used to analyze time-to-event outcomes. RESULTS Among 213,643 ESE, 134 patients met the eligibility criteria. The mean age of the population was 66 ± 10 years, 76% were women, and 16% had AF at baseline. Mean LVEF was 58% ± 4% at rest and 43% ± 4% at peak stress. Stress ECG met the criteria for ischemia in 14% of these patients. The 10-year estimated incidence of HF hospitalization was 17.6% (95% CI, 9.0%-26.2%). Among the subgroup without AF at baseline, the 10-year estimated incidence of developing AF was 23.4% (95% CI, 13.4%-33.4%). The 10-year estimated incidence of all-cause mortality was 12.9% (95% CI, 5.5%-20.3%), with 89% of deaths occurring due to noncardiovascular causes. CONCLUSION Patients with exercise-induced reduction in LVEF in the absence of obstructive CAD have a high incidence of HF hospitalizations and AF. The underlying pathophysiology of this disease process needs to be further investigated.
Collapse
Affiliation(s)
- Luca Fazzini
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota; Clinical Cardiology Unit, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Scott A Hubers
- Minneapolis Heart Institute, United Hospital, Nasseff Specialty Center, St. Paul, Minnesota
| | - Jenny J Cao
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Christopher G Scott
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Robert B McCully
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Matteo Castrichini
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Marta Figueiral
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Akanksha Mohananey
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Li Wang
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Rajiv Gulati
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Roberta Montisci
- Clinical Cardiology Unit, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | | | - Naveen L Pereira
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
29
|
Dregoesc I, Hung JW, Dal-Bianco JP. Diastolic Function in Acute Myocardial Infarction: Do We Need to Relax More? Echocardiography 2025; 42:e70199. [PMID: 40387012 DOI: 10.1111/echo.70199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2025] [Accepted: 05/05/2025] [Indexed: 05/20/2025] Open
Affiliation(s)
- Ioana Dregoesc
- Echocardiography Laboratory, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Judy W Hung
- Echocardiography Laboratory, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jacob P Dal-Bianco
- Echocardiography Laboratory, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
30
|
Velollari O, Rommel KP, Kresoja KP, Lurz P, Gori T. Focusing on microvascular function in heart failure with preserved ejection fraction. Heart Fail Rev 2025; 30:493-503. [PMID: 39804445 PMCID: PMC11992002 DOI: 10.1007/s10741-024-10479-7] [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] [Accepted: 12/20/2024] [Indexed: 04/12/2025]
Abstract
Heart failure is a prevalent global health issue. Heart failure with preserved ejection fraction (HFpEF), which already represents half of all heart cases worldwide, is projected to further increase, driven by aging populations and rising cardiovascular risk factors. Effective therapies for HFpEF remain limited, particularly due to its pathophysiological heterogeneity and incomplete understanding of underlying pathomechanisms and implications. Coronary microvascular dysfunction (CMD), characterized by structural and functional changes in the coronary microcirculation, is increasingly recognized as a significant factor in HFpEF even though the exact nature of their causal relationship is still unclear. This review explores prevalence, prognostic implications, and potential therapeutic targets for CMD in HFpEF. CMD's role in HFpEF might involve impaired coronary blood flow regulation, leading to myocardial ischemia, impaired relaxation, and/or adverse remodeling. Vice versa, increased wall stress in patients with HFpEF might elevate coronary resistances, further worsening microvascular perfusion. Finally, abnormalities in substrate metabolism might cause both CMD and HFpEF. Current treatments, including pharmacotherapy and device-based therapies, show limited success, highlighting the need for more targeted approaches. New possible therapies, such as the coronary sinus reducer device, may show promise in improving myocardial perfusion and function. However, further large-scale studies are required to elucidate the mechanistic links between CMD and HFpEF and to develop specialized treatments for distinct heart failure phenotypes.
Collapse
Affiliation(s)
- Ornela Velollari
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
- German Centre for Cardiovascular Research (DZHK), Standort Rhein-Main, Frankfurt, Germany
| | - Karl-Philipp Rommel
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
- German Centre for Cardiovascular Research (DZHK), Standort Rhein-Main, Frankfurt, Germany
| | - Karl-Patrik Kresoja
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
- German Centre for Cardiovascular Research (DZHK), Standort Rhein-Main, Frankfurt, Germany
| | - Philipp Lurz
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
- German Centre for Cardiovascular Research (DZHK), Standort Rhein-Main, Frankfurt, Germany
| | - Tommaso Gori
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany.
- German Centre for Cardiovascular Research (DZHK), Standort Rhein-Main, Frankfurt, Germany.
| |
Collapse
|
31
|
Iacovoni A, Navazio A, De Luca L, Gori M, Corda M, Milli M, Iacoviello M, Di Lenarda A, Di Tano G, Marini M, Iorio A, Mortara A, Mureddu GF, Zilio F, Chimenti C, Cipriani MG, Senni M, Bilato C, Di Marco M, Geraci G, Pascale V, Riccio C, Scicchitano P, Tizzani E, Gulizia MM, Nardi F, Gabrielli D, Colivicchi F, Grimaldi M, Oliva F. ANMCO position paper: diagnosis and treatment of heart failure with preserved systolic function. Eur Heart J Suppl 2025; 27:v216-v246. [PMID: 40385467 PMCID: PMC12078774 DOI: 10.1093/eurheartjsupp/suaf070] [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] [Indexed: 05/20/2025]
Abstract
Heart failure is the leading cardiovascular cause of hospitalization with an increasing prevalence, especially in older patients. About 50% of patients with heart failure have preserved ventricular function, a form of heart failure that, until a few years ago, was orphaned by pharmacological treatments effective in reducing hospitalization and mortality. New trials, which have tested the use of gliflozins in patients with heart failure with preserved ejection fraction (HFpEF), have for the first time demonstrated their effectiveness in changing the natural history of this insidious and frequent form of heart failure. Therefore, diagnosing those patients early is crucial to provide the best treatment. Moreover, the diagnosis is influenced by the patient's comorbidities, and some HFpEF patients have symptoms common to other rare diseases that, if unrecognized, develop an unfavourable prognosis. This position paper aims to provide the clinician with a useful tool for diagnosing and treating patients with HFpEF, guiding the clinician towards the most appropriate diagnostic and therapeutic pathway.
