1
|
Kawanami S, Egami Y, Abe M, Osuga M, Nohara H, Ukita K, Kawamura A, Yasumoto K, Okamoto N, Matsunaga‐Lee Y, Yano M, Nishino M. Randomized trial to assess worsening renal function by adding dapagliflozin for acute decompensated heart failure. ESC Heart Fail 2025; 12:2023-2033. [PMID: 39888286 PMCID: PMC12055397 DOI: 10.1002/ehf2.15212] [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] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 12/11/2024] [Accepted: 01/02/2025] [Indexed: 02/01/2025] Open
Abstract
AIMS Dapagliflozin (DAPA), a sodium-glucose co-transporter 2 inhibitor, has been shown to reduce cardiovascular mortality among patients with chronic heart failure. We aimed to evaluate the impact on a worsening renal function (WRF) by adding DAPA as compared to standard decongestive therapy with loop diuretics alone. METHODS AND RESULTS We enrolled 114 consecutive acute decompensated heart failure (ADHF) patients with a left ventricular ejection fraction (LVEF) of less than 50%. The patients were prospectively randomized to be assigned either to DAPA group who received DAPA at a dose of 10 mg once daily within 24 h after admission or conventional therapy group (CON group) who received loop diuretics alone. All patients were adjusted by increasing or decreasing the loop diuretic by 10 mg to maintain a 1-2 mL/kg/h urine output. The primary endpoint was the incidence of WRF, which was defined as an increase in the serum creatinine of ≥0.3 mg/dL from baseline. The median age of the patients was 77 [interquartile range (IQR): 64, 85] years, 35% were female and the median LVEF was 33 [IQR: 28, 38] %. There was no significant difference in the incidence of WRF between the two groups (16.1%, n = 9 vs. 12.1%, n = 7, P value = 0.54). The total dose of loop diuretics through day 7 was lower in the DAPA group than CON group (184 ± 79.5 mg vs. 214 ± 66.5 mg, P value = 0.03). CONCLUSIONS This randomized prospective trial revealed the addition of DAPA within 24 h after admission reduced the diuretic dose without WRF.
Collapse
Affiliation(s)
| | | | - Masaru Abe
- Division of CardiologyOsaka Rosai HospitalOsakaJapan
| | - Mizuki Osuga
- Division of CardiologyOsaka Rosai HospitalOsakaJapan
| | | | - Kohei Ukita
- Division of CardiologyOsaka Rosai HospitalOsakaJapan
| | | | - Koji Yasumoto
- Division of CardiologyOsaka Rosai HospitalOsakaJapan
| | | | | | | | | |
Collapse
|
2
|
Ferreira JP, Anker SD, Butler J, Filippatos G, Januzzi JL, Schueler E, Panova-Noeva M, Wetzel K, Prochaska J, Pocock SJ, Sattar N, Sumin M, Zannad F, Packer M. Effect of Empagliflozin on the Mechanisms Driving Erythropoiesis and Iron Mobilization in Patients With Heart Failure: The EMPEROR Program. J Am Coll Cardiol 2025; 85:1757-1770. [PMID: 40335252 DOI: 10.1016/j.jacc.2025.03.503] [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: 01/09/2025] [Revised: 03/05/2025] [Accepted: 03/12/2025] [Indexed: 05/09/2025]
Abstract
BACKGROUND Sodium-glucose cotransporter 2 (SGLT2) inhibitors stimulate erythropoiesis, but the mechanisms and clinical relevance of the effect of SGLT2 inhibitors on systemic iron metabolism in patients with heart failure is not well understood. OBJECTIVES The authors sought to characterize a comprehensive suite of iron metabolism biomarkers-particularly the erythroblast signaling molecule, erythroferrone-in patients with heart failure before and after short- and long-term treatment with empagliflozin in patients with heart failure and a reduced or preserved ejection fraction. METHODS We measured serum iron metabolism biomarkers at baseline, 12 weeks, and 52 weeks in 1,139 patients who were treated with placebo or empagliflozin in the EMPEROR (EMPagliflozin outcomE tRial in Patients With chrOnic heaRt Failure) program, and we characterized the inter-relationships of these biomarkers with clinical status and with the effect of empagliflozin on erythropoiesis and heart failure outcomes. RESULTS Correlations among iron biomarkers indicated the presence of a functional erythropoietin-erythroferrone-transferrin-receptor-protein-1 (TfR1)-hepcidin axis. As heart failure advanced, patients showed higher levels of erythropoietin, erythroferrone, and TfR1 (P trend <0.01), and levels of these proteins predicted a heightened risk of cardiovascular death or heart failure hospitalization (all P < 0.01). Compared with placebo, at 12 weeks, empagliflozin increased hemoglobin by 0.6 to 0.9 g/dL (P < 0.001), an effect that was accompanied by further activation of the erythropoietin-erythroferrone-TfR1 axis and increased iron use. Empagliflozin increased serum levels of erythroferrone by >40% (along with increases in erythropoietin and TfR1), while simultaneously decreasing hepcidin levels and reducing serum iron concentrations and transferrin saturation (all P < 0.01). When treated with empagliflozin, patients with evidence of iron deficiency at baseline showed attenuation of the erythrocytic response (P trend = 0.04) but no diminution of the heart failure benefits. CONCLUSIONS The erythropoietin-erythroferrone-TfR1-hepcidin axis is activated in patients with heart failure as the disease advances and is further heightened by SGLT2 inhibitors, in parallel with their effect to enhance erythropoiesis and iron mobilization and use. These changes have important implications for understanding the mechanism of action of SGLT2 inhibitors and for monitoring the response to treatment.
Collapse
Affiliation(s)
- João Pedro Ferreira
- UnIC@RISE, Cardiovascular Research and Development Center, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal; Heart Failure Clinic, Internal Medicine Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, Portugal; Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, and INSERM U1116, CHRU, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France.
| | - Stefan D Anker
- Department of Cardiology (CVK) and Berlin Institute of Health Center for Regenerative Therapies (BCRT), German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany; Institute of Heart Diseases, Wrocław Medical University, Wroclaw, Poland
| | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, Texas, USA; University of Mississippi, Jackson, Mississippi, USA
| | - Gerasimos Filippatos
- National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - James L Januzzi
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA; Baim Institute for Clinical Research, Boston, Massachusetts, USA
| | | | - Marina Panova-Noeva
- Boehringer Ingelheim Pharma GmbH & Co KG, Ingelheim, Germany; Center for Thrombosis and Haemostasis, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | | | - Juergen Prochaska
- Boehringer Ingelheim International GmbH, Ingelheim, Germany; Preventive Cardiology and Preventive Medicine, Department of Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Stuart J Pocock
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Naveed Sattar
- School of Cardiovascular and Metabolic Health, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Mikhail Sumin
- Boehringer Ingelheim International GmbH, Ingelheim, Germany
| | - Faiez Zannad
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, and INSERM U1116, CHRU, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France
| | - Milton Packer
- Baylor Heart and Vascular Institute, Dallas, Texas, USA; Imperial College, London, United Kingdom.
| |
Collapse
|
3
|
Wang R, Yan J, Zhang H, Zhu X, Xie D, Wang T, Li X. New insights into heavy metal cadmium-induced liver injury: prominent role of programmed cell death mechanisms. Toxicology 2025:154169. [PMID: 40318836 DOI: 10.1016/j.tox.2025.154169] [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: 03/07/2025] [Revised: 04/30/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
The heavy metal cadmium (Cd) is an important environmental factor that induces liver injury and contributes to liver disease. Ongoing research aims to refine our understanding of the pathogenesis of cadmium-induced liver injury and the interactions between the various mechanisms. Oxidative stress, described as a pathophysiological basis of liver injury, is a process in which reactive oxygen species are generated, causing the destruction of hepatocyte structure and cellular dysfunction. Additionally, the activation of oxidative stress downstream signals regulates several forms of cell death, such as apoptosis, necroptosis, autophagy, ferroptosis, and pyroptosis, which significantly contributes to liver damage. Furthermore, the interplay between different types of programmed cell death highlights the complexity of liver injury mechanisms. This review summarizes the role of programmed cell death in Cd-induced liver injury and explores the relationships between different programmed cell death pathways, which is expected to provide new insights into the mechanisms of Cd-induced liver injury.
Collapse
Affiliation(s)
- Ruipeng Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Jun Yan
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China; Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, China; Cancer Prevention and Treatment Center of Lanzhou University School of Medicine, Lanzhou 730000, China; Hepatopancreatobiliary Surgery Institute of Gansu Province, Lanzhou 730000, China
| | - Honglong Zhang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Xingwang Zhu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Danna Xie
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Tingting Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Xun Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China; Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, China; Cancer Prevention and Treatment Center of Lanzhou University School of Medicine, Lanzhou 730000, China; Hepatopancreatobiliary Surgery Institute of Gansu Province, Lanzhou 730000, China; Clinical Research Center for General Surgery of Gansu Province, Lanzhou 730000, China.
| |
Collapse
|
4
|
Anker SD, Friede T, Butler J, Talha KM, Placzek M, Diek M, Nosko A, Stas A, Kluge S, Jarczak D, Deheer G, Rybczynski M, Bayes-Genis A, Edelmann F, Filippatos G, Hasenfuß G, Haverkamp W, Lainscak M, Landmesser U, Macdougall IC, Merkely B, Pieske BM, Pinto FJ, Rassaf T, Volterrani M, von Haehling S, Anker MS, Doehner W, Ince H, Koehler F, Savarese G, Rauch-Kröhnert U, Gori T, Trenkwalder T, Akin I, Paitazoglou C, Kobielusz-Gembala I, Zmuda W, Kuthi L, Frey N, Licka M, Kääb S, Laugwitz KL, Ponikowski P, Karakas M. Ferric carboxymaltose assessment of morbidity and mortality in patients with iron deficiency and chronic heart failure (FAIR-HF2-DZHK05) trial: Baseline characteristics and comparison to other relevant clinical trials. Eur J Heart Fail 2025. [PMID: 40300786 DOI: 10.1002/ejhf.3658] [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: 12/22/2024] [Revised: 03/19/2025] [Accepted: 03/24/2025] [Indexed: 05/01/2025] Open
Abstract
AIMS Prior randomized trials have reported conflicting evidence regarding the efficacy of intravenous (IV) iron in patients with heart failure with reduced ejection fraction (HFrEF) and iron deficiency (ID). METHODS AND RESULTS FAIR-HF2 is a double-blind, randomized, controlled trial evaluating the efficacy of IV ferric carboxymaltose in patients with HFrEF and ID. We report the baseline characteristics of enrolled patients and compare them with other major trials of IV iron in HFrEF (FAIR-HF, CONFIRM-HF, AFFIRM-AHF, IRONMAN, and HEART-FID). A total of 1105 patients were randomized between March 2017 and November 2023. Most patients were men (67%) and median age was 72 (interquartile range [IQR] 63-79) years. More than one-third had a heart failure hospitalization within the preceding 12 months (36%), and 53% were hospitalized at randomization. Common comorbidities included hypertension (79%), coronary artery disease (74%), dyslipidaemia (67%), and diabetes (46%). The median left ventricular ejection fraction was 58% (IQR 42-77) and mean estimated glomerular filtration rate was 58 (IQR 42-77) ml/min/1.73 m2. A total of 1064 (96%) patients were on renin-angiotensin system inhibitors (angiotensin receptor-neprilysin inhibitors [ARNI] 38%), 1016 (92%) on beta-blockers, and 779 (71%) on mineralocorticoid receptor antagonists; and 261 (24%) of patients were on sodium-glucose cotransporter 2 (SGLT2) inhibitors, which is much higher than prior trials. A higher proportion of patients had ischaemic HFrEF (78%) compared to preceding trials. The baseline median haemoglobin (g/dl) was 12.7 (IQR 11.8-13.4), median serum ferritin (μg/dl) was 63 (IQR 36-90), and median transferrin saturation (%) was 16.5 (IQR 11.8-22.9), resembling that of other trials. The mean 6-min walk distance at enrolment was 314 ± 118 m. CONCLUSION The FAIR-HF2 trial represents a contemporary cohort of patients with baseline characteristics mostly similar to prior trial populations. Use of SGLT2 inhibitors and ARNI in FAIR-HF2 was higher than in prior trials. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov ID NCT03036462.