Collapse
Affiliation(s)
- Attilio Iacovoni
- S.S.D. Chirurgia dei Trapianti e del Trattamento Chirurgico dello Scompenso, Dipartimento Cardiovascolare, ASST Papa Giovanni XXIII, Piazza OMS 1, Bergamo 24127, Italy
| | - Alessandro Navazio
- S.O.C. Cardiologia Ospedaliera, Presidio Ospedaliero Arcispedale Santa Maria Nuova, Azienda USL di Reggio Emilia—IRCCS, Reggio Emilia, Italy
| | - Leonardo De Luca
- S.C. Cardiologia, Dipartimento Cardio-Toraco-Vascolare, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Mauro Gori
- U.O.C. Cardiologia 1, Dipartimento Cardiovascolare, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Marco Corda
- S.C. Cardiologia, Azienda di Rilievo Nazionale e Alta Specializzazione ‘G. Brotzu’, Cagliari, Italy
| | - Massimo Milli
- Cardiologia Firenze 1 (Ospedali S. Maria Nuova e Nuovo San Giovanni di Dio), Azienda USL Toscana Centro, Florence, Italy
| | | | - Andrea Di Lenarda
- S.C. Patologie Cardiovascolari, Dipartimento Specialistico Territoriale, Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI), Trieste, Italy
| | - Giuseppe Di Tano
- U.O. CARDIOLOGIA - UCC, Ospedale Cernusco sul Naviglio, Cernusco sul Naviglio, MI, Italy
| | - Marco Marini
- S.O.S. Terapia Intensiva Cardiologica, S.O.D. Cardiologia-UTIC, Dipartimento di Scienze Cardiovascolari, AOU delle Marche, Ancona, Italy
| | - Annamaria Iorio
- S.S.D. Chirurgia dei Trapianti e del Trattamento Chirurgico dello Scompenso, Dipartimento Cardiovascolare, ASST Papa Giovanni XXIII, Piazza OMS 1, Bergamo 24127, Italy
| | - Andrea Mortara
- Dipartimento di Cardiologia Clinica, Policlinico di Monza, Monza, Italy
| | - Gian Francesco Mureddu
- U.O.S.D. Cardiologia Riabilitativa, Azienda Ospedaliera San Giovanni Addolorata, Rome, Italy
| | - Filippo Zilio
- U.O. Cardiologia, Ospedale Santa Chiara, Trento, Italy
| | - Cristina Chimenti
- Dipartimento di Scienze Cliniche, Internistiche, Anestesiologiche e Cardiovascolari, Azienda Ospedaliera Policlinico Umberto I, Sapienza Università di Roma, Rome, Italy
| | - Manlio Gianni Cipriani
- Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (ISMETT)-IRCCS, Palermo, Italy
| | - Michele Senni
- S.S.D. Chirurgia dei Trapianti e del Trattamento Chirurgico dello Scompenso, Dipartimento Cardiovascolare, ASST Papa Giovanni XXIII, Piazza OMS 1, Bergamo 24127, Italy
| | - Claudio Bilato
- U.O.C. Cardiologia, Ospedali dell’Ovest Vicentino, Azienda ULSS 8 Berica, Vicenza, Italy
| | | | - Giovanna Geraci
- U.O.C. Cardiologia, Presidio Ospedaliero Sant’Antonio Abate, ASP Trapani, Erice, TP, Italy
| | - Vittorio Pascale
- UTIC-Emodinamica e Cardiologia Interventistica, Ospedale Civile Pugliese, Catanzaro, Italy
| | - Carmine Riccio
- U.O.S.D. Follow-up del Paziente Post-Acuto, Dipartimento Cardio-Vascolare, AORN Sant’Anna e San Sebastiano, Caserta, Italy
| | | | - Emanuele Tizzani
- Dipartimento di Cardiologia, Ospedale degli Infermi, Rivoli, TO, Italy
| | - Michele Massimo Gulizia
- U.O.C. Cardiologia, Ospedale Garibaldi-Nesima, Azienda di Rilievo Nazionale e Alta Specializzazione ‘Garibaldi’, Catania, Italy
| | - Federico Nardi
- Dipartimento di Cardiologia, Ospedale Santo Spirito, Casale Monferrato, AL, Italy
| | - Domenico Gabrielli
- U.O.C. Cardiologia, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, Rome, Italy
- Fondazione per il Tuo cuore—Heart Care Foundation, Florence, Italy
| | - Furio Colivicchi
- U.O.C. Cardiologia Clinica e Riabilitativa, Presidio Ospedaliero San Filippo Neri—ASL Roma 1, Rome, Italy
| | - Massimo Grimaldi
- U.O.C. Cardiologia-UTIC, Ospedale Miulli, Acquaviva delle Fonti, BA, Italy
| | - Fabrizio Oliva
- Fondazione per il Tuo cuore—Heart Care Foundation, Florence, Italy
- Cardiologia 1-Emodinamica, Dipartimento Cardiotoracovascolare ‘A. De Gasperis’, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Associazione Nazionale Medici Cardiologi Ospedalieri (ANMCO), Florence, Italy
| |
Collapse
|
32
|
Lecointe J, Gan S, Tripathi D, Ichimura S, Clouthier KL, Kushwaha A, Mercer-Rosa L, Reddy S. Plasma Proteomics of the Fontan Circulation Reveal Signatures of Oxidative Stress and Cell Death. Circ Heart Fail 2025; 18:e012136. [PMID: 40235440 PMCID: PMC12084025 DOI: 10.1161/circheartfailure.124.012136] [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: 06/20/2024] [Accepted: 03/24/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND Single ventricle congenital heart disease like hypoplastic left heart syndrome with a Fontan circulation constitutes, the largest group of children hospitalized with circulation failure, experiencing an in-hospital mortality rate of 20% to 50%. We investigated the mechanisms leading to Fontan failure to identify novel therapeutic targets. METHODS Blood was collected from patients with hypoplastic left heart syndrome post-Fontan and controls (n=6/group). Plasma microvesicles were isolated, and proteomics assessed using data-independent acquisition mass spectroscopy. Dysregulated proteins with a fold change >1.5 or ≤1.5, P<0.05, were evaluated using the Database for Annotation, Visualization, and Integrated Discovery and Ingenuity pathway analysis. Correlation of highly dysregulated proteins was assessed with New York Heart Association class, right ventricular fractional area change, oxygen saturation, and hemoglobin. RESULTS The age of Fontan patients versus controls was 16.0±2.1 versus 15.3±2.2. Three of 6 Fontan patients were in New York Heart Association class II, and 3 of 6 were in New York Heart Association III/IV; 4 of 6 had Fontan-associated liver disease. Overall, 72 proteins were upregulated, and 187 proteins were downregulated in Fontan failure. Proteins upregulated in Fontan failure predicted cell death pathways (Solute carrier family 2, Angiotensinogen, CD14) and mitochondrial reactive oxygen species signaling (ATP5F1A, S100A8); downregulated proteins predicted impaired cell survival (tyrosine-protein kinase, endothelial growth factors) and mitochondrial antioxidant enzymes (GPX1, PRDX5) Increasing expression of the following proteins was associated with worsening New York Heart Association class, ventricular function and cyanosis: complement system (C1QA, r=0.91), mitochondrial reactive oxygen species generation (HSPD1, r=0.81; ATP5F1A, r=0.75), and cytoskeletal proteins (ANK1, r=0.63; ACTN1, r=0.76). CONCLUSIONS Proteins from circulating microvesicles from patients with hypoplastic left heart syndrome post-Fontan are mostly from the liver. While this pilot study is limited by its sample size and may not represent the broader Fontan population, the proteomic changes were associated with worsening heart failure and cyanosis, suggesting their potential utility as biomarkers.
Collapse
Affiliation(s)
| | - Sushrima Gan
- Department of Pediatrics (Cardiology) and Cardiovascular Institute (S.G., S.I., A.K., S.R.), Stanford University, Palo Alto, CA
| | - Dipti Tripathi
- Department of Surgery (D.T.), Stanford University, Palo Alto, CA
| | - Shoko Ichimura
- Department of Pediatrics (Cardiology) and Cardiovascular Institute (S.G., S.I., A.K., S.R.), Stanford University, Palo Alto, CA
| | - Katie L. Clouthier
- Department of Anesthesia Critical Care Medicine, Children’s Hospital Los Angeles, CA (K.L.C.)
| | - Ankit Kushwaha
- Department of Pediatrics (Cardiology) and Cardiovascular Institute (S.G., S.I., A.K., S.R.), Stanford University, Palo Alto, CA
| | | | - Sushma Reddy
- Department of Pediatrics (Cardiology) and Cardiovascular Institute (S.G., S.I., A.K., S.R.), Stanford University, Palo Alto, CA
| |
Collapse
|
33
|
Kawano Y, Weber BN, Weisenfeld D, Jeffway MI, Cai T, McDermott GC, Liu Q, Sparks JA, Stuart J, Joseph J, Cai T, Liao KP. Risk of Incident Heart Failure and Heart Failure Subtypes in Patients With Rheumatoid Arthritis. Arthritis Care Res (Hoboken) 2025; 77:631-639. [PMID: 39651569 PMCID: PMC12040578 DOI: 10.1002/acr.25481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 11/01/2024] [Accepted: 12/02/2024] [Indexed: 12/11/2024]
Abstract
OBJECTIVE Patients with rheumatoid arthritis (RA) are at increased risk of cardiovascular disease (CVD) including heart failure (HF). However, little is known regarding the relative risks of HF subtypes such as HF with preserved ejection fraction (HFpEF) or reduced ejection fraction (HFrEF) in RA compared with non-RA. METHODS We identified patients with RA and matched non-RA comparators among participants consenting to broad research from two large academic centers. We identified incident HF and categorized HF subtypes based on EF closest to the HF incident date. Covariates included age, sex, and established CVD risk factors. Cox proportional hazards models were used to estimate the hazard ratios (HRs) for incident HF and HF subtypes. RESULTS We studied 1,445 patients with RA and 4,335 matched non-RA comparators (mean age 51.4 and 51.7 years, respectively; 78.7% female). HFpEF was the most common HF subtype in both groups (65% in RA vs 59% in non-RA). Patients with RA had an HR of 1.79 (95% confidence interval [CI] 1.38-2.32) for incident HF compared with those without RA after adjusting for CVD risk factors. Patients with RA had a higher rate of HFpEF (HR 1.99, 95% CI 1.43-2.77), but there was no statistical difference in the HFrEF rate (HR 1.45, 95% CI 0.81-2.60). CONCLUSION RA was associated with a higher rate of HF overall compared with non-RA, even after adjustment for established CVD risk factors. The elevated risk was driven by HFpEF, supporting a role for inflammation in HFpEF and highlighting potential opportunities to address this excess risk in RA.
Collapse
Affiliation(s)
- Yumeko Kawano
- Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Brittany N. Weber
- Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | | | | | | | | | - Qing Liu
- Brigham and Women’s Hospital, Boston, MA
| | - Jeffrey A. Sparks
- Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Jennifer Stuart
- Brigham and Women’s Hospital, Boston, MA
- Harvard T. H. Chan School of Public Health, Boston, MA
| | - Jacob Joseph
- Veteran’s Affairs Healthcare System, Boston, MA
- Veterans Affairs Healthcare System, Providence, RI
- Brown University, Providence, RI, USA
| | - Tianxi Cai
- Harvard T. H. Chan School of Public Health, Boston, MA
- Veteran’s Affairs Healthcare System, Boston, MA
| | - Katherine P. Liao
- Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Veteran’s Affairs Healthcare System, Boston, MA
| |
Collapse
|
34
|
Gu H, Simpson JM, Cansick J, Finlay E, Gilbert R, Lunn A, Maxwell H, Morgan H, Shenoy M, Shroff R, Subramaniam P, Tizard J, Tse Y, Chowienczyk P, Sinha MD, HOT-KID study. An exploratory analysis on diastolic function in the intensive compared with less intensive blood pressure control to prevent adverse cardiac remodelling in children with chronic kidney disease (HOT-KID): a parallel-group, open-label, multicentre, randomised, controlled trial. EBioMedicine 2025; 115:105691. [PMID: 40262383 PMCID: PMC12052684 DOI: 10.1016/j.ebiom.2025.105691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 03/05/2025] [Accepted: 03/26/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Relationship between blood pressure (BP) control and left ventricular (LV) diastolic function in children with chronic kidney disease (CKD) is uncertain. The aim of this study is to investigate whether achieving lower BP yields a favourable impact on diastolic function. METHODS We performed an exploratory analysis in the HOT-KID, a parallel group, open-label, multicentre, randomised, controlled trial (ISRCTN25006406). Children with CKD were randomised to standard (50th-75th percentile) or intensive (<40th percentile) standardised office systolic BP targets. Echocardiograms were performed at baseline and at follow-up visits. Diastolic function was assessed by early (E) and late mitral inflow (A) E/A ratio, mitral annular motion of myocardial relaxation (e') and atrial contraction (a') velocity, LV compliance of E/e' and e'/a' ratio, and left atrial volume index (LAVi) by a blinded observer. FINDINGS There was a difference in the average annual rate of change in E/A ratio (difference in means -0·07 per year, 95% CI: -0·14 to -0·01), septal e' (difference in means -0·003 m/s per year, 95% CI: -0·005 to 0·001), and LAVi (difference in means 0·82 ml/m2 per year, 95% CI: 0·22-1·42) in the standard (n = 60) compared to the intensive treatment arm (n = 64). However, the average annual changes in all other diastolic function measures were similar between standard and intensive treatment groups. There was no difference for overall adverse events or serious adverse events between the two treatment groups. INTERPRETATION Our exploratory analysis in a small, open label RCT suggests that achieving lower blood pressure may favourably impact some measures of LV diastolic function in children with CKD. FUNDING British Heart Foundation (PG/11/90/28,994); The authors MDS, PJC acknowledge financial support from the Department of Health via the National Institute for Health Research (NIHR) comprehensive Biomedical Research Centre and Clinical Research Facilities awards to Guy's and St Thomas' NHS Foundation Trust in partnership with King's College London and King's College Hospital NHS Foundation Trust. There are no relationships with industry.
Collapse
Affiliation(s)
- Haotian Gu
- King's College London British Heart Foundation Centre, London, UK.
| | - John M Simpson
- King's College London British Heart Foundation Centre, London, UK; Department of Paediatric Cardiology, Evelina London Children's Hospital, Guy's & St Thomas NHS Foundation Trust, Westminster Bridge Road, London SE1 7EH, UK
| | - Janette Cansick
- Department of Paediatrics, Medway Maritime Hospital, Medway, UK
| | - Eric Finlay
- Department of Paediatric Nephrology, Leeds General Infirmary, Leeds, UK
| | - Rodney Gilbert
- Department of Paediatric Nephrology, Southampton General Hospital, Southampton, UK
| | - Andrew Lunn
- Department of Paediatric Nephrology, Nottingham University Hospital NHS Trust, Nottingham, UK
| | - Heather Maxwell
- Department of Paediatric Nephrology, Royal Hospital for Sick Children, Glasgow, UK
| | - Henry Morgan
- Department of Paediatric Nephrology, Alder Hey Children's Hospital, Liverpool, UK
| | - Mohan Shenoy
- Department of Paediatric Nephrology, Royal Manchester Children's Hospital, Manchester, UK
| | - Rukshana Shroff
- Department of Paediatric Nephrology, UCL Great Ormond Street Hospital and Institute of Child Health, London, UK
| | | | - Jane Tizard
- Department of Paediatric Nephrology, Bristol Royal Hospital for Children, Bristol, UK
| | - Yincent Tse
- Department of Paediatric Nephrology, Great North Children's Hospital, Newcastle Upon Tyne, UK
| | - Phil Chowienczyk
- King's College London British Heart Foundation Centre, London, UK
| | - Manish D Sinha
- King's College London British Heart Foundation Centre, London, UK; Department of Paediatric Nephrology, Evelina London Children's Hospital, Guy's & St Thomas NHS Foundation Trust, Westminster Bridge Road, London SE1 7EH, UK.
| | | |
Collapse
Collaborators
Janette Cansick, Abdel Douiri, Eric Finlay, Rodney Gilbert, Haotian Gu, Larissa Kerecuk, Andrew Lunn, Heather Maxwell, Henry Morgan, Reza Razavi, Mohan Shenoy, Rukshana Shroff, Pushpa Subramaniam, Jane Tizard, Yincent Tse, Poothirikovil Venugopalan, John Simpson, Phil Chowienczyk, Manish Sinha,
Collapse
|
35
|
Koppikar S, Kuriya B, Udell JA, Yu B, Chu A, Lee DS, Widdifield J, Eder L. Risk of Heart Failure in Patients With Immune-Mediated Inflammatory Diseases: A Population-Based Study. J Rheumatol 2025; 52:489-497. [PMID: 40041997 DOI: 10.3899/jrheum.2024-0866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2025] [Indexed: 03/19/2025]
Abstract
OBJECTIVE To evaluate the risk of heart failure (HF) in patients with immune-mediated inflammatory diseases (IMIDs) compared to the general population with and without diabetes mellitus (DM). METHODS A population-based cohort study was conducted in patients with rheumatoid arthritis (RA), radiographic axial spondyloarthritis (r-axSpA), psoriatic arthritis, and psoriasis (PsO) in Ontario from 2011 until 2019. The study outcome was first hospitalization for HF. Incidence rates of HF were calculated for each cohort. Hazard ratios (HRs) for HF were calculated using Cox proportional hazard models. The etiology of HF was descriptively classified into mutually exclusive groups based on comorbidities during HF hospitalization. RESULTS A total of 243,061 patients with IMID, 748,517 with DM, and 8,278,934 non-IMID, non-DM controls were analyzed. The crude incidence rate for HF in IMID was 2.70 per 1000 person-years, with the highest rate in RA and lowest in r-axSpA. The risk of being hospitalized for HF was higher in IMID compared with non-IMID comparators (HR 1.34, 95% CI 1.30-1.38). This risk was highest among patients with RA (HR 1.61, 95% CI 1.54-1.68) and lowest in PsO (HR 1.09, 95% CI 1.03-1.15). In comparison, the risk of HF hospitalization in patients with DM was higher (HR 2.19, 95% CI 2.16-2.21). The most common antecedent comorbidity associated with HF in all patients with IMID was ischemic heart disease. In patients with IMID without DM, atrial fibrillation had a similar effect as ischemic heart disease. CONCLUSION The risk of HF hospitalization is increased in patients with IMID compared to the general population.