Collapse
Affiliation(s)
- Stefan D Anker
- Deutsches Herzzentrum der Charité, Campus Virchow Klinikum, Berlin, Germany
- Institute of Health Center for Regenerative Therapies (BCRT), German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Charité Universitätsmedizin, Berlin, Germany
| | - Tim Friede
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Gottingen, Germany
| | - Javed Butler
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Baylor Scott and White Research Institute, Dallas, Texas, USA
| | - Khawaja M Talha
- Department of Cardiovascular Disease, Loyola University Medical Center, Maywood, Illinois, USA
| | - Marius Placzek
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| | - Monika Diek
- Deutsches Herzzentrum der Charité, Campus Virchow Klinikum, Berlin, Germany
- Institute of Health Center for Regenerative Therapies (BCRT), German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Charité Universitätsmedizin, Berlin, Germany
| | - Anna Nosko
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Adriane Stas
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Gottingen, Germany
- Department of Medical Informatics, University Medical Center Göttingen, Göttingen, Germany
| | - Stefan Kluge
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dominik Jarczak
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Geraldine Deheer
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Meike Rybczynski
- Department of Cardiology, University Medical Center Hamburg-Eppendorf, University Heart & Vascular Center Hamburg, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Antoni Bayes-Genis
- Heart Institute, Hospital Universitari Germans Trias i Pujol, CIBERCV, Barcelona, Spain
| | - Frank Edelmann
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Virchow Klinikum, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Charité Universitätsmedizin, Berlin, Germany
| | - Gerasimos Filippatos
- Department of Cardiology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Gerd Hasenfuß
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Gottingen, Germany
- Department of Cardiology and Pneumology, University Medical Centre Göttingen, Georg August University of Göttingen, Göttingen, Germany
| | - Wilhelm Haverkamp
- Deutsches Herzzentrum der Charité, Campus Virchow Klinikum, Berlin, Germany
- Institute of Health Center for Regenerative Therapies (BCRT), German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Charité Universitätsmedizin, Berlin, Germany
| | - Mitja Lainscak
- Division of Cardiology, General Hospital Murska Sobota, Murska Sobota, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Ulf Landmesser
- German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Charité Universitätsmedizin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | | | - Bela Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Burkert M Pieske
- Division of Cardiology, Department of Internal Medicine, University Medicine Rostock, Rostock, Germany
| | - Fausto J Pinto
- Centro Academico de Medicina de Lisboa, CCUL@RISE, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Tienush Rassaf
- West German Heart- and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Maurizio Volterrani
- IRCCS San Raffaele Roma, Rome, Italy
- San Raffaele Open University in Rome, Rome, Italy
| | - Stephan von Haehling
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Gottingen, Germany
- Department of Cardiology and Pneumology, University Medical Centre Göttingen, Georg August University of Göttingen, Göttingen, Germany
| | - Markus S Anker
- German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Charité Universitätsmedizin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, Scotland
- Berlin Institute of Health - Center for Regenerative Therapies, and Department of Cardiology (CVK), Deutsches Herzzentrum der Charité and German Centre for Cardiovascular Research Partner Site Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Wolfram Doehner
- Deutsches Herzzentrum der Charité (DHZC), Department of Cardiology, Angiology and Intensive Care Medicine, Campus Charité Mitte, Berlin, Germany
| | - Hüseyin Ince
- Division of Cardiology, Department of Internal Medicine, University Medicine Rostock, Rostock, Germany
| | - Friedrich Koehler
- German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Charité Universitätsmedizin, Berlin, Germany
- Center for Cardiovascular Telemedicine; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Gianluigi Savarese
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany
| | - Ursula Rauch-Kröhnert
- German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Charité Universitätsmedizin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, Scotland
| | - Tommaso Gori
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany
- German Centre for Cardiovascular Research (DZHK), Standort RheinMain, Frankfurt, Germany
| | - Teresa Trenkwalder
- Technical University of Munich, School of Medicine and Health, Department of Cardiovascular Diseases, German Heart Centre Munich, TUM University Hospital, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Ibrahim Akin
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research (DZHK)), Partner Site, Heidelberg/Mannheim, Mannheim, Germany
| | - Christina Paitazoglou
- Department of Cardiology, Angiology and Intensive Care Medicine, University Heart Center Lübeck, Medical Clinic II, University Hospital Schleswig-Holstein, Lübeck, Germany
| | | | - Witold Zmuda
- Oświęcimskie Centrum Badań Klinicznych, Oswiecim, Poland
| | - Luca Kuthi
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumolgy, Clinical Trial Unit, University Hospital Heidelberg, Heidelberg, Germany
| | - Manuela Licka
- Department of Cardiology, Angiology and Pneumolgy, Clinical Trial Unit, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Kääb
- LMU Munich, Department of Medicine I, LMU University Hospital, Munich, Germany
| | - Karl-Ludwig Laugwitz
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Department of Internal Medicine I, Technical University Munich University Hospital, Munich, Germany
| | - Piotr Ponikowski
- Institute of Heart Disease, Medical University and University Hospitals, Wroclaw, Poland
| | - Mahir Karakas
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| |
Collapse
|
5
|
Liu B, Jin Q, Sun YK, Yang ZM, Meng P, Zhang X, Chen Q, Gan P, Zhao T, He JJ, He GP, Xue Q. From bench to bedside: targeting ferroptosis and mitochondrial damage in the treatment of diabetic cardiomyopathy. Front Endocrinol (Lausanne) 2025; 16:1563362. [PMID: 40352456 PMCID: PMC12061709 DOI: 10.3389/fendo.2025.1563362] [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: 01/19/2025] [Accepted: 03/31/2025] [Indexed: 05/14/2025] Open
Abstract
Diabetic cardiomyopathy (DCM) is a common and fatal cardiac complication caused by diabetes, with its pathogenesis involving various forms of cell death and mitochondrial dysfunction, particularly ferroptosis and mitochondrial injury. Recent studies have indicated that ferroptosis and mitochondrial damage play crucial roles in the onset and progression of DCM, though their precise regulatory mechanisms remain unclear. Of particular interest is the interaction between ferroptosis and mitochondrial damage, as well as their synergistic effects, which are not fully understood. This review summarizes the roles of ferroptosis and mitochondrial injury in the progression of DCM and explores the molecular mechanisms involved, with an emphasis on the interplay between these two processes. Additionally, the article offers an overview of targeted drugs shown to be effective in cellular experiments, animal models, and clinical trials, analyzing their mechanisms of action and potential side effects. The goal is to provide insights for future drug development and clinical applications. Moreover, the review explores the challenges and prospects of multi-target combination therapies and personalized medicine interventions in clinical practice to offer strategic guidance for the comprehensive prevention and management of DCM.
Collapse
Affiliation(s)
- Bin Liu
- Department of Cardiology, The Fifth Affiliated Hospital of Kunming Medical University, Gejiu People’s Hospital, Gejiu, Yunnan, China
| | - Qing Jin
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Yi Kang Sun
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Zhi Ming Yang
- Department of Cardiology, The Fifth Affiliated Hospital of Kunming Medical University, Gejiu People’s Hospital, Gejiu, Yunnan, China
| | - Ping Meng
- Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Xi Zhang
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Qiu Chen
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
- Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Pin Gan
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Tao Zhao
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Jia Ji He
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Gui Ping He
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Qiang Xue
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| |
Collapse
|
6
|
Koshino A, Heerspink HJL, Jongs N, Badve SV, Arnott C, Neal B, Jardine M, Mahaffey KW, Pollock C, Perkovic V, Hansen MK, Bakker SJL, Wada T, Neuen BL. Canagliflozin and iron metabolism in the CREDENCE trial. Nephrol Dial Transplant 2025; 40:696-706. [PMID: 39304530 PMCID: PMC11960735 DOI: 10.1093/ndt/gfae198] [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: 05/06/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Studies in patients with heart failure have indicated that sodium-glucose cotransporter 2 (SGLT2) inhibitors increase iron use and enhance erythropoiesis. In this post hoc analysis of the Canagliflozin and Renal Endpoints in Diabetes with Established Nephropathy Clinical Evaluation (CREDENCE) trial, we evaluated the effects of canagliflozin on iron metabolism in patients with chronic kidney disease (CKD) and whether the effects of canagliflozin on hemoglobin and cardiorenal outcomes were modified by iron deficiency. METHODS We measured serum iron, total iron binding capacity (TIBC), transferrin saturation (TSAT) and ferritin at baseline and 12 months. The effects of canagliflozin, relative to placebo, on iron markers were assessed with analysis of covariance. Interactions between baseline iron deficiency, defined as TSAT <20%, and the effects of canagliflozin on hemoglobin and cardiorenal outcomes were evaluated with mixed effect models and Cox regression models, respectively. RESULTS Of 4401 participants randomized in CREDENCE, 2416 (54.9%) had iron markers measured at baseline, of whom 924 (38.2%) were iron deficient. Canagliflozin, compared with placebo, increased TIBC by 2.1% [95% confidence interval (CI) 0.4, 3.8; P = .014] and decreased ferritin by 11.5% (95% CI 7.1, 15.7; P < .001) with no clear effect on serum iron or TSAT. Canagliflozin increased hemoglobin over the trial duration by 7.3 g/L (95% CI 6.2, 8.5; P < .001) and 6.7 g/L (95% CI 5.2, 8.2; P < .001) in patients with and without iron deficiency, respectively (P for interaction = .38). The relative effect of canagliflozin on the primary outcome of doubling of serum creatinine, kidney failure or death due to cardiovascular disease or kidney failure (hazard ratio 0.70, 95% CI 0.56, 0.87) was consistent regardless of iron deficiency (P for interaction = .83), as were effects on other cardiovascular and mortality outcomes (all P for interactions ≥0.10). CONCLUSION Iron deficiency is highly prevalent in patients with type 2 diabetes and CKD. Canagliflozin increased TIBC and decreased ferritin in patients with type 2 diabetes and CKD, suggesting increased iron utilization, and improved hemoglobin levels and clinical outcomes regardless of iron deficiency.