Collapse
Affiliation(s)
- Sahil Koppikar
- S. Koppikar, MD, Lihi Eder, MD, PhD, Department of Medicine, University of Toronto, and Women's College Hospital, University of Toronto, Toronto
| | - Bindee Kuriya
- B. Kuriya, MD, Department of Medicine, University of Toronto, and Sinai Health System, University of Toronto, and ICES, Toronto
| | - Jacob A Udell
- J.A. Udell, MD, Department of Medicine, University of Toronto, Toronto, Women's College Hospital, University of Toronto, and Peter Munk Cardiac Centre, University Health Network, and ICES, Toronto
| | - Bing Yu
- B. Yu, PhD, A. Chu, MHSc, ICES, Toronto
| | - Anna Chu
- B. Yu, PhD, A. Chu, MHSc, ICES, Toronto
| | - Douglas S Lee
- D.S. Lee, MD, PhD, Department of Medicine, University of Toronto, and Peter Munk Cardiac Centre, University Health Network, and ICES, Toronto
| | - Jessica Widdifield
- J. Widdifield, PhD, ICES, Toronto, and Institute of Health Policy Management and Evaluation, University of Toronto, and Sunnybrook Health Sciences Centre, Research Institute, Toronto, Ontario, Canada
| | - Lihi Eder
- S. Koppikar, MD, Lihi Eder, MD, PhD, Department of Medicine, University of Toronto, and Women's College Hospital, University of Toronto, Toronto;
| |
Collapse
|
36
|
Suthahar N, Mourmans SGJ, Achten A, Aboumsallem JP, Meijers WC, Bomer N, Kardys I, Gansevoort RT, Bakker SJL, Weerts J, Eringa EC, Damman K, van Empel V, de Boer RA. Peroxiredoxin-4, a marker of systemic oxidative stress, is associated with incident heart failure. Eur J Heart Fail 2025; 27:905-911. [PMID: 40189227 DOI: 10.1002/ejhf.3653] [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: 01/13/2025] [Revised: 02/25/2025] [Accepted: 03/17/2025] [Indexed: 05/27/2025] Open
Abstract
AIMS Oxidative stress is known to be involved in the pathophysiology of heart failure (HF). To assess oxidative stress, direct quantification of reactive oxygen species would be ideal but this is not feasible due to their short half-lives. Antioxidant enzymes such as peroxiredoxins, produced as a direct response to oxidative stress, mirror the process and can be more easily quantified. The aim of this study was to examine whether circulating peroxiredoxin-4 (Prx4), a marker of systemic oxidative stress, associates with incident HF and its subtypes. METHODS AND RESULTS We included a total of 8199 individuals from the Prevention of REnal and Vascular End-stage Disease (PREVEND) community-based cohort (mean age: 49.8 years; 50.1% women). During a median follow-up of 12.6 years, 349 (4.3%) HF events occurred of which 118 (33.8%) had HF with preserved ejection fraction. In a Cox proportional hazards model adjusting for age, sex, smoking, diabetes, hypertension, obesity, total and high-density lipoprotein cholesterol, cholesterol-lowering medication and renal disease, Prx4 was significantly associated with incident HF (hazard ratio [HR] per 1 standard deviation increase in log-Prx4: 1.22; 95% confidence interval [CI] 1.09-1.36; p < 0.001). Among HF subtypes, Prx4 remained associated with incident HF with preserved (HR 1.27; 95% CI 1.05-1.53) as well as reduced ejection fraction (HR 1.19; 95% CI 1.04-1.37), with no significant difference between the subtypes (p = 0.64). CONCLUSION Circulating Prx4 associates with the risk of developing HF, both with preserved and reduced ejection fraction. Future studies should examine whether Prx4 can serve as a real-time marker of oxidative stress status.
Collapse
Affiliation(s)
- Navin Suthahar
- Department of Cardiology, Erasmus MC, Cardiovascular Institute, Thorax Center, Rotterdam, The Netherlands
| | - Sanne G J Mourmans
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Anouk Achten
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Joseph Pierre Aboumsallem
- Department of Cardiology, Erasmus MC, Cardiovascular Institute, Thorax Center, Rotterdam, The Netherlands
| | - Wouter C Meijers
- Department of Cardiology, Erasmus MC, Cardiovascular Institute, Thorax Center, Rotterdam, The Netherlands
| | - Nils Bomer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Isabella Kardys
- Department of Cardiology, Erasmus MC, Cardiovascular Institute, Thorax Center, Rotterdam, The Netherlands
| | - Ron T Gansevoort
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Stephan J L Bakker
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jerremy Weerts
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Etto C Eringa
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Department of Physiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Kevin Damman
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Vanessa van Empel
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Rudolf A de Boer
- Department of Cardiology, Erasmus MC, Cardiovascular Institute, Thorax Center, Rotterdam, The Netherlands
| |
Collapse
|
37
|
Palmiero P, Caretto P, Ciccone MM, Maiello M, on behalf of the I.C.I.S.C.U. (Italian Chapter of International Society Cardiovascular Ultrasound). Long-Term Cardiovascular Risk and Maternal History of Pre-Eclampsia. J Clin Med 2025; 14:3121. [PMID: 40364153 PMCID: PMC12072551 DOI: 10.3390/jcm14093121] [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: 03/08/2025] [Revised: 04/22/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Pre-eclampsia is a severe pregnancy complication affecting 5-8% of pregnancies worldwide, marked by high blood pressure and organ damage typically occurring after 20 weeks of gestation. It is a leading cause of maternal and fetal morbidity and mortality. Though its exact cause is unknown, it involves placental abnormalities and improper blood vessel development. Risk factors include a history of pre-eclampsia, chronic hypertension, diabetes, obesity, and autoimmune disorders. Symptoms include high blood pressure, proteinuria, headaches, vision changes, and abdominal pain. Untreated, it can lead to seizures, stroke, preterm birth, or death. Delivery is the definitive treatment, with management strategies such as monitoring and blood pressure control. Pre-eclampsia significantly increases long-term cardiovascular disease (CVD) risks, including hypertension, ischemic heart disease, and stroke, linked to shared mechanisms like endothelial dysfunction and inflammation. Women with severe or recurrent pre-eclampsia have heightened risks, often developing chronic hypertension within a decade postpartum. It also impacts offspring, with daughters at elevated risk for pre-eclampsia and CVD. Hypertensive disorders of pregnancy, including pre-eclampsia, induce changes like left ventricular hypertrophy and diastolic dysfunction, raising risks for heart failure with preserved ejection fraction and coronary atherosclerosis. Overlapping with peripartum cardiomyopathy, pre-eclampsia underscores a spectrum of pregnancy-related cardiovascular disorders. Long-term monitoring and lifestyle interventions are crucial for managing risks, with research into genetic and biological mechanisms offering the potential for targeted prevention.
Collapse
Affiliation(s)
- Pasquale Palmiero
- ASL Brindisi, Cardiology Equipe, District of Brindisi, 72100 Brindisi, Italy;
- Medical School, University of Bari, 70122 Bari, Italy
| | - Pierpaolo Caretto
- University Cardiology Unit, Interdisciplinary Department of Medicine, Polyclinic University Hospital, 70124 Bari, Italy; (P.C.); (M.M.C.)
| | - Marco Matteo Ciccone
- University Cardiology Unit, Interdisciplinary Department of Medicine, Polyclinic University Hospital, 70124 Bari, Italy; (P.C.); (M.M.C.)
| | - Maria Maiello
- ASL Brindisi, Cardiology Equipe, District of Brindisi, 72100 Brindisi, Italy;
| | | |
Collapse
|
38
|
Duan X, Zhang X, Sun B. The landscape of novel antidiabetic drugs in diabetic HFpEF: relevant mechanisms and clinical implications. Cardiovasc Diabetol 2025; 24:186. [PMID: 40295996 PMCID: PMC12038999 DOI: 10.1186/s12933-025-02750-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/19/2025] [Indexed: 04/30/2025] Open
Abstract
As a heterogeneous syndrome, heart failure with preserved ejection fraction (HFpEF) has become the leading form of heart failure worldwide. Increasing evidence has identified that diabetes mellitus (DM) increases the risk of HFpEF. Worse still, the coexistence of both diseases poses a great threat to human health by further worsening the cardiovascular system and accelerating the progression of diabetes. Although several studies have indicated that the novel antidiabetic drugs, including sodium glucose cotransporter 2 inhibitors (SGLT2i), glucagon-like peptide-1 receptor agonists (GLP-1 RA) and dipeptidyl peptidase 4 inhibitors (DPP4i) provide the cardiovascular benefits in T2DM patients with HFpEF, the elaborated roles and mechanisms are not fully understood. In this review, we summarize the state-of-the-art evidence regarding the epidemiology and pathophysiology of diabetic HFpEF, and the landscape of the novel antidiabetic drugs in the treatment of diabetic HFpEF, as well as discuss the relevant mechanisms, aiming to broaden the understanding of diabetic HFpEF and gain new insight into the treatment of this disease.
Collapse
Affiliation(s)
- Xiangling Duan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, No. 139, People's Middle Street, Changsha, 410011, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Xiaomeng Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Bao Sun
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, No. 139, People's Middle Street, Changsha, 410011, China.
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
39
|
Zhu T, Song Y. Efficacy of Sacubitril/Valsartan Combined With Metoprolol on Cardiac Function, Cardiac Remodeling, and Endothelial Function in Patients With Coronary Heart Disease and Heart Failure. Br J Hosp Med (Lond) 2025; 86:1-16. [PMID: 40265535 DOI: 10.12968/hmed.2025.0120] [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: 04/24/2025]
Abstract
Aims/Background Coronary heart disease (CHD) combined with heart failure results in a rapidly progressing disease with an acute onset, posing a significant threat to a patient's survival. Metoprolol, a β-blocker, is effective in treating heart failure; however, due to its complex pathogenesis, the efficacy of monotherapy in managing disease progression remains suboptimal. Sacubitril/valsartan, an angiotensin II receptor antagonist, is another widely used drug for treating heart failure. The combination of the two drugs may play a synergistic role in effectively managing heart failure through different mechanisms. This study aims to investigate the effects of sacubitril/valsartan combined with metoprolol on cardiac function, cardiac remodeling, and endothelial function in patients with CHD and heart failure. Methods This retrospective analysis included 138 CHD patients combined with heart failure who received care at Linhai Hospital of Traditional Chinese Medicine between January 2022 and January 2024. Based on the treatment regimen, patients were divided into two groups. Patients receiving metoprolol monotherapy were included in the Metoprolol group (n = 61), while those receiving a combination of sacubitril/valsartan and metoprolol were assigned to the Combination group (n = 77). The cardiac function [New York Heart Association (NYHA) cardiac function classification], myocardial injury markers [serum cardiac troponin I (cTnI), N-terminal pro-brain natriuretic peptide (NT-proBNP)], cardiac remodeling function [left ventricular ejection fraction (LVEF), left ventricular end-diastolic diameter (LVEDD), left ventricular end-systolic diameter (LVESD)], endothelial function [serum endothelin-1 (ET-1), nitric oxide (NO)] were compared between these two groups before treatment and 3 months post-treatment. Additionally, the two groups were comparatively assessed for the incidence of adverse reactions during the treatment period. Results Following treatment, the NYHA cardiac function grading was significantly improved in the Combination group than in the Metoprolol group (p = 0.014). After treatment, the Combination group demonstrated significantly lower serum cTnI and NT-proBNP levels than the Metoprolol group (p < 0.05). After treatment, the Combination group had substantially higher LVEF and lower LVEDD and LVESD than the Metoprolol group (p < 0.05). Furthermore, the Combination group showed a significant decrease in serum ET-1 levels and an increase in serum NO levels compared to the Metoprolol group (p < 0.05). During the treatment period, there was no significant difference in the incidence of adverse reactions between the two groups (p > 0.05). Conclusion Sacubitril/valsartan combined with metoprolol is a safe, effective, and viable treatment option for patients with CHD combined with heart failure. This combination therapy may further improve cardiac and endothelial function by reducing cardiac remodeling, without increasing the risk of adverse reactions. This study offers a new drug combination regimen (sacubitril/valsartan combined with metoprolol) for patients with CHD combined with heart failure. This regimen further improves the cardiac and endothelial function, inhibits cardiac remodeling, and has good safety.
Collapse
Affiliation(s)
- Tongyu Zhu
- Department of Internal Medicine, Linhai Hospital of Traditional Chinese Medicine, Linhai, Zhejiang, China
| | - Yingjing Song
- Department of Cardiovascular Medicine, Linhai Hospital of Traditional Chinese Medicine, Linhai, Zhejiang, China
| |
Collapse
|
40
|
Mourmans SGJ, Achten A, Hermans R, Scheepers MJE, D'Alessandro E, Swennen G, Woudstra J, Appelman Y, Goor HV, Schalkwijk C, Knackstedt C, Weerts J, Eringa EC, van Empel VPM. The effect of empagliflozin on peripheral microvascular dysfunction in patients with heart failure with preserved ejection fraction. Cardiovasc Diabetol 2025; 24:182. [PMID: 40281528 PMCID: PMC12023568 DOI: 10.1186/s12933-025-02679-8] [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: 12/10/2024] [Accepted: 03/10/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Empagliflozin is an effective treatment for heart failure with preserved ejection fraction (HFpEF), but its definite mechanism of action is unclear. Systemic microvascular dysfunction strongly relates to HFpEF aetiology, and we hypothesised that empagliflozin improves microvascular function in HFpEF. OBJECTIVE To investigate the effect of the sodium-glucose cotransporter-2 inhibitor empagliflozin on peripheral microvascular function in HFpEF. METHODS This is a pre-post intervention study in patients diagnosed with HFpEF who are eligible for treatment with empagliflozin. Microvascular function assessment using laser speckle contrast analysis of the dorsal forearm during iontophoresis of vasoactive stimuli (acetylcholine, insulin sodium nitroprusside) was performed at baseline and after 3 months of empagliflozin treatment (10 mg daily). The primary outcome was the difference in blood flow measured in the forearm microvasculature between baseline and at follow-up (cutaneous vascular conductance, CVC). Secondarily we investigated quality-of-life based on the EQ-5D-5 L questionnaire at baseline and follow-up. RESULTS Twenty six patients finished the study according to protocol (mean age of 74 ± 7 years, 62% female). We observed a decreased blood flow response to acetylcholine after 3 months of empagliflozin (CVC: 0.77 ± 0.24 vs. 0.64 ± 0.20, p < 0.001). In contrast, the response to insulin improved (CVC: 0.61 ± 0.43 vs. 0.81 ± 0.32, p = 0.03), and the response to sodium nitroprusside remained stable after 3 months. No significant correlations were found between the changes in blood flow and quality of life. CONCLUSION This study shows that three months treatment with empagliflozin changed peripheral microvascular function in patients with HFpEF. Empagliflozin may enhance microvascular blood flow specifically via vascular actions of insulin, rather than a general effect on endothelial vasoregulation or smooth muscle cell function. As such, systemic microvascular dysfunction can be a modifiable factor in patients with HFpEF, while the clinical implications thereof warrant further investigations. TRIAL REGISTRATION The trial was preregistered at clinicaltrials.gov (NCT06046612).
Collapse
Affiliation(s)
- Sanne G J Mourmans
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| | - Anouk Achten
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| | - Raquel Hermans
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| | - Marijne J E Scheepers
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| | - Elisa D'Alessandro
- Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
| | - Geertje Swennen
- Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
| | - Janneke Woudstra
- Department of Cardiology, Amsterdam UMC Heart Centre, Amsterdam, The Netherlands
| | - Yolande Appelman
- Department of Cardiology, Amsterdam UMC Heart Centre, Amsterdam, The Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Casper Schalkwijk
- Department of Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| | - Christian Knackstedt
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| | - Jerremy Weerts
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| | - Etto C Eringa
- Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, The Netherlands
| | - Vanessa P M van Empel
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands.
| |
Collapse
|
41
|
Zhou W, Zhou L, Qi Z, Dai S, Zhang P, Zhong H, Xu H, Zhao X, Lian X, Lin J, Wu H. The soluble guanylate cyclase (sGC) stimulator vericiguat inhibits platelet activation and thrombosis. Eur J Pharmacol 2025; 999:177670. [PMID: 40287044 DOI: 10.1016/j.ejphar.2025.177670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 04/24/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Vericiguat, a soluble guanylate cyclase (sGC) stimulator, is used to treat chronic heart failure. Vericiguat can directly bind to the sGC in the absence of NO or stabilize the binding of NO to sGC, thereby stimulating cGMP production. Vericiguat correlates closely with the platelet activation. However, the precise effect of vericiguat on platelet activation and thrombosis in vivo remains to be elucidated. METHODS We investigated the effects of vericiguat on agonist-induced platelet aggregation, secretion, integrin αIIbβ3 activation, spreading, clot retraction, and thrombus formation in vivo, elucidating the underlying mechanisms. Additionally, we performed whole blood aggregometry and Microfluidic whole-blood perfusion assay to determine whether vericiguat could alleviate thrombosis. RESULTS Vericiguat concentration-dependently inhibited aggregation and ATP release induced by agonists both in human and mouse platelets. P-selection expression, integrin αIIbβ3 activation, spreading, and clot retraction induced by thrombin were all inhibited by vericiguat. Mechanistically, vericiguat bound to the sGC in platelets, avtivating the cGMP/PKG signaling pathway to inhibit the platelet. Vericiguat also inhibited the FeCl3-injured thrombus formation in mesenteric arterioles in wild-type (WT) mice and pulmonary vascular thrombi after constructing the pulmonary embolism model. Oral administration of vericiguat for 2 weeks attenuated thromboembolism in brain, too. CONCLUSION Vericiguat directly inhibits platelet activation and thrombosis in vivo by binding to the sGC and activating cGMP/PKG pathway. In addition to the treatment for chronic heart failure, it may have therapeutic advantages in treating thrombotic diseases.