Collapse
Affiliation(s)
- Akihiko Koshino
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Nephrology and Rheumatology, Kanazawa University, Ishikawa, Japan
| | - Hiddo J L Heerspink
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- The George Institute for Global Health, UNSW Sydney, Sydney, Australia
| | - Niels Jongs
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sunil V Badve
- The George Institute for Global Health, UNSW Sydney, Sydney, Australia
- Department of Nephrology, St George Hospital, Sydney, Australia
- Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Clare Arnott
- The George Institute for Global Health, UNSW Sydney, Sydney, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Bruce Neal
- The George Institute for Global Health, UNSW Sydney, Sydney, Australia
- School of Public Health, Imperial College London, UK
| | - Meg Jardine
- The George Institute for Global Health, UNSW Sydney, Sydney, Australia
- NHMRC Clinical Trials Centre University of Sydney NSW, Sydney, Australia
- Concord Repatriation General Hospital, Sydney, Australia
| | - Kenneth W Mahaffey
- Stanford Center for Clinical Research, Stanford University School of Medicine, Stanford, CA, USA
| | - Carol Pollock
- Kolling Institute of Medical Research, Sydney Medical School, University of Sydney, Sydney, Australia
- Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Vlado Perkovic
- The George Institute for Global Health, UNSW Sydney, Sydney, Australia
- Faculty of Medicine, University of New South Wales, Sydney, Australia
| | | | - Stephan J L Bakker
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Takashi Wada
- Department of Nephrology and Rheumatology, Kanazawa University, Ishikawa, Japan
| | - Brendon L Neuen
- The George Institute for Global Health, UNSW Sydney, Sydney, Australia
- Royal North Shore Hospital, St Leonards, New South Wales, Australia
| |
Collapse
|
7
|
Chen D, Shi J, Wu Y, Miao L, Wang Z, Wang Y, Xu S, Lou Y. Dapagliflozin alleviates high-fat-induced obesity cardiomyopathy by inhibiting ferroptosis. ESC Heart Fail 2025; 12:1358-1373. [PMID: 39523867 PMCID: PMC11911578 DOI: 10.1002/ehf2.15150] [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: 11/08/2023] [Revised: 04/10/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
AIM Dapagliflozin (Dapa) is a novel hypoglycaemic agent with multiple cardiovascular protective effects, and it is widely used in treatment of heart failure patients, but whether it can improve obese phenotype of heart failure and its mechanism is still unclear. Ferroptosis is an iron dependent form of cell death and has been proved to be an important role in heart failure. The aim of this study is to determine whether Dapa improves obesity-related heart failure by regulating ferroptosis in high-fat diet rats. METHODS AND RESULTS Male SD rats were fed a high-fat diet for 12 weeks and confirmed of obese heart failure by metabolic parameters and cardiac ultrasound. Being overweight by 20% compared with the normal group, with elevated systolic blood pressure and abnormal levels of insulin and blood lipid (TG and LDL-c), is recognized as obesity. The obese rats with reduced EF, FS, and E/A shown on ultrasound are defined as the obese heart failure (OHF) group. Histological tests confirmed the more pronounced cardiac fibrosis, mitochondrial volume and collagen deposition in OHF group. Dapa treatment effectively reduced body weight, INS, ISI/IRI index, TG and HDL-C levels (P < 0.05). Also, Dapa administration can slightly decrease the SBP and DBP levels; however, there was no statistical difference among those four groups. Furthermore, Dapa treatment can significantly improve high-fat induced systolic and diastolic dysfunction via regulating cardiac histological abnormalities, including less obvious mitochondrial swelling, muscle fibre dissolution and collagen deposition. Additionally, genes from the OHF group were used by GO enrichment analysis, and it shows that ferroptosis metabolic pathway participated in the development of obese phenotype of heart failure. More importantly, Dapa significantly inhibited Fe2+ and MDA levels (P < 0.05), but augmented GSH content (P < 0.05). In addition, the mRNAs and protein expression of some important regulators of ferroptosis, like GPX4, SLC7A11, FTH1 and FPN1, were all decreased after Dapa intervention. CONCLUSION Dapa improved high-fat induced obese cardiac dysfunction via regulating ferroptosis pathway.
Collapse
Affiliation(s)
- Di Chen
- Affiliated Zhongshan Hospital of Dalian UniversityDalianChina
| | - Jiahao Shi
- Zhongshan Hospital of Xiamen University, School of MedicineXiamen UniversityXiamenChina
| | - Yue Wu
- Zhongshan Hospital of Xiamen University, School of MedicineXiamen UniversityXiamenChina
| | - Lizhu Miao
- Yancheng No. 1 People's HospitalYanchengChina
| | - Zilin Wang
- Affiliated Zhongshan Hospital of Dalian UniversityDalianChina
| | - Yixuan Wang
- Affiliated Zhongshan Hospital of Dalian UniversityDalianChina
| | - Siwei Xu
- Central Hospital of Dalian University of Technology (Dalian Municipal Central Hospital)DalianChina
| | - Yu Lou
- The Second Hospital of Dalian Medical UniversityDalianChina
| |
Collapse
|
8
|
Anker SD, Friede T, Butler J, Talha KM, Diek M, Nosko A, Placzek M, Hasenfuß G, Ponikowski P, Karakas M. Rationale and design of the FAIR-HF2-DZHK05 trial: Ferric carboxymaltose assessment of morbidity and mortality in patients with iron deficiency and chronic heart failure. Eur J Heart Fail 2025; 27:681-689. [PMID: 39876699 PMCID: PMC12034437 DOI: 10.1002/ejhf.3574] [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: 10/16/2024] [Revised: 12/05/2024] [Accepted: 12/18/2024] [Indexed: 01/30/2025] Open
Abstract
AIMS While it is widely accepted that intravenous (IV) iron improves functional capacity, symptoms, and cardiovascular outcomes in patients with heart failure (HF) with reduced ejection fraction (HFrEF) diagnosed with iron deficiency (ID), three recently published cardiovascular outcome trials (AFFIRM-AHF, IRONMAN and HEART-FID) of IV iron supplementation in HF failed to demonstrate a significant benefit on their respective primary endpoints. Dosing of IV iron after the initial correction of baseline ID - by design or as a result of trial circumstances - was relatively low (i.e. <500 mg/year). The primary objective of the FAIR-HF2 trial is to evaluate the treatment effect of ferric carboxymaltose (FCM) compared with placebo in ambulatory patients with HFrEF using a higher dose of IV iron during follow-up (i.e. >1000 mg/year). The second objective of the study is to create prospective evidence for patients fulfilling the new definition of ID for patients with HF, i.e. for those with a transferrin saturation <20%. METHODS FAIR-HF2 is an investigator-initiated, multicentre, randomized, double-blind, placebo-controlled trial that has recruited 1105 patients with chronic HF with a left ventricular ejection fraction of ≤45% and concomitant ID, defined as serum ferritin <100 ng/ml or serum ferritin 100-299 ng/ml with a transferrin saturation <20%. Patients were consented and randomized to receive either IV FCM (treatment) or saline (placebo). During an estimated median follow-up of over 2 years, patients underwent a placebo-controlled repletion and maintenance phase, with an initial iron supplementation of up to 2000 mg, followed by 500 mg every 4 months unless stop criteria of haemoglobin >16 mg/dl or serum ferritin >800 ng/ml are met on repeat visits. The trial will evaluate three primary hypotheses: (i) time to first event of cardiovascular death or hospitalization for HF, (ii) the rate of total (first and recurrent) HF hospitalizations (both analysed in the full study population), and (iii) the time to first event of cardiovascular death or hospitalization for HF in patients with a transferrin saturation <20% at baseline. The familywise type I error rate across the three primary endpoint hypotheses will be controlled using the Hochberg procedure (alpha 0.05). CONCLUSION The FAIR-HF2 will evaluate the efficacy of FCM in patients with HFrEF in improving cardiovascular outcomes by utilizing a more aggressive approach towards iron supplementation ensuring prevention of transitional ID after initial repletion targets have been met.
Collapse
Affiliation(s)
- Stefan D. Anker
- Department of Cardiology (CVK) of German Heart Center CharitéInstitute of Health Center for Regenerative Therapies (BCRT)BerlinGermany
- German Centre for Cardiovascular Research (DZHK) partner site BerlinCharité UniversitätsmedizinBerlinGermany
| | - Tim Friede
- Department of Medical StatisticsUniversity Medical Center Göttingen, German Centre for Cardiovascular Research (DZHK), partner site Lower SaxonyGöttingenGermany
| | - Javed Butler
- Department of MedicineUniversity of Mississippi Medical CenterJacksonMSUSA
- Baylor Scott and White Research InstituteDallasTXUSA
| | - Khawaja M. Talha
- Department of Cardiovascular DiseaseLoyola University Medical CenterMaywoodILUSA
| | - Monika Diek
- Department of Cardiology (CVK) of German Heart Center CharitéInstitute of Health Center for Regenerative Therapies (BCRT)BerlinGermany
| | - Anna Nosko
- Department of Intensive Care MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LübeckHamburgGermany
| | - Marius Placzek
- Department of Medical StatisticsUniversity Medical Center Göttingen, German Centre for Cardiovascular Research (DZHK), partner site Lower SaxonyGöttingenGermany
| | - Gerd Hasenfuß
- Clinic for Cardiology and PneumologyUniversity Medical Center Göttingen, German Centre for Cardiovascular Research (DZHK), partner site Lower SaxonyGöttingenGermany
| | - Piotr Ponikowski
- Institute of Heart DiseasesWroclaw Medical UniversityWroclawPoland
| | - Mahir Karakas
- Department of Intensive Care MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LübeckHamburgGermany
| |
Collapse
|
9
|
Marques Vidas M, Portolés J, Cobo M, Gorriz JL, Nuñez J, Cases A. Anemia Management in the Cardiorenal Patient: A Nephrological Perspective. J Am Heart Assoc 2025; 14:e037363. [PMID: 40028884 DOI: 10.1161/jaha.124.037363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Heart failure (HF) and chronic kidney disease (CKD) frequently coexist, sharing significant overlap in prevalence and pathophysiological mechanisms. This coexistence, termed cardiorenal syndrome (CRS), often leads to anemia, which exacerbates both HF and CKD, thereby increasing morbidity and death. Managing anemia in CRS is complex due to conflicting guidelines and the multifactorial nature of the condition. Anemia in CRS is influenced by factors such as inadequate erythropoietin production, iron deficiency, reduced red blood cell life span, and chronic inflammation, which inhibit iron absorption and mobilization. This interplay of mechanisms worsens anemia, further aggravating HF and CKD. Anemia significantly impacts the prognosis of both HF and CKD, and recent trials have shown that hemoglobin increases, particularly with sodium-glucose cotransporter 2 inhibitors, can improve outcomes in patients with HF and CKD. Iron deficiency is also prevalent in both patients with HF and patients with CKD and is associated with poorer exercise capacity and a higher mortality rate. Guidelines for diagnosing and treating iron deficiency differ between HF and CKD. Furthermore, treatment of anemia in CRS is controversial: While sodium-glucose cotransporter 2 inhibitors and intravenous iron has shown consistent benefits in patients with CRS, normalization of hemoglobin with erythropoiesis-stimulating agents improves symptoms and quality of life but have not consistently demonstrated cardiovascular benefits. There are no definitive guidelines for anemia management in CRS. Treatment should address HF, CKD, and anemia concurrently. A proposed algorithm includes correcting iron deficiency, initiating sodium-glucose cotransporter 2 inhibitors, and considering erythropoiesis-stimulating agents if hemoglobin remains <10 g/dL. Further research is needed to optimize anemia management strategies in patients with CRS.
Collapse
Affiliation(s)
- María Marques Vidas
- Nephrology Department Hospital Universitario Puerta de Hierro-Majadahonda, IDIPHISA Madrid Spain
- Medicine Department, Facultad de Medicina Universidad Autónoma de Madrid Madrid Spain
| | - José Portolés
- Nephrology Department Hospital Universitario Puerta de Hierro-Majadahonda, IDIPHISA Madrid Spain
- Medicine Department, Facultad de Medicina Universidad Autónoma de Madrid Madrid Spain
- Anemia Working Group of the Spanish Society of Nephrology Madrid Spain
| | - Marta Cobo
- Cardiology Department Hospital Universitario Puerta de Hierro-Majadahonda Madrid Spain
- Centro de Investigación Biomédica en Red (CIBER Cardiovascular) Madrid Spain
| | - José Luis Gorriz
- Anemia Working Group of the Spanish Society of Nephrology Madrid Spain
- Nephrology Department Hospital Clínico Universitario de Valencia (INCLIVA), Valencia, Universitat de València Valencia Spain
| | - Julio Nuñez
- Cardiology Department, Department of Medicine Hospital Clínico Universitario de Valencia, Universidad de Valencia, CIBER Cardiovascular, INCLIVA Valencia Spain
| | - Aleix Cases
- Anemia Working Group of the Spanish Society of Nephrology Madrid Spain
- Nephrology Unit Hospital Clinic Barcelona Spain
| |
Collapse
|
10
|
Lu H, Claggett BL, Packer M, Pfeffer MA, Lam CSP, Zile MR, Desai AS, Jhund P, Lefkowitz M, McMurray JJV, Solomon SD, Vaduganathan M. Sacubitril/valsartan reduces incident anaemia and iron therapy utilization in heart failure: The PARAGON-HF trial. Eur J Heart Fail 2025; 27:85-95. [PMID: 39217577 DOI: 10.1002/ejhf.3414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/17/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
AIMS Renin-angiotensin system inhibitors (RASi) have been shown to lower haemoglobin levels, potentially related to reductions in erythropoietin levels and haematopoiesis. We examined whether sacubitril/valsartan might attenuate this effect of RASi alone on incident anaemia in patients with heart failure (HF) with mildly reduced or preserved ejection fraction (HFmrEF/HFpEF). METHODS AND RESULTS PARAGON-HF was a global, multicentre randomized clinical trial of sacubitril/valsartan versus the RASi valsartan in patients with HF and left ventricular ejection fraction ≥45%. We evaluated haemoglobin trajectory and risks of incident anaemia and new iron therapy initiation during follow-up. Among 4795 participants, 1111 (23.2%) had anaemia at randomization and 5.6% were treated with iron at baseline. Over a median follow-up of 2.9 years, patients with anaemia were at significantly higher risk for total HF hospitalizations and cardiovascular death, compared with those without anaemia (21.6 vs. 11.5 per 100 patient-years; adjusted rate ratio 1.31; 95% confidence interval [CI] 1.12-1.54; p = 0.001). Sacubitril/valsartan slightly slowed the decline in haemoglobin levels by 0.1 g/dl (95% CI 0.0-0.2 g/dl; p = 0.005). Participants treated with sacubitril/valsartan were at significantly lower risk of developing anaemia (30.3% vs. 37.6%; hazard ratio [HR] 0.76; 95% CI 0.68-0.85; p < 0.001) and starting iron therapy (8.1% vs. 10.0%; HR 0.81; 95% CI 0.67-0.97; p = 0.026). Treatment effects of sacubitril/valsartan versus valsartan on total HF hospitalizations and cardiovascular death were consistent among patients across the haemoglobin spectrum (pinteraction = 0.60). CONCLUSIONS Among patients with HFmrEF/HFpEF, treatment with sacubitril/valsartan resulted in modestly smaller declines in haemoglobin, lower rates of incident anaemia, and fewer new initiations of iron therapy compared with RASi. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov ID NCT01920711.
Collapse
Affiliation(s)
- Henri Lu
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Cardiology, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Brian L Claggett
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Milton Packer
- Baylor University Medical Center, Baylor Heart and Vascular Institute, Dallas, TX, USA
| | - Marc A Pfeffer
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Michael R Zile
- Medical University of South Carolina, Charleston, SC, USA
| | - Akshay S Desai
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pardeep Jhund
- BHF Glasgow Cardiovascular Research Centre, Glasgow, UK
| | | | | | - Scott D Solomon
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Muthiah Vaduganathan
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
11
|
Hopkins S, Baqai F, Gajagowni S, Hickey G. Direct Cardiac Mechanisms of the Sodium Glucose Co-Transporter 2 Inhibitor Class. J Cardiovasc Pharmacol Ther 2025; 30:10742484251323428. [PMID: 40221961 DOI: 10.1177/10742484251323428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
BackgroundSodium-glucose co-transporter 2 (SGLT2) inhibitors have demonstrated significant cardiovascular benefits in clinical trial. While their role in reducing heart failure hospitalizations and cardiovascular mortality is well established, the precise mechanisms underlying their direct cardiac effects remain unclear. This literature review aims to synthesize current knowledge on the molecular and physiological pathways by which SGLT2 inhibitors may exert effects on cardiac tissue, independent of glycemic control.MethodsA comprehensive review of peer-reviewed articles, randomized controlled trials, meta-analyses, and mechanistic studies published in PubMed and related databases was conducted. The search focused on studies examining the direct impact of SGLT2 inhibitors on cardiac function, remodeling, metabolism, and intracellular signaling pathways. Only studies evaluating the cardiac effects separate from their glucose-lowering action were included in the analysis.ResultsThis review identified several key mechanisms by which SGLT2 inhibitors may benefit the heart directly, including reductions in oxidative stress, inflammation, and myocardial fibrosis. Emerging evidence suggests that these drugs modulate key pathways such as sodium-hydrogen exchange (NHE) inhibition, improvement of mitochondrial function, and promotion of ketone body utilization in cardiomyocytes.ConclusionsSGLT2 inhibitors appear to confer direct cardioprotective effects. These include anti-inflammatory, anti-fibrotic, and energy efficiency improvements in the myocardium. The findings highlight new potential therapeutic mechanisms and provide a foundation for further research into the non-diabetic use of SGLT2 inhibitors in heart failure and other cardiac conditions. Understanding these direct effects could lead to optimized treatment strategies for patients with and without diabetes.
Collapse
Affiliation(s)
| | - Faiz Baqai
- Department of Internal Medicine, Baylor College of Medicine, Houston, USA
| | | | - Gavin Hickey
- Heart and Vascular Institute, UPMC, Pittsburgh, USA
| |
Collapse
|
12
|
Mareev VY, Kobalava ZD, Mareev YV, Begrambekova YL, Karapetyan LV, Galochkin SA, Kazakhmedov ER, Lapshin AA, Garganeeva AA, Kuzheleva EA, Efremushkina AA, Kiseleva EV, Barbarash OL, Pecherina TB, Galyavich AS, Galeeva ZM, Baleeva LV, Koziolova NA, Veklich AS, Duplyakov DV, Maksimova MN, Yakushin SS, Smirnova EA, Sedykh EV, Shaposhnik II, Makarova NA, Zemlyanukhina AA, Skibitskiy VV, Fendrikova AV, Skibitskiy AV, Spiropulos NA, Seredenina EM, Orlova YA, Eruslanova KA, Kotovskaya YV, Тkacheva ON, Fedin MA. Relationship of Iron Deficiency, Anemia and Combination of Iron Deficiency With Anemia With Severity of Manifestations of Chronic Heart Failure. Additional Analysis of the Study "Prevalence of Iron Deficiency in Patients With Chronic Heart Failure in the Russian Federation (J-CHF-RF)". KARDIOLOGIIA 2024; 64:62-75. [PMID: 39637392 DOI: 10.18087/cardio.2024.11.n2786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/24/2024] [Indexed: 12/07/2024]
Abstract
Aim To evaluate the role of iron deficiency (ID) identified by various criteria, anemia, and the combination of ID and anemia in determining the severity of the clinical course of chronic heart failure (CHF) in a retrospective analysis of data from 498 patients who participated in the ID-CHF-RF Russian multicenter program.Material and methods ID was diagnosed by the following three criteria established by the European Society of Cardiology (ESC) and the Russian Society of Cardiology (RSC): 1) ferritin concentration <100 μg/l or ferritin concentration 100-299 μg/l in combination with a decreased transferrin saturation (TS) <20%; 2) ID criteria that showed a high sensitivity and specificity when compared with bone marrow morphology (BMM): TS ≤19.8% or serum iron (SI) ≤13 μmol/l; and 3) a composite index including a ferritin concentration <100 μg/l in combination with TS <20% and SI ≤13 μmol/l. The presence of anemia was defined as a hemoglobin concentration of less than 12.0 g/dl in women and less than 13.0 g/dl in men according to the criteria of the World Health Organization.Results Concomitant anemia was detected in 40.3% of patients with CHF; in 85.1% of cases, anemia was combined with the SI concentration below normal. CHF patients with concomitant anemia were significantly older and had low levels of not only red blood cells and hemoglobin but also all parameters of iron metabolism, i.e., SI, ferritin concentration, and TS. The mean deviation of the red blood cell size, that characterizes the degree of anisocytosis, was significantly increased in patients with anemia, especially with a low SI. These patients had a higher CHF functional class, elevated levels of N-terminal fragment of pro-brain natriuretic peptide (NT-proBNP) and walked a shorter distance in the 6-minute walk test, which reflects significantly more severe manifestations of CHF with concomitant anemia, particularly in combination with a low SI. The incidence of ID was 83.1% (including 23.3% in combination with anemia) according to the ESC/RSC criteria; 74.5% (including 43.3% with anemia) according to the BMM criteria; and 51.6% (including 51.7% with anemia) according to the composite index, which seems to be stricter compared to the first two criteria. Regardless of the assessment method (by total weighted average data), in ID combined with anemia, not only the hemoglobin concentration was significantly reduced but all three analyzed parameters of iron metabolism were also significantly reduced (SI 9.0 μmol/l vs. 10.4 μmol/l; ferritin 41 μg/l vs. 59 μg/l; TS 8.5% vs. 12.9%) compared to ID without anemia, respectively. The CHF severity and the NT-proBNP concentration were also maximum for the combination of ID and anemia, in contrast to ID without anemia, regardless of the ID criterion used. A more accurate comparison of the methods for determining ID in CHF in the context of their prognostic value will be obtained by analyzing the data of a two-year follow-up of patients in this study, which will be the subject of the next article.Conclusion This analysis suggests that the presence of concomitant ID without anemia or anemia without ID moderately affects the severity of clinical manifestations of CHF and may be rather markers than factors determining the course of the disease, and in this case, does not require special correction with iron medications. And only ID anemia (a combination of ID with anemia) in patients with CHF can be considered a condition requiring special correction (for example, with intravenous medication) in addition to optimal therapy for CHF. This conclusion does not change depending on the used criteria for ID and requires verification in new RCTs.