Collapse
Affiliation(s)
- Wenxuan Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, China
| | - Luning Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, China
| | - Zhiyong Qi
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, China
| | - Shimo Dai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, China
| | - Peng Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, China
| | - Haoxuan Zhong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, China
| | - Huajie Xu
- Department of Infectious Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin Zhao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, China
| | - Xiaoyu Lian
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, China
| | - Jiaxiong Lin
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, China.
| | - Hongyi Wu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, China.
| |
Collapse
|
42
|
Ratchford SM, Clifton HL, Gifford JR, LaSalle DT, Thurston TS, Bunsawat K, Alpenglow JK, Wright JB, Amann M, Ryan JJ, Wray DW. Impact of Acute Antioxidant and Tetrahydrobiopterin (BH 4) Administration on Locomotor Muscle Microvascular Function in Patients With Heart Failure. Circ Heart Fail 2025:e012446. [PMID: 40270242 DOI: 10.1161/circheartfailure.124.012446] [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: 09/16/2024] [Accepted: 04/14/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Peripheral microvascular dysfunction is a hallmark feature of both heart failure with reduced ejection fraction (HFrEF) and preserved ejection fraction (HFpEF) pathophysiology, due partly to impairments in nitric oxide signaling secondary to tetrahydrobiopterin (BH4) deficiency and oxidative stress. METHODS Using a randomized, double-blind, placebo-controlled crossover design, this study examined the impact of enteral BH4 (10 mg/kg), an antioxidant cocktail (AOx), and coadministration of these 2 agents (BH4+AOx) on microvascular function in patients with HFrEF (n=14, 64±10 years) and HFpEF (n=19, 74±9 years). Passive limb movement was utilized to assess locomotor muscle microvascular function, and biomarkers of inflammation and oxidative damage were measured. RESULTS Compared with placebo, the peak change in leg blood flow was not statistically different after AOx administration (HFrEF, P=0.60; HFpEF, P=0.61), but improved following BH4 (P=0.033) and BH4+AOx (P=0.019) in both HFrEF (placebo: 234±31; BH4: 357±45; BH4+AOx: 355±49 mL/min) and HFpEF (placebo: 269±33; BH4: 367±47; BH4+AOx: 394±65 mL/min). The total hyperemic response to passive limb movement (leg blood flow area under the curve) was not statistically different across treatments in patients with HFrEF (P=0.29), but increased following BH4 (P=0.016) and BH4+AOx (P=0.040) in the HFpEF group. CRP (C-reactive protein) was lower following BH4 (P=0.007) and BH4+AOx (P=0.007) in HFpEF (placebo: 4268±547; BH4: 2721±391; BH4+AOx: 2779±376 ng/mL), but was not statistically different in HFrEF (P=0.39). CONCLUSIONS Together, these results provide new evidence for the efficacy of acute BH4 administration to improve some aspects of locomotor muscle microvascular function in patients with HFrEF and HFpEF, with no apparent benefit of AOx administration, alone or in combination with BH4, in either group. These findings lend further conceptual support for the nitric oxide pathway as a modifiable target in the treatment of heart failure.
Collapse
Affiliation(s)
- Stephen M Ratchford
- Geriatric Research, Education, and Clinical Center, George E. Wahlen VA Medical Center, Salt Lake City, UT (S.M.R., H.L.C., K.B., M.A., D.W.W.)
- Department of Internal Medicine, Division of Geriatrics (S.M.R., H.L.C., K.B., D.W.W.), University of Utah, Salt Lake City
| | - Heather L Clifton
- Geriatric Research, Education, and Clinical Center, George E. Wahlen VA Medical Center, Salt Lake City, UT (S.M.R., H.L.C., K.B., M.A., D.W.W.)
- Department of Internal Medicine, Division of Geriatrics (S.M.R., H.L.C., K.B., D.W.W.), University of Utah, Salt Lake City
| | - Jayson R Gifford
- Department of Exercise Sciences, Brigham Young University, Salt Lake City, UT (J.R.G.)
| | - D Taylor LaSalle
- Department of Nutrition and Integrative Physiology (D.T.L.S., T.S.T., J.K.A., D.W.W.), University of Utah, Salt Lake City
| | - Taylor S Thurston
- Department of Nutrition and Integrative Physiology (D.T.L.S., T.S.T., J.K.A., D.W.W.), University of Utah, Salt Lake City
| | - Kanokwan Bunsawat
- Geriatric Research, Education, and Clinical Center, George E. Wahlen VA Medical Center, Salt Lake City, UT (S.M.R., H.L.C., K.B., M.A., D.W.W.)
- Department of Internal Medicine, Division of Geriatrics (S.M.R., H.L.C., K.B., D.W.W.), University of Utah, Salt Lake City
| | - Jeremy K Alpenglow
- Department of Nutrition and Integrative Physiology (D.T.L.S., T.S.T., J.K.A., D.W.W.), University of Utah, Salt Lake City
| | - Josephine B Wright
- Division of Cardiovascular Medicine (J.B.W., J.J.R.), University of Utah, Salt Lake City
| | - Markus Amann
- Geriatric Research, Education, and Clinical Center, George E. Wahlen VA Medical Center, Salt Lake City, UT (S.M.R., H.L.C., K.B., M.A., D.W.W.)
- Department of Anesthesiology (M.A.) University of Utah, Salt Lake City
| | - John J Ryan
- Division of Cardiovascular Medicine (J.B.W., J.J.R.), University of Utah, Salt Lake City
| | - D Walter Wray
- Geriatric Research, Education, and Clinical Center, George E. Wahlen VA Medical Center, Salt Lake City, UT (S.M.R., H.L.C., K.B., M.A., D.W.W.)
- Department of Internal Medicine, Division of Geriatrics (S.M.R., H.L.C., K.B., D.W.W.), University of Utah, Salt Lake City
- Department of Nutrition and Integrative Physiology (D.T.L.S., T.S.T., J.K.A., D.W.W.), University of Utah, Salt Lake City
| |
Collapse
|
43
|
Malik MK, Kinno M, Liebo M, Yu MD, Syed M. Evolving role of myocardial fibrosis in heart failure with preserved ejection fraction. Front Cardiovasc Med 2025; 12:1573346. [PMID: 40336640 PMCID: PMC12055812 DOI: 10.3389/fcvm.2025.1573346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 04/07/2025] [Indexed: 05/09/2025] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a complex clinical diagnosis with a heterogeneous pathophysiology and clinical presentation. The hallmark of HFpEF is diastolic dysfunction associated with left ventricular remodeling and fibrosis. Myocardial interstitial fibrosis (MIF) occurs as the result of collagen deposition and is dependent on the underlying etiology of heart failure. Detection of MIF can be done by invasive histopathologic sampling or by imaging. More recently, novel biomarkers have been investigated as an alternative tool for not only the detection of MIF but also for the prognostication of patients with HFpEF which may in turn alleviate the need for invasive and expensive imaging in the future.
Collapse
Affiliation(s)
- Muhammad K. Malik
- Department of Internal Medicine, Loyola University Medical Center, Maywood, IL, United States
- Department of Cardiology, Baylor Scott & White, The Heart Hospital, Plano, TX, United States
| | - Menhel Kinno
- Division of Cardiology, Department of Internal Medicine, Loyola University Medical Center, Maywood, IL, United States
- Division of Cardiology, Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Max Liebo
- Division of Cardiology, Department of Internal Medicine, Loyola University Medical Center, Maywood, IL, United States
| | - Mingxi D. Yu
- Division of Cardiology, Department of Internal Medicine, Loyola University Medical Center, Maywood, IL, United States
| | - Mushabbar Syed
- Division of Cardiology, Department of Internal Medicine, Loyola University Medical Center, Maywood, IL, United States
| |
Collapse
|
44
|
Butler J, Hammonds K, Talha KM, Alhamdow A, Bennett MM, Bomar JVA, Ettlinger JA, Traba MM, Priest EL, Schmedt N, Zeballos C, Shaver CN, Afzal A, Widmer RJ, Gottlieb RL, Mack MJ, Packer M. Incident heart failure and recurrent coronary events following acute myocardial infarction. Eur Heart J 2025; 46:1540-1550. [PMID: 39874177 PMCID: PMC12011519 DOI: 10.1093/eurheartj/ehae885] [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: 03/27/2024] [Revised: 09/12/2024] [Accepted: 12/06/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND AND AIMS Recurrent myocardial infarction (MI) and incident heart failure (HF) are major post-MI complications. Herein, contemporary post-MI risks for recurrent MI and HF are described. METHODS A total of 6804 patients with a primary discharge diagnosis of MI at 28 Baylor Scott & White Health hospitals (January 2015 to December 2021) were studied. Patient characteristics, treatment, and outcomes, including incident HF, recurrent MI, all-cause death, and all-cause and cardiovascular rehospitalizations, were assessed. Landmark approach anchored at 3 months post-discharge was used to assess 1-year outcomes. RESULTS Median age was 69 years, 59.7% were male, and 76.7% had non-ST-elevation MI. Comorbidities included hypertension (89%), dyslipidaemia (87%), Type 2 diabetes (48%), and chronic kidney disease (34%); 17% had a history of MI and 23% of HF; 63% underwent percutaneous/surgical revascularization. In landmark-anchored 1-year outcomes (N = 6210), 413 (6.7%) patients died, 1730 (27.9%) had all-cause and 735 (11.8%) cardiovascular hospitalizations, 234 (3.8%) had recurrent MI. Of patients without history of HF, 1160 (23.8%) developed incident HF [42.2%, 26.7%, and 31.1% with ejection fraction (EF) < 40%, 41-49%, and >50%, respectively) within 3 months of discharge. Patients who developed HF had higher risk of death and hospitalizations (all P < .001), irrespective of EF. Of 2179 patients with EF > 50% without prevalent HF or HF during index hospitalization, 257 (11.8%) developed HF and 77 (3.5%) recurrent MI within 1 year. CONCLUSIONS In a contemporary post-MI cohort, the risk for incident HF was greater than recurrent MI, even among those with normal EF and no HF at discharge.