Collapse
Affiliation(s)
- V Yu Mareev
- Medical Research and Educational Institute of the Lomonosov Moscow State University, Moscow
| | - Zh D Kobalava
- Moiseev Department of Internal Diseases with a Course of Cardiology and Functional Diagnostics, Medical Institute, Patrice Lumumba Peoples' Friendship University of Russia, Moscow; Vinogradov Municipal Clinical Hospital, Moscow
| | - Yu V Mareev
- Robertson Center for Biostatistics, Glasgow University, Glasgow
| | - Yu L Begrambekova
- Medical Research and Educational Institute of the Lomonosov Moscow State University, Moscow
| | - L V Karapetyan
- Moiseev Department of Internal Diseases with a Course of Cardiology and Functional Diagnostics, Medical Institute, Patrice Lumumba Peoples' Friendship University of Russia, Moscow; Vinogradov Municipal Clinical Hospital, Moscow
| | - S A Galochkin
- Moiseev Department of Internal Diseases with a Course of Cardiology and Functional Diagnostics, Medical Institute, Patrice Lumumba Peoples' Friendship University of Russia, Moscow; Vinogradov Municipal Clinical Hospital, Moscow
| | - E R Kazakhmedov
- Moiseev Department of Internal Diseases with a Course of Cardiology and Functional Diagnostics, Medical Institute, Patrice Lumumba Peoples' Friendship University of Russia, Moscow; Vinogradov Municipal Clinical Hospital, Moscow
| | - A A Lapshin
- Moiseev Department of Internal Diseases with a Course of Cardiology and Functional Diagnostics, Medical Institute, Patrice Lumumba Peoples' Friendship University of Russia, Moscow; Vinogradov Municipal Clinical Hospital, Moscow
| | - A A Garganeeva
- Research Institute of Cardiology, Tomsk Research Medical Center of the Russian Academy of Sciences, Tomsk
| | - E A Kuzheleva
- Research Institute of Cardiology, Tomsk Research Medical Center of the Russian Academy of Sciences, Tomsk
| | - A A Efremushkina
- Altai State Medical University, Barnaul; Altai Territorial Cardiological Dispensary, Barnaul
| | - E V Kiseleva
- Altai Territorial Cardiological Dispensary, Barnaul
| | - O L Barbarash
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo
| | - T B Pecherina
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo
| | | | | | | | | | | | - D V Duplyakov
- Research Institute of Cardiology, Samara State Medical University, Samara; Polyakov Samara Regional Clinical Cardiological Dispensary, Samara
| | - M N Maksimova
- Polyakov Samara Regional Clinical Cardiological Dispensary, Samara
| | - S S Yakushin
- Pavlov Ryazan State Medical University, Ryazan; Regional Clinical Cardiology Dispensary, Ryazan
| | | | - E V Sedykh
- Pavlov Ryazan State Medical University, Ryazan; Regional Clinical Cardiology Dispensary, Ryazan
| | | | - N A Makarova
- South Ural State Medical University, Chelyabinsk
| | | | | | | | | | | | - E M Seredenina
- Medical Research and Educational Institute of the Lomonosov Moscow State University, Moscow
| | - Ya A Orlova
- Medical Research and Educational Institute of the Lomonosov Moscow State University, Moscow
| | - K A Eruslanova
- Russian Gerontological Research and Clinical Center, Pirogov Russian National Research Medical University, Moscow
| | - Yu V Kotovskaya
- Russian Gerontological Research and Clinical Center, Pirogov Russian National Research Medical University, Moscow
| | - O N Тkacheva
- Russian Gerontological Research and Clinical Center, Pirogov Russian National Research Medical University, Moscow
| | - M A Fedin
- Russian Gerontological Research and Clinical Center, Pirogov Russian National Research Medical University, Moscow; Pirogov Russian National Research Medical University, Moscow
| |
Collapse
|
13
|
Aristizábal-Colorado D, Ocampo-Posada M, Rivera-Martínez WA, Corredor-Rengifo D, Rico-Fontalvo J, Gómez-Mesa JE, Duque-Ossman JJ, Abreu-Lomba A. SGLT2 Inhibitors and How They Work Beyond the Glucosuric Effect. State of the Art. Am J Cardiovasc Drugs 2024; 24:707-718. [PMID: 39179723 DOI: 10.1007/s40256-024-00673-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/06/2024] [Indexed: 08/26/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is associated with a heightened risk of cardiovascular and renal complications. While glycemic control remains essential, newer therapeutic options, such as SGLT2 inhibitors, offer additional benefits beyond glucose reduction. This review delves into the mechanisms underlying the cardio-renal protective effects of SGLT2 inhibitors. By inducing relative hypoglycemia, these agents promote ketogenesis, optimize myocardial energy metabolism, and reduce lipotoxicity. Additionally, SGLT2 inhibitors exert renoprotective actions by enhancing renal perfusion, attenuating inflammation, and improving iron metabolism. These pleiotropic effects, including modulation of blood pressure, reduction of uric acid, and improved endothelial function, collectively contribute to the cardiovascular and renal benefits observed with SGLT2 inhibitor therapy. This review will provide clinicians with essential knowledge, understanding, and a clear recollection of this pharmacological group's mechanism of action.
Collapse
Affiliation(s)
- David Aristizábal-Colorado
- Department of Internal Medicine, Universidad Libre, Cali, Colombia
- Internal Medicine Research Group, Universidad Libre, Cali, Colombia
- Interamerican Society of Cardiology (SIAC), Mexico City, Mexico
| | - Martín Ocampo-Posada
- Department of Internal Medicine, Universidad Libre, Cali, Colombia
- Internal Medicine Research Group, Universidad Libre, Cali, Colombia
- Faculty of Health, Pontificia Universidad Javeriana, Cali, Colombia
- Grupo de Investigación en Ciencias Básicas y Clínicas de la Salud, Universidad Javeriana, Cali, Colombia
| | - Wilfredo Antonio Rivera-Martínez
- Internal Medicine Research Group, Universidad Libre, Cali, Colombia
- Department of Endocrinology, Faculty of Medicine, Universidad de Antioquia, Medellin, Colombia
| | - David Corredor-Rengifo
- Department of Internal Medicine, Universidad Libre, Cali, Colombia
- Internal Medicine Research Group, Universidad Libre, Cali, Colombia
| | - Jorge Rico-Fontalvo
- Department of Nephrology. Faculty of Medicine, Universidad Simón Bolívar, Barranquilla, Colombia
- Latin American Society of Nephrology and Arterial Hypertension (SLANH), Panama City, Panamá
| | - Juan Esteban Gómez-Mesa
- Interamerican Society of Cardiology (SIAC), Mexico City, Mexico.
- Cardiology Department, Fundación Valle del Lili, Cali, Colombia.
- Department of Health Sciences, Universidad Icesi, Cali, Colombia.
| | - John Jairo Duque-Ossman
- Universidad Del Quindío, Armenia, Colombia
- Latin American Federation of Endocrinology (FELAEN), Armenia, Colombia
| | - Alin Abreu-Lomba
- Internal Medicine Research Group, Universidad Libre, Cali, Colombia
- Endocrinology Department, Clínica Imbanaco, Cali, Colombia
| |
Collapse
|
14
|
Sanna GD, Milani P, Palladini G. Rethinking the Role of Co-morbidities in Transthyretin Cardiac Amyloidosis: Cinderella or Princess? The Case of Anemia and Iron Deficiency. Am J Cardiol 2024; 229:79-80. [PMID: 39117007 DOI: 10.1016/j.amjcard.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024]
Affiliation(s)
- Giuseppe D Sanna
- Department of Molecular Medicine, University of Pavia, Pavia, Italy; Amyloidosis Research and Treatment Center, Foundation IRCCS Policlinico San Matteo, Pavia, Italy; Clinical and Interventional Cardiology, Sassari University Hospital, Sassari, Italy.
| | - Paolo Milani
- Department of Molecular Medicine, University of Pavia, Pavia, Italy; Amyloidosis Research and Treatment Center, Foundation IRCCS Policlinico San Matteo, Pavia, Italy
| | - Giovanni Palladini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy; Amyloidosis Research and Treatment Center, Foundation IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
15
|
Ru Q, Li Y, Chen L, Wu Y, Min J, Wang F. Iron homeostasis and ferroptosis in human diseases: mechanisms and therapeutic prospects. Signal Transduct Target Ther 2024; 9:271. [PMID: 39396974 PMCID: PMC11486532 DOI: 10.1038/s41392-024-01969-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/08/2024] [Accepted: 09/02/2024] [Indexed: 10/15/2024] Open
Abstract
Iron, an essential mineral in the body, is involved in numerous physiological processes, making the maintenance of iron homeostasis crucial for overall health. Both iron overload and deficiency can cause various disorders and human diseases. Ferroptosis, a form of cell death dependent on iron, is characterized by the extensive peroxidation of lipids. Unlike other kinds of classical unprogrammed cell death, ferroptosis is primarily linked to disruptions in iron metabolism, lipid peroxidation, and antioxidant system imbalance. Ferroptosis is regulated through transcription, translation, and post-translational modifications, which affect cellular sensitivity to ferroptosis. Over the past decade or so, numerous diseases have been linked to ferroptosis as part of their etiology, including cancers, metabolic disorders, autoimmune diseases, central nervous system diseases, cardiovascular diseases, and musculoskeletal diseases. Ferroptosis-related proteins have become attractive targets for many major human diseases that are currently incurable, and some ferroptosis regulators have shown therapeutic effects in clinical trials although further validation of their clinical potential is needed. Therefore, in-depth analysis of ferroptosis and its potential molecular mechanisms in human diseases may offer additional strategies for clinical prevention and treatment. In this review, we discuss the physiological significance of iron homeostasis in the body, the potential contribution of ferroptosis to the etiology and development of human diseases, along with the evidence supporting targeting ferroptosis as a therapeutic approach. Importantly, we evaluate recent potential therapeutic targets and promising interventions, providing guidance for future targeted treatment therapies against human diseases.
Collapse
Affiliation(s)
- Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
16
|
Girardi ACC, Polidoro JZ, Castro PC, Pio-Abreu A, Noronha IL, Drager LF. Mechanisms of heart failure and chronic kidney disease protection by SGLT2 inhibitors in nondiabetic conditions. Am J Physiol Cell Physiol 2024; 327:C525-C544. [PMID: 38881421 DOI: 10.1152/ajpcell.00143.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 06/18/2024]
Abstract
Sodium-glucose cotransporter 2 inhibitors (SGLT2is), initially developed for type 2 diabetes (T2D) treatment, have demonstrated significant cardiovascular and renal benefits in heart failure (HF) and chronic kidney disease (CKD), irrespective of T2D. This review provides an analysis of the multifaceted mechanisms underlying the cardiorenal benefits of SGLT2i in HF and CKD outside of the T2D context. Eight major aspects of the protective effects of SGLT2i beyond glycemic control are explored: 1) the impact on renal hemodynamics and tubuloglomerular feedback; 2) the natriuretic effects via proximal tubule Na+/H+ exchanger NHE3 inhibition; 3) the modulation of neurohumoral pathways with evidence of attenuated sympathetic activity; 4) the impact on erythropoiesis, not only in the context of local hypoxia but also systemic inflammation and iron regulation; 5) the uricosuria and mitigation of the hyperuricemic environment in cardiorenal syndromes; 6) the multiorgan metabolic reprogramming including the potential induction of a fasting-like state, improvement in glucose and insulin tolerance, and stimulation of lipolysis and ketogenesis; 7) the vascular endothelial growth factor A (VEGF-A) upregulation and angiogenesis, and 8) the direct cardiac effects. The intricate interplay between renal, neurohumoral, metabolic, and cardiac effects underscores the complexity of SGLT2i actions and provides valuable insights into their therapeutic implications for HF and CKD. Furthermore, this review sets the stage for future research to evaluate the individual contributions of these mechanisms in diverse clinical settings.
Collapse
Affiliation(s)
- Adriana C C Girardi
- Laboratório de Genética e Cardiologia Molecular, Faculdade de Medicina, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Juliano Z Polidoro
- Laboratório de Genética e Cardiologia Molecular, Faculdade de Medicina, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Paulo C Castro
- Laboratório de Genética e Cardiologia Molecular, Faculdade de Medicina, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Andrea Pio-Abreu
- Disciplina de Nefrologia, Faculdade de Medicina, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Irene L Noronha
- Disciplina de Nefrologia, Faculdade de Medicina, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Luciano F Drager
- Disciplina de Nefrologia, Faculdade de Medicina, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, Brazil
- Unidade de Hipertensão, Faculdade de Medicina, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
17
|
Hiraiwa H, Yura Y, Okumura T, Murohara T. Interplay of the heart, spleen, and bone marrow in heart failure: the role of splenic extramedullary hematopoiesis. Heart Fail Rev 2024; 29:1049-1063. [PMID: 38985383 PMCID: PMC11306273 DOI: 10.1007/s10741-024-10418-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/03/2024] [Indexed: 07/11/2024]
Abstract
Improvements in therapies for heart failure with preserved ejection fraction (HFpEF) are crucial for improving patient outcomes and quality of life. Although HFpEF is the predominant heart failure type among older individuals, its prognosis is often poor owing to the lack of effective therapies. The roles of the spleen and bone marrow are often overlooked in the context of HFpEF. Recent studies suggest that the spleen and bone marrow could play key roles in HFpEF, especially in relation to inflammation and immune responses. The bone marrow can increase production of certain immune cells that can migrate to the heart and contribute to disease. The spleen can contribute to immune responses that either protect or exacerbate heart failure. Extramedullary hematopoiesis in the spleen could play a crucial role in HFpEF. Increased metabolic activity in the spleen, immune cell production and mobilization to the heart, and concomitant cytokine production may occur in heart failure. This leads to systemic chronic inflammation, along with an imbalance of immune cells (macrophages) in the heart, resulting in chronic inflammation and progressive fibrosis, potentially leading to decreased cardiac function. The bone marrow and spleen are involved in altered iron metabolism and anemia, which also contribute to HFpEF. This review presents the concept of an interplay between the heart, spleen, and bone marrow in the setting of HFpEF, with a particular focus on extramedullary hematopoiesis in the spleen. The aim of this review is to discern whether the spleen can serve as a new therapeutic target for HFpEF.
Collapse
Affiliation(s)
- Hiroaki Hiraiwa
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Yoshimitsu Yura
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Takahiro Okumura
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
18
|
Jin EJ, Jo Y, Wei S, Rizzo M, Ryu D, Gariani K. Ferroptosis and iron metabolism in diabetes: Pathogenesis, associated complications, and therapeutic implications. Front Endocrinol (Lausanne) 2024; 15:1447148. [PMID: 39279996 PMCID: PMC11392752 DOI: 10.3389/fendo.2024.1447148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/14/2024] [Indexed: 09/18/2024] Open
Abstract
Diabetes mellitus is a complex chronic disease, considered as one of the most common metabolic disorders worldwide, posing a major threat to global public health. Ferroptosis emerges as a novel mechanism of programmed cell death, distinct from apoptosis, necrosis, and autophagy, driven by iron-dependent lipid peroxidation accumulation and GPx4 downregulation. A mounting body of evidence highlights the interconnection between iron metabolism, ferroptosis, and diabetes pathogenesis, encompassing complications like diabetic nephropathy, cardiomyopathy, and neuropathy. Moreover, ferroptosis inhibitors hold promise as potential pharmacological targets for mitigating diabetes-related complications. A better understanding of the role of ferroptosis in diabetes may lead to an improvement in global diabetes management. In this review, we delve into the intricate relationship between ferroptosis and diabetes development, exploring associated complications and current pharmacological treatments.
Collapse
Affiliation(s)
- Eun-Ju Jin
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Yunju Jo
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Shibo Wei
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Manfredi Rizzo
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Dongryeol Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Karim Gariani
- Service of Endocrinology, Diabetes, Nutrition, and Therapeutic Education, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
- Diabetes Center of the Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland
| |
Collapse
|
19
|
Cirovic A, Cirovic A. Iron Status Screening in Individuals with Heart Failure Before Initiating Sodium-Glucose Cotransporter 2 Inhibitor Therapy: Is It Necessary? Cardiovasc Drugs Ther 2024; 38:859-860. [PMID: 37548810 DOI: 10.1007/s10557-023-07498-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/28/2023] [Indexed: 08/08/2023]
Affiliation(s)
- Ana Cirovic
- Institute of Anatomy, Faculty of Medicine, University of Belgrade, 4/2 Dr Subotica Street, Belgrade, Serbia
| | - Aleksandar Cirovic
- Institute of Anatomy, Faculty of Medicine, University of Belgrade, 4/2 Dr Subotica Street, Belgrade, Serbia.
| |
Collapse
|
20
|
Docherty KF, McMurray JJ, Kalra PR, Cleland JG, Lang NN, Petrie MC, Robertson M, Ford I. Intravenous iron and SGLT2 inhibitors in iron-deficient patients with heart failure and reduced ejection fraction. ESC Heart Fail 2024; 11:1875-1879. [PMID: 38549192 PMCID: PMC11287326 DOI: 10.1002/ehf2.14742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/11/2024] [Indexed: 07/31/2024] Open
Abstract
AIMS To explore the potential interaction between use of SGLT2 inhibitors and the increase in haemoglobin in patients randomized to intravenous iron or the control group in the IRONMAN (Effectiveness of Intravenous Iron Treatment versus Standard Care in Patients with Heart Failure and Iron Deficiency) trial. METHODS AND RESULTS This was a post hoc exploratory analysis of the IRONMAN trial which randomized patients with heart failure, a left ventricular ejection fraction (LVEF) ≤ 45% and iron deficiency (transferrin saturation <20% or ferritin <100 μg/L) to open label intravenous ferric derisomaltose or usual care. Of the 1137 randomized patients, 29 (2.6%) were taking an SGLT2 inhibitor at baseline. The mean (SD) change in haemoglobin from baseline at 4 weeks in those taking an SGLT2 inhibitor at baseline was 1.3 (1.2) g/dL in patients randomized to ferric derisomaltose and 0.1 (0.7) g/dL in the usual care group; between-group difference = 1.0 g/dL (95% CI 0.1, 1.8). The equivalent numbers in the no SGLT2 inhibitor group were 0.6 (0.9) g/dL in those randomized to ferric derisomaltose and 0.1 (0.8) g/dL in the usual care group; between-group difference = 0.4 g/dL (95% CI 0.3, 1.6); interaction P value = 0.10. No patient receiving an SGLT2 inhibitor at baseline developed polycythaemia during follow-up (defined as haemoglobin >16.5 g/dL [men] or >16 g/dL [women]). CONCLUSIONS In the IRONMAN trial, there was a trend to a greater increase in haemoglobin with ferric derisomaltose in iron-deficient patients taking an SGLT2 inhibitor at baseline, as compared with those not taking one.
Collapse
Affiliation(s)
- Kieran F. Docherty
- BHF Cardiovascular Research CentreSchool of Cardiovascular and Metabolic Health University of GlasgowGlasgowUK
| | - John J.V. McMurray
- BHF Cardiovascular Research CentreSchool of Cardiovascular and Metabolic Health University of GlasgowGlasgowUK
| | - Paul R. Kalra
- Department of CardiologyPortsmouth Hospitals University NHS TrustPortsmouthUK
- Faculty of Science and HealthUniversity of PortsmouthPortsmouthUK
| | - John G.F. Cleland
- BHF Cardiovascular Research CentreSchool of Cardiovascular and Metabolic Health University of GlasgowGlasgowUK
| | - Ninian N. Lang
- BHF Cardiovascular Research CentreSchool of Cardiovascular and Metabolic Health University of GlasgowGlasgowUK
| | - Mark C. Petrie
- BHF Cardiovascular Research CentreSchool of Cardiovascular and Metabolic Health University of GlasgowGlasgowUK
| | | | - Ian Ford
- Robertson Centre for BiostatisticsUniversity of GlasgowGlasgowUK
| |
Collapse
|
21
|
Lakhal-Littleton S, Cleland JGF. Iron deficiency and supplementation in heart failure. Nat Rev Cardiol 2024; 21:463-486. [PMID: 38326440 DOI: 10.1038/s41569-024-00988-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/08/2024] [Indexed: 02/09/2024]
Abstract
Non-anaemic iron deficiency (NAID) is a strategic target in cardiovascular medicine because of its association with a range of adverse effects in various conditions. Endeavours to tackle NAID in heart failure have yielded mixed results, exposing knowledge gaps in how best to define 'iron deficiency' and the handling of iron therapies by the body. To address these gaps, we harness the latest understanding of the mechanisms of iron homeostasis outside the erythron and integrate clinical and preclinical lines of evidence. The emerging picture is that current definitions of iron deficiency do not assimilate the multiple influences at play in patients with heart failure and, consequently, fail to identify those with a truly unmet need for iron. Additionally, current iron supplementation therapies benefit only certain patients with heart failure, reflecting differences in the nature of the unmet need for iron and the modifying effects of anaemia and inflammation on the handling of iron therapies by the body. Building on these insights, we identify untapped opportunities in the management of NAID, including the refinement of current approaches and the development of novel strategies. Lessons learned from NAID in cardiovascular disease could ultimately translate into benefits for patients with other chronic conditions such as chronic kidney disease, chronic obstructive pulmonary disease and cancer.
Collapse
Affiliation(s)
| | - John G F Cleland
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| |
Collapse
|
22
|
Gale SE, Willeford A, Sandquist K, Watson K. Intravenous iron in patients with iron deficiency and heart failure: a review of modern evidence. Curr Opin Cardiol 2024; 39:178-187. [PMID: 38353280 DOI: 10.1097/hco.0000000000001121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
PURPOSE OF REVIEW Iron deficiency is common in patients with heart failure, affecting up to half of ambulatory patients and an even greater percentage of patients admitted for acute decompensation. Iron deficiency in this population is also associated with poor outcomes, including worse quality of life in addition to increased hospitalizations for heart failure and mortality. Evidence suggests that patients with iron deficiency in heart failure may benefit from repletion with IV iron. RECENT FINDINGS In this review, we outline the etiology and pathophysiology of iron deficiency in heart failure as well as various iron formulations available. We discuss evidence for intravenous iron repletion with a particular focus on recent studies that have evaluated its effects on hospitalizations and mortality. Finally, we discuss areas of uncertainty and future study and provide practical guidance for iron repletion. SUMMARY In summary, there is overwhelming evidence that intravenous iron repletion in patients with iron deficiency in heart failure is both beneficial and safe. However, further evidence is needed to better identify which patients would most benefit from iron repletion as well as the ideal repletion strategy.
Collapse
Affiliation(s)
- Stormi E Gale
- Novant Health Heart and Vascular Institute, Huntersville, North Carolina
| | - Andrew Willeford
- University of California San Diego Skaggs School of Pharmacy and Pharmaceutical Sciences, San Diego, California
| | | | - Kristin Watson
- University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| |
Collapse
|
23
|
Bhatia K, Sabharwal B, Gupta K, Lopez PD, Kaur A, Bhatia HK, Gandhi KD, Niroula S, Correa A, Birati EY, Argulian E, Fox A, Mahmood K. Clinical outcomes of intravenous iron therapy in patients with heart failure and iron deficiency: Meta-analysis and trial sequential analysis of randomized clinical trials. J Cardiol 2024; 83:105-112. [PMID: 37380069 DOI: 10.1016/j.jjcc.2023.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/13/2023] [Accepted: 06/22/2023] [Indexed: 06/30/2023]
Abstract
BACKGROUND Iron deficiency in patients with heart failure (HF) is underdiagnosed and undertreated. The role of intravenous (IV) iron is well-established to improve quality of life measures. Emerging evidence also supports its role in preventing cardiovascular events in patients with HF. METHODOLOGY We conducted a literature search of multiple electronic databases. Randomized controlled trials that compared IV iron to usual care among patients with HF and reported cardiovascular (CV) outcomes were included. Primary outcome was the composite of first heart failure hospitalization (HFH) or CV death. Secondary outcomes included HFH (first or recurrent), CV death, all-cause mortality, hospitalization for any cause, gastrointestinal (GI) side effects, or any infection. We performed trial sequential and cumulative meta-analyses to evaluate the effect of IV iron on the primary endpoint, and on HFH. RESULTS Nine trials enrolling 3337 patients were included. Adding IV iron to usual care significantly reduced the risk of first HFH or CV death [risk ratio (RR) 0.84; 95 % confidence interval (CI) 0.75-0.93; I2 = 0 %; number needed to treat (NNT) 18], which was primarily driven by a reduction in the risk of HFH of 25 %. IV iron also reduced the risk of the composite of hospitalization for any cause or death (RR 0.92; 95 % CI 0.85-0.99; I2 = 0 %; NNT 19). There was no significant difference in the risk of CV death, all-cause mortality, adverse GI events, or any infection among patients receiving IV iron compared to usual care. The observed benefits of IV iron were directionally consistent across trials and crossed both the statistical and trial sequential boundaries of benefit. CONCLUSION In patients with HF and iron deficiency, the addition of IV iron to usual care reduces the risk of HFH without affecting the risk of CV or all-cause mortality.
Collapse
Affiliation(s)
- Kirtipal Bhatia
- Mount Sinai Heart, Mount Sinai Morningside Hospital, New York, NY, USA
| | - Basera Sabharwal
- Mount Sinai Heart, Mount Sinai Morningside Hospital, New York, NY, USA
| | - Kartik Gupta
- Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
| | - Persio D Lopez
- Mount Sinai Heart, Mount Sinai Morningside Hospital, New York, NY, USA
| | - Arpanjeet Kaur
- Department of Internal Medicine, Mount Sinai Morningside/West, New York, NY, USA
| | - Harsimran K Bhatia
- Department of Medicine, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | | | - Shailesh Niroula
- Department of Internal Medicine, Beaumont Hospital, Royal Oak, MI, USA
| | - Ashish Correa
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Edo Y Birati
- The Lydia and Carol Kittner, Lea and Benjamin Davidai Division of Cardiovascular Medicine, Poriya Medical Center, Azrieli Faculty of Medicine, Bar-Ilan University, Tel Aviv, Israel
| | - Edgar Argulian
- Mount Sinai Heart, Mount Sinai Morningside Hospital, New York, NY, USA
| | - Arieh Fox
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kiran Mahmood
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
24
|
Luo Y, Bai R, Zhang W, Qin G. Selective sodium-glucose cotransporter-2 inhibitors in the improvement of hemoglobin and hematocrit in patients with type 2 diabetes mellitus: a network meta-analysis. Front Endocrinol (Lausanne) 2024; 15:1333624. [PMID: 38362282 PMCID: PMC10867125 DOI: 10.3389/fendo.2024.1333624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 01/16/2024] [Indexed: 02/17/2024] Open
Abstract
Objective To compare the effects of different selective sodium-glucose cotransporter-2 inhibitors (SGLT2i) on hemoglobin and hematocrit in patients with type 2 diabetes mellitus (T2DM) with a network meta-analysis (NMA). Methods Randomized controlled trials (RCTs) on SGLT2i for patients with T2DM were searched in PubMed, Embase, Cochrane Library, and Web of Science from inception of these databases to July 1, 2023. The risk of bias (RoB) tool was used to evaluate the quality of the included studies, and R software was adopted for data analysis. Results Twenty-two articles were included, involving a total of 14,001 T2DM patients. SGLT2i included empagliflozin, dapagliflozin, and canagliflozin. The NMA results showed that compared with placebo, canagliflozin 100mg, canagliflozin 300mg, dapagliflozin 10mg, dapagliflozin 2mg, dapagliflozin 50mg, dapagliflozin 5mg, empagliflozin 25mg, and dapagliflozin 20mg increased hematocrit in patients with T2DM, while canagliflozin 100mg, canagliflozin 200mg, canagliflozin 300mg increased hemoglobin in patients with T2DM. In addition, the NMA results indicated that canagliflozin 100mg had the best effect on the improvement of hematocrit, and canagliflozin 200mg had the best effect on the improvement of hemoglobin. Conclusion Based on the existing studies, we concluded that SGLT2i could increase hematocrit and hemoglobin levels in patients with T2DM, and canagliflozin 100mg had the best effect on the improvement of hematocrit, while canagliflozin 200mg had the best effect on the improvement of hemoglobin. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/#loginpage, identifier PROSPERO (CRD42023477103).
Collapse
Affiliation(s)
- Yuanyuan Luo
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Ruojing Bai
- Department of Geriatric Medicine, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Wei Zhang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Guijun Qin
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
25
|
Yu H, Gan D, Luo Z, Yang Q, An D, Zhang H, Hu Y, Ma Z, Zeng Q, Xu D, Ren H. α-Ketoglutarate improves cardiac insufficiency through NAD +-SIRT1 signaling-mediated mitophagy and ferroptosis in pressure overload-induced mice. Mol Med 2024; 30:15. [PMID: 38254035 PMCID: PMC10804789 DOI: 10.1186/s10020-024-00783-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND In heart failure (HF), mitochondrial dysfunction and metabolic remodeling lead to a reduction in energy productivity and aggravate cardiomyocyte injury. Supplementation with α-ketoglutarate (AKG) alleviated myocardial hypertrophy and fibrosis in mice with HF and improved cardiac insufficiency. However, the myocardial protective mechanism of AKG remains unclear. We verified the hypothesis that AKG improves mitochondrial function by upregulating NAD+ levels and activating silent information regulator 2 homolog 1 (SIRT1) in cardiomyocytes. METHODS In vivo, 2% AKG was added to the drinking water of mice undergoing transverse aortic constriction (TAC) surgery. Echocardiography and biopsy were performed to evaluate cardiac function and pathological changes. Myocardial metabolomics was analyzed by liquid chromatography‒mass spectrometry (LC‒MS/MS) at 8 weeks after surgery. In vitro, the expression of SIRT1 or PINK1 proteins was inhibited by selective inhibitors and siRNA in cardiomyocytes stimulated with angiotensin II (AngII) and AKG. NAD+ levels were detected using an NAD test kit. Mitophagy and ferroptosis levels were evaluated by Western blotting, qPCR, JC-1 staining and lipid peroxidation analysis. RESULTS AKG supplementation after TAC surgery could alleviate myocardial hypertrophy and fibrosis and improve cardiac function in mice. Metabolites of the malate-aspartate shuttle (MAS) were increased, but the TCA cycle and fatty acid metabolism pathway could be inhibited in the myocardium of TAC mice after AKG supplementation. Decreased NAD+ levels and SIRT1 protein expression were observed in heart of mice and AngII-treated cardiomyocytes. After AKG treatment, these changes were reversed, and increased mitophagy, inhibited ferroptosis, and alleviated damage in cardiomyocytes were observed. When the expression of SIRT1 was inhibited by a selective inhibitor and siRNA, the protective effect of AKG was suppressed. CONCLUSION Supplementation with AKG can improve myocardial hypertrophy, fibrosis and chronic cardiac insufficiency caused by pressure overload. By increasing the level of NAD+, the SIRT-PINK1 and SIRT1-GPX4 signaling pathways are activated to promote mitophagy and inhibit ferroptosis in cardiomyocytes, which ultimately alleviates cardiomyocyte damage.
Collapse
Affiliation(s)
- Hao Yu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Daojing Gan
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Zhen Luo
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Qilin Yang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Dongqi An
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Hao Zhang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Yingchun Hu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Zhuang Ma
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Qingchun Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Dingli Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China.
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China.
| | - Hao Ren
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China.
- Department of Rheumatology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
26
|
Packer M. Mechanisms of enhanced renal and hepatic erythropoietin synthesis by sodium-glucose cotransporter 2 inhibitors. Eur Heart J 2023; 44:5027-5035. [PMID: 37086098 PMCID: PMC10733737 DOI: 10.1093/eurheartj/ehad235] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 03/06/2023] [Accepted: 04/03/2023] [Indexed: 04/23/2023] Open
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors reduce the risk of major heart failure events, an action that is statistically linked to enhanced erythropoiesis, suggesting that stimulation of erythropoietin and cardioprotection are related to a shared mechanism. Four hypotheses have been proposed to explain how these drugs increase erythropoietin production: (i) renal cortical reoxygenation with rejuvenation of erythropoietin-producing cells; (ii) counterregulatory distal sodium reabsorption leading to increased tubular workload and oxygen consumption, and thus, to localized hypoxia; (iii) increased iron mobilization as a stimulus of hypoxia-inducible factor-2α (HIF-2α)-mediated erythropoietin synthesis; and (iv) direct HIF-2α activation and enhanced erythropoietin gene transcription due to increased sirtuin-1 (SIRT1) signaling. The first two hypotheses assume that the source of increased erythropoietin is the interstitial fibroblast-like cells in the deep renal cortex. However, SGLT2 inhibitors do not alter regional tissue oxygen tension in the non-diabetic kidney, and renal erythropoietin synthesis is markedly impaired in patients with anemia due to chronic kidney disease, and yet, SGLT2 inhibitors produce an unattenuated erythrocytic response in these patients. This observation raises the possibility that the liver contributes to the production of erythropoietin during SGLT2 inhibition. Hypoxia-inducible factor-2α and erythropoietin are coexpressed not only in the kidney but also in hepatocytes; the liver is a major site of production when erythropoietin stimulation is maintained for prolonged periods. The ability of SGLT2 inhibitors to improve iron mobilization by derepressing hepcidin and ferritin would be expected to increase cytosolic ferrous iron, which might stimulate HIF-2α expression in both the kidney and liver through the action of iron regulatory protein 1. Alternatively, the established ability of SGLT2 inhibitors to enhance SIRT1 might be the mechanism of enhanced erythropoietin production with these drugs. In hepatic cell lines, SIRT1 can directly activate HIF-2α by deacetylation, and additionally, through an effect of SIRT in the liver, peroxisome proliferator-activated receptor-γ coactivator-1α binds to hepatic nuclear factor 4 to promote transcription of the erythropoietin gene and synthesis of erythropoietin. Since SIRT1 up-regulation exerts direct cytoprotective effects on the heart and stimulates erythropoietin, it is well-positioned to represent the shared mechanism that links erythropoiesis to cardioprotection during SGLT2 inhibition.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, 621 North Hall Street, Dallas, TX 75226, USA
- Imperial College, London, UK
| |
Collapse
|
27
|
Tziastoudi M, Pissas G, Golfinopoulos S, Filippidis G, Dousdampanis P, Eleftheriadis T, Stefanidis I. Sodium-Glucose Transporter 2 (SGLT2) Inhibitors and Iron Deficiency in Heart Failure and Chronic Kidney Disease: A Literature Review. Life (Basel) 2023; 13:2338. [PMID: 38137939 PMCID: PMC10744560 DOI: 10.3390/life13122338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Heart failure (HF) and chronic kidney disease (CKD) are associated with high mortality. In both disorders, impaired iron homeostasis, mostly in the form of a functional iron deficiency, is a frequent co-morbidity. In HF, functional iron deficiency and management by i.v. iron supplementation have been proven to affect both prognosis and functional capacity. In the same context, iron supplementation is routine for the adequate management of renal anemia in CKD. In numerous recent studies in HF and in CKD, sodium-glucose transporter 2 (SGLT2) inhibitor treatment has been proven to significantly reduce mortality. Furthermore, the same trials showed that these drugs alleviate iron deficiency and anemia. These effects of SGLT2 inhibitors may be due to an amelioration of inflammation with reduced interleukin-6 (IL-6) and to an enhancement of autophagy with increased sirtuin 1 (SIRT1), both associated with modified production of hepcidin and enhanced ferritinophagy. However, the exact pathogenic basis of the beneficial SGLT2 inhibitor action is not fully elucidated. Nevertheless, effects on iron homeostasis might be a potential explanatory mechanism for the powerful SGLT2 inhibitors' cardiovascular and renal outcome benefits. In addition, the interaction between iron supplementation and SGLT2 inhibitors and its potential impact on prognosis remains to be clarified by future studies. This review represents a significant effort to explore the complex relationships involved, seeking to elucidate the intricate mechanisms by which SGLT2 inhibitors influence iron homeostasis.