Collapse
Affiliation(s)
- Javed Butler
- Baylor Scott & White Research Institute, 3434 Live Oak St Ste 501, Dallas, TX 75204, USA
- Department of Medicine, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA
| | - Kendall Hammonds
- Baylor Scott & White Research Institute, 3434 Live Oak St Ste 501, Dallas, TX 75204, USA
| | - Khawaja M Talha
- Department of Medicine, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA
| | - Ayman Alhamdow
- Boehringer Ingelheim International GmbH, Binger Straße 173, Ingelheim, 55218 Ingelheim am Rhein, Germany
| | - Monica M Bennett
- Baylor Scott & White Research Institute, 3434 Live Oak St Ste 501, Dallas, TX 75204, USA
| | - J Vee Anne Bomar
- Baylor Scott & White Research Institute, 3434 Live Oak St Ste 501, Dallas, TX 75204, USA
| | - Jason A Ettlinger
- Baylor Scott & White Research Institute, 3434 Live Oak St Ste 501, Dallas, TX 75204, USA
| | - Monica Martinez Traba
- Boehringer Ingelheim International GmbH, Binger Straße 173, Ingelheim, 55218 Ingelheim am Rhein, Germany
| | - Elisa L Priest
- Baylor Scott & White Research Institute, 3434 Live Oak St Ste 501, Dallas, TX 75204, USA
| | - Niklas Schmedt
- Boehringer Ingelheim International GmbH, Binger Straße 173, Ingelheim, 55218 Ingelheim am Rhein, Germany
| | - Cecilia Zeballos
- Boehringer Ingelheim International GmbH, Binger Straße 173, Ingelheim, 55218 Ingelheim am Rhein, Germany
| | - Courtney N Shaver
- Baylor Scott & White Research Institute, 3434 Live Oak St Ste 501, Dallas, TX 75204, USA
| | - Aasim Afzal
- Departments of Cardiology and Cardiothoracic Surgery, Baylor Scott &White The Heart Hospital, 1100 Allied Dr, Plano, TX 75093, USA
- Center for Advanced Heart and Lung Disease and Baylor Heart and Vascular Institute, Baylor University Medical Center, 3410 Worth St, Ste 250, Dallas, TX 75226, USA
- Department of Medicine, Texas A&M Health Science Center, 3302 Gaston Avenue, Dallas, TX 75246, USA
| | - Robert J Widmer
- Department of Cardiology, Baylor Scott & White Medical Center—Temple, 2401 S 31st St, Temple, TX 76508, USA
| | - Robert L Gottlieb
- Baylor Scott & White Research Institute, 3434 Live Oak St Ste 501, Dallas, TX 75204, USA
- Departments of Cardiology and Cardiothoracic Surgery, Baylor Scott &White The Heart Hospital, 1100 Allied Dr, Plano, TX 75093, USA
- Center for Advanced Heart and Lung Disease and Baylor Heart and Vascular Institute, Baylor University Medical Center, 3410 Worth St, Ste 250, Dallas, TX 75226, USA
- Department of Medicine, Texas A&M Health Science Center, 3302 Gaston Avenue, Dallas, TX 75246, USA
- Department of Medicine, Burnett School of Medicine, Texas Christian University, 1100 W. Rosedale St., Fort Worth, TX 76104, USA
| | - Michael J Mack
- Baylor Scott & White Research Institute, 3434 Live Oak St Ste 501, Dallas, TX 75204, USA
- Departments of Cardiology and Cardiothoracic Surgery, Baylor Scott &White The Heart Hospital, 1100 Allied Dr, Plano, TX 75093, USA
| | - Milton Packer
- Center for Advanced Heart and Lung Disease and Baylor Heart and Vascular Institute, Baylor University Medical Center, 3410 Worth St, Ste 250, Dallas, TX 75226, USA
- Department of Medicine, Texas A&M Health Science Center, 3302 Gaston Avenue, Dallas, TX 75246, USA
- Department of Cardiology, Baylor Scott & White Medical Center—Temple, 2401 S 31st St, Temple, TX 76508, USA
- The Imperial College, London, UK
| |
Collapse
|
45
|
Lindberg F, Savarese G. Opportunities and challenges in preventing heart failure: when is risk modifiable? Eur Heart J 2025; 46:1537-1539. [PMID: 40036736 DOI: 10.1093/eurheartj/ehaf042] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/06/2025] Open
Affiliation(s)
- Felix Lindberg
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Gianluigi Savarese
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
46
|
Tatekoshi Y, Mahmoodzadeh A, Shapiro JS, Liu M, Bianco GM, Tatekoshi A, Camp SD, De Jesus A, Koleini N, De La Torre S, Wasserstrom JA, Dillmann WH, Thomson BR, Bedi KC, Margulies KB, Weinberg SE, Ardehali H. Protein O-GlcNAcylation and hexokinase mitochondrial dissociation drive heart failure with preserved ejection fraction. Cell Metab 2025:S1550-4131(25)00211-6. [PMID: 40267914 DOI: 10.1016/j.cmet.2025.04.001] [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: 10/27/2024] [Revised: 03/03/2025] [Accepted: 04/02/2025] [Indexed: 04/25/2025]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a common cause of morbidity and mortality worldwide, but its pathophysiology remains unclear. Here, we report a mouse model of HFpEF and show that hexokinase (HK)-1 mitochondrial binding in endothelial cells (ECs) is critical for protein O-GlcNAcylation and the development of HFpEF. We demonstrate increased mitochondrial dislocation of HK1 within ECs in HFpEF mice. Mice with deletion of the mitochondrial-binding domain of HK1 spontaneously develop HFpEF and display impaired angiogenesis. Spatial proximity of dislocated HK1 and O-linked N-acetylglucosamine transferase (OGT) causes increased OGT activity, shifting the balance of the hexosamine biosynthetic pathway intermediates into the O-GlcNAcylation machinery. EC-specific overexpression of O-GlcNAcase and an OGT inhibitor reverse angiogenic defects and the HFpEF phenotype, highlighting the importance of protein O-GlcNAcylation in the development of HFpEF. Our study demonstrates a new mechanism for HFpEF through HK1 cellular localization and resultant protein O-GlcNAcylation, and provides a potential therapy for HFpEF.
Collapse
Affiliation(s)
- Yuki Tatekoshi
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA
| | - Amir Mahmoodzadeh
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA
| | - Jason S Shapiro
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA
| | - Mingyang Liu
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA
| | - George M Bianco
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA
| | - Ayumi Tatekoshi
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA
| | - Spencer Duncan Camp
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA
| | - Adam De Jesus
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA
| | - Navid Koleini
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA
| | - Santiago De La Torre
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA
| | - J Andrew Wasserstrom
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA
| | - Wolfgang H Dillmann
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Benjamin R Thomson
- Feinberg Cardiovascular and Renal Research Institute and Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Kenneth C Bedi
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kenneth B Margulies
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Samuel E Weinberg
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Hossein Ardehali
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
47
|
Janssen-Telders C, Eringa EC, de Groot JR, de Man FS, Handoko ML. The role of epicardial adipose tissue remodelling in heart failure with preserved ejection fraction. Cardiovasc Res 2025:cvaf056. [PMID: 40238568 DOI: 10.1093/cvr/cvaf056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/12/2024] [Accepted: 01/22/2025] [Indexed: 04/18/2025] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a growing global health problem characterized by high morbidity and mortality, with limited effective therapies available. Obesity significantly influences haemodynamic and structural changes in the myocardium and vasculature, primarily through the accumulation and action of visceral adipose tissue. Particularly, epicardial adipose tissue (EAT) contributes to HFpEF through inflammation and lipotoxic infiltration of the myocardium. However, the precise signalling pathways leading to diastolic stiffness in HFpEF require further elucidation. This review explores the dynamic role of EAT in health and disease. Drawing upon insights from studies in other conditions, we discuss potential EAT-mediated inflammatory pathways in HFpEF and how they may contribute to functional and structural myocardial and endothelial derangements, including intramyocardial lipid infiltration, fibrosis, endothelial dysfunction, cardiomyocyte stiffening, and left ventricular hypertrophy. Lastly, we propose potential targets for novel therapeutic avenues.