Collapse
Affiliation(s)
- Maria Tziastoudi
- Clinic of Nephrology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41334 Larisa, Greece; (G.P.); (S.G.); (G.F.); (P.D.); (T.E.)
| | | | | | | | | | | | - Ioannis Stefanidis
- Clinic of Nephrology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41334 Larisa, Greece; (G.P.); (S.G.); (G.F.); (P.D.); (T.E.)
| |
Collapse
|
28
|
Marques P, Matias P, Packer M, Vieira JT, Vasques-Nóvoa F, Sharma A, Mavrakanas TA, Friões F, Ferreira JP. Erythropoietic response after intravenous iron in patients with heart failure and reduced ejection fraction with and without background treatment with sodium-glucose cotransporter 2 inhibitors. Eur J Heart Fail 2023; 25:2191-2198. [PMID: 37559543 DOI: 10.1002/ejhf.2992] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/11/2023] Open
Abstract
AIMS Intravenous (IV) iron increases haemoglobin/haematocrit and improves outcomes in patients with heart failure with reduced ejection fraction (HFrEF) and iron deficiency. Sodium-glucose cotransporter 2 inhibitors (SGLT2i) also increase haemoglobin/haematocrit and improve outcomes in heart failure by mechanisms linked to nutrient deprivation signalling and reduction of inflammation and oxidative stress. The effect of IV iron among patients using SGLT2i has not yet been studied. The aim of this study was to evaluate the changes in haemoglobin, haematocrit, and iron biomarkers in HFrEF patients treated with IV iron with and without background SGLT2i treatment. Secondary outcomes included changes in natriuretic peptides, kidney function and heart failure-associated outcomes. METHODS AND RESULTS Retrospective, single-centre analysis of HFrEF patients with iron deficiency treated with IV iron using (n = 60) and not using (n = 60) SGLT2i, matched for age and sex. Mean age was 73 ± 12 years, 48% were men, with more than 65% of patients having chronic kidney disease and anaemia. After adjustment for all baseline differences, SGLT2i users experienced a greater increase in haemoglobin and haematocrit compared to SGLT2i non-users: haemoglobin +0.57 g/dl (95% confidence interval [CI] 0.04-1.10, p = 0.036) and haematocrit +1.64% (95% CI 0.18-3.11, p = 0.029). No significant differences were noted for iron biomarkers or any of the secondary outcomes. CONCLUSION Combined treatment with IV iron and background SGLT2i was associated with a greater increase in haemoglobin and haematocrit than IV iron without background SGLT2i. These results suggest that in HFrEF patients treated with IV iron, SGLT2i may increase the erythropoietic response. Further studies are needed to ascertain the potential benefit or harm of combining these two treatments in heart failure patients.
Collapse
Affiliation(s)
- Pedro Marques
- Department of Internal Medicine, Centro Hospitalar Universitário de São João, Porto, Portugal
- Department of Surgery and Physiology, Cardiovascular Research and Development Center (UnIC@RISE), Faculty of Medicine of the University of Porto, Porto, Portugal
- Division of Nephrology, McGill University Health Centre, Montreal, QC, Canada
| | - Paula Matias
- Department of Internal Medicine, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Milton Packer
- Baylor Heart and Vascular Institute, Dallas, TX, USA
- Imperial College, London, UK
| | - Joana T Vieira
- Department of Internal Medicine, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Francisco Vasques-Nóvoa
- Department of Internal Medicine, Centro Hospitalar Universitário de São João, Porto, Portugal
- Department of Surgery and Physiology, Cardiovascular Research and Development Center (UnIC@RISE), Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Abhinav Sharma
- Division of Cardiology, DREAM-CV Lab, McGill University Health Centre, Montreal, QC, Canada
| | - Thomas A Mavrakanas
- Division of Nephrology, McGill University Health Centre, Montreal, QC, Canada
| | - Fernando Friões
- Department of Internal Medicine, Centro Hospitalar Universitário de São João, Porto, Portugal
- Department of Surgery and Physiology, Cardiovascular Research and Development Center (UnIC@RISE), Faculty of Medicine of the University of Porto, Porto, Portugal
- Department of Medicine, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - João Pedro Ferreira
- Department of Surgery and Physiology, Cardiovascular Research and Development Center (UnIC@RISE), Faculty of Medicine of the University of Porto, Porto, Portugal
- Inserm, Centre d'Investigations Cliniques, CHRU Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Université de Lorraine, Nancy, France
| |
Collapse
|
29
|
Koshino A, Neuen BL, Jongs N, Pollock C, Greasley PJ, Andersson EM, Hammarstedt A, Karlsson C, Langkilde AM, Wada T, Heerspink HJL. Effects of dapagliflozin and dapagliflozin-saxagliptin on erythropoiesis, iron and inflammation markers in patients with type 2 diabetes and chronic kidney disease: data from the DELIGHT trial. Cardiovasc Diabetol 2023; 22:330. [PMID: 38017482 PMCID: PMC10685512 DOI: 10.1186/s12933-023-02027-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 10/12/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND This post-hoc analysis of the DELIGHT trial assessed effects of the SGLT2 inhibitor dapagliflozin on iron metabolism and markers of inflammation. METHODS Patients with type 2 diabetes and albuminuria were randomized to dapagliflozin, dapagliflozin and saxagliptin, or placebo. We measured hemoglobin, iron markers (serum iron, transferrin saturation, and ferritin), plasma erythropoietin, and inflammatory markers (urinary MCP-1 and urinary/serum IL-6) at baseline and week 24. RESULTS 360/461 (78.1%) participants had available biosamples. Dapagliflozin and dapagliflozin-saxagliptin, compared to placebo, increased hemoglobin by 5.7 g/L (95%CI 4.0, 7.3; p < 0.001) and 4.4 g/L (2.7, 6.0; p < 0.001) and reduced ferritin by 18.6% (8.7, 27.5; p < 0.001) and 18.4% (8.7, 27.1; p < 0.001), respectively. Dapagliflozin reduced urinary MCP-1/Cr by 29.0% (14.6, 41.0; p < 0.001) and urinary IL-6/Cr by 26.6% (9.1, 40.7; p = 0.005) with no changes in other markers. CONCLUSIONS Dapagliflozin increased hemoglobin and reduced ferritin and urinary markers of inflammation, suggesting potentially important effects on iron metabolism and inflammation. TRIAL REGISTRATION ClinicalTrials.gov NCT02547935.
Collapse
Affiliation(s)
- Akihiko Koshino
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, Groningen, the Netherlands
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Ishikawa, Japan
| | - Brendon L Neuen
- The George Institute for Global Health, UNSW Sydney, Sydney, Australia
| | - Niels Jongs
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, Groningen, the Netherlands
| | - Carol Pollock
- Kolling Institute of Medical Research, Sydney Medical School, University of Sydney, Sydney, Australia
- Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Peter J Greasley
- BioPharmaceuticals Research and Development, AstraZeneca, Gothenburg, Sweden
| | - Eva-Marie Andersson
- BioPharmaceuticals Research and Development, AstraZeneca, Gothenburg, Sweden
| | - Ann Hammarstedt
- BioPharmaceuticals Research and Development, AstraZeneca, Gothenburg, Sweden
| | - Cecilia Karlsson
- BioPharmaceuticals Research and Development, AstraZeneca, Gothenburg, Sweden
| | | | - Takashi Wada
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Ishikawa, Japan
| | - Hiddo J L Heerspink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, Groningen, the Netherlands.
- The George Institute for Global Health, UNSW Sydney, Sydney, Australia.
| |
Collapse
|
30
|
Wang Y, Wu J. Ferroptosis: a new strategy for cardiovascular disease. Front Cardiovasc Med 2023; 10:1241282. [PMID: 37731525 PMCID: PMC10507265 DOI: 10.3389/fcvm.2023.1241282] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/21/2023] [Indexed: 09/22/2023] Open
Abstract
Cardiovascular disease (CVD) is currently one of the prevalent causes of human death. Iron is one of the essential trace elements in the human body and a vital component of living tissues. All organ systems require iron for various metabolic processes, including myocardial and skeletal muscle metabolism, erythropoiesis, mitochondrial function, and oxygen transport. Its deficiency or excess in the human body remains one of the nutritional problems worldwide. The total amount of iron in a normal human body is about 3-5 g. Iron deficiency may cause symptoms such as general fatigue, pica, and nerve deafness, while excessive iron plays a crucial role in the pathophysiological processes of the heart through ferroptosis triggered by the Fenton reaction. It differs from other cell death modes based on its dependence on the accumulation of lipid peroxides and REDOX imbalance, opening a new pathway underlying the pathogenesis and mechanism of CVDs. In this review, we describe the latest research progress on the mechanism of ferroptosis and report its crucial role and association with miRNA in various CVDs. Finally, we summarise the potential therapeutic value of ferroptosis-related drugs or ferroptosis inhibitors in CVDs.
Collapse
Affiliation(s)
| | - Junduo Wu
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
31
|
Tkaczyszyn M, Fudim M, Ponikowski P, Biegus J. Pathophysiology and Treatment Opportunities of Iron Deficiency in Heart Failure: Is There a Need for Further Trials? Curr Heart Fail Rep 2023; 20:300-307. [PMID: 37428429 PMCID: PMC10421819 DOI: 10.1007/s11897-023-00611-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/07/2023] [Indexed: 07/11/2023]
Abstract
PURPOSE OF REVIEW Iron deficiency (ID) complicates heart failure (HF) at different stages of the natural history of the disease; however, this frequent comorbidity is still not comprehensively understood and investigated in terms of pathophysiology. Intravenous iron therapy with ferric carboxymaltose (FCM) should be considered to improve the quality of life, exercise capacity, and symptoms in stable HF with ID, as well as to reduce HF hospitalizations in iron-deficient patients stabilized after an episode of acute HF. The therapy with intravenous iron, however, continues to generate important clinical questions for cardiologists. RECENT FINDINGS In the current paper, we discuss the class effect concept for intravenous iron formulations beyond FCM, based on the experiences of nephrologists who administer different intravenous iron formulations in advanced chronic kidney disease complicated with ID and anemia. Furthermore, we discuss the neutral effects of oral iron therapy in patients with HF