Collapse
Affiliation(s)
- Carolina Janssen-Telders
- Department of Cardiology Amsterdam UMC, Heart Centre, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Etto C Eringa
- Amsterdam Cardiovascular Sciences, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
- Department of Physiology, Amsterdam UMC, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht UMC, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Joris R de Groot
- Department of Cardiology Amsterdam UMC, Heart Centre, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Frances S de Man
- Amsterdam Cardiovascular Sciences, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht UMC, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - M Louis Handoko
- Department of Cardiology Amsterdam UMC, Heart Centre, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Department of Pulmonology, Amsterdam UMC, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Department of Cardiology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
48
|
Hung TC, Chien SC, Tsao CH, Wu YW, Yen CH, Hsiao CC, Lin HC, Tsai JP, Peng MC, Tsai CT, Hung CL. The association of coronary microvascular dysfunction and cardiac maladaptation with clinical outcomes in heart failure with preserved ejection fraction: A prospective dynamic SPECT study. Int J Cardiol 2025; 425:133064. [PMID: 39952474 DOI: 10.1016/j.ijcard.2025.133064] [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: 12/03/2024] [Revised: 02/04/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Accumulating evidences have suggested a pathophysiological link between coronary microvascular dysfunction (CMD) and heart failure with preserved ejection fraction (HFpEF). The clinical relevance and prognostic impact of myocardial blood flow (MBF) using single-photon emission computed tomography (SPECT) among HFpEF patients remain unexplored clearly. METHODS Fifty-five patients with HFpEF and 48 controls were prospectively enrolled. All underwent dynamic myocardial perfusion imaging with (99 m) Tc-sestamibi at rest and a stress test at peak with dipyridamole. MBF and myocardial flow reserve (MFR) parameters were obtained after perfusion and further related to cardiac structural and advanced functional strain measures using echocardiography and clinical outcomes. RESULTS CMD (defined as global MFR <2.5) was more prevalent in HFpEF than in control (71 % vs 16.7 %). Patients with HFpEF had worse longitudinal strain indices, lower post-stress MBF (2.37 ± 0.78 vs. 3.30 ± 0.94 ml/min/g, p < 0.001), higher global rest MBF (1.11 ± 0.55 vs. 0.89 ± 0.19 ml/min/g, p = 0.008), and lower global MFR (2.34 ± 0.84 vs. 3.68 ± 1.04, p < 0.001) than the controls. These results were similar for the regional MFRs in the vascular-based analysis. Lower MFR correlated with more impaired left ventricular, left atrial, and right ventricular strain measures (r = -0.44, 0.38, and 0.27, respectively, all p < 0.05). Patients with HFpEF and CMD had increased risks of all-cause mortality and hospitalization compared with those without HFpEF nor CMD (adjusted hazard: 6.42, 95 % CI: 1.40-29.43, p = 0.017). CONCLUSIONS CMD by dynamic SPECT is frequently observed among patients with HFpEF and correlated with more impaired overall cardiac mechanics. HFpEF with comorbid CMD had worse clinical outcome.
Collapse
Affiliation(s)
- Ta-Chuan Hung
- Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan; MacKay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
| | - Shih-Chieh Chien
- Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan; Department of Nuclear Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan.
| | - Chin-Ho Tsao
- Department of Nuclear Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Yen-Wen Wu
- Department of Nuclear Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan; Division of Cardiology, Cardiovascular Medical Center, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Chih-Hsuan Yen
- Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chih-Chung Hsiao
- Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Hui-Chun Lin
- Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Jui-Peng Tsai
- Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Ming-Cheng Peng
- Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Cheng-Ting Tsai
- Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chung-Lieh Hung
- Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
49
|
Pedicino D, Volpe M. Weekly Journal Scan: a deep insight into aldosterone antagonism across the heart failure spectrum. Eur Heart J 2025:ehaf233. [PMID: 40233031 DOI: 10.1093/eurheartj/ehaf233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/17/2025] Open
Affiliation(s)
- Daniela Pedicino
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Catholic University of the Sacred Heart, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Massimo Volpe
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Via di Grottarossa 1035, Rome, Italy
- IRCCS San Raffaele Roma, Via di Valcannuta 250, Rome, Italy
| |
Collapse
|
50
|
Vranken NPA, Li X, Bouman H, Mourmans SGJ, Achten A, Barandiarán Aizpurua A, Brunner-La Rocca HP, Knackstedt C, van Empel VPM, Weerts J. Temporal prevalence and prognostic impact of diabetes mellitus and albuminuria in heart failure with preserved ejection fraction. Cardiovasc Diabetol 2025; 24:156. [PMID: 40188061 PMCID: PMC11972526 DOI: 10.1186/s12933-025-02708-6] [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: 02/20/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Most patients with heart failure with preserved ejection fraction (HFpEF) have a metabolic phenotype in which comorbidities including diabetes mellitus play an important role. Factors related to impaired glucose metabolism, such as kidney disease, may contribute to adverse clinical events. Albuminuria is an early marker of kidney disease. We assessed the prevalence of impaired glucose metabolism and albuminuria in HFpEF over time, and evaluated its prognostic implications. METHODS Consecutive patients referred to our outpatient clinic and diagnosed with HFpEF between March 2015-November 2023 were included in this study. Patients with type 1 diabetes were excluded. Patients were stratified according to baseline glucose metabolism status (DM + for prediabetes and diabetes, or DM-) and albuminuria status (ALB+ or ALB- for albuminuria > 3.0 mg/mmol and normoalbuminuria, respectively). The primary outcome was a composite of HF hospitalizations (HFH) and all-cause mortality, and was analysed using multivariable-adjusted Cox-regression models. RESULTS Among 332 patients with HFpEF (median age 77 years; 67% female), 121 (36.4%) were classified as DM-/ALB-, 106 (31.9%) as DM+ /ALB-, 44 (13.3%) as DM-/ALB+, and 61 (18.4%) as DM+ /ALB+. Both baseline DM and ALB were independently associated with the primary outcome after approximately 3 years: adjusted hazard ratio (aHR) 1.93; 95% confidence interval (CI) 1.25-2.97 and 1.58; 95%CI 1.04-2.41, respectively. Patients in the DM+ /ALB+ group showed the highest risk (aHR 2.85; 95%CI 1.57-5.15). After one year, DM/ALB status was re-evaluated in 250 (75%) patients. New DM+ and ALB+ incidence was 3.9% and 22%in those at risk, respectively. Patients particularly changed ALB groups compared to baseline (n = 63, 25.2%); 27 (10.8%) patients recovered from albuminuria. At 3 years follow-up, the primary outcome mainly occurred in patients who consistently showed albuminuria (27.1%) or who recovered from albuminuria (22.2%), and less so in patients who developed albuminuria after one year (13.9%) or who remained free of albuminuria (8.6%) (p = 0.008). CONCLUSIONS DM and albuminuria are prevalent in HFpEF at baseline, and re-evaluation one year later still reveals new diagnoses. Both factors are independently associated with adverse outcomes. Albuminuria at any time point remains predictive of adverse outcomes in HFpEF. RESEARCH INSIGHTS WHAT IS CURRENTLY KNOWN ABOUT THIS TOPIC?: Diabetes mellitus is an important cardiovascular risk factor in patients with HFpEF, contributing to disease progression and worse outcomes. Albuminuria is a prognostic marker in heart failure patients and more prevalent in patients with diabetes WHAT IS THE KEY RESEARCH QUESTION?: What is prevalence of impaired glucose metabolism and albuminuria in HFpEF over time and how does this translate to prognosis? WHAT IS NEW?: Both DM and albuminuria each independently associated with worse prognosis in HFpEF. Screening 1 year after HFpEF diagnosis yielded incidence rates of 3.9% and 10.8% for DM and prediabetes, respectively, and 22% for albuminuria. Albuminuria at any time point appeared prognostic in HFpEF, also when albuminuria recovered HOW MIGHT THIS STUDY INFLUENCE CLINICAL PRACTICE?: Intermittent screening of HFpEF patients for abnormal glucose metabolism and albuminuria is warranted to optimize risk management.
Collapse
Affiliation(s)
- Nousjka P A Vranken
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre (MUMC+), PO Box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Xinyu Li
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre (MUMC+), PO Box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Heleen Bouman
- Division of Nephrology, Department of Internal Medicine, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| | - Sanne G J Mourmans
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre (MUMC+), PO Box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Anouk Achten
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre (MUMC+), PO Box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Arantxa Barandiarán Aizpurua
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre (MUMC+), PO Box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Hans-Peter Brunner-La Rocca
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre (MUMC+), PO Box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Christian Knackstedt
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre (MUMC+), PO Box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Vanessa P M van Empel
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre (MUMC+), PO Box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Jerremy Weerts
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre (MUMC+), PO Box 5800, 6202 AZ, Maastricht, The Netherlands.
| |
Collapse
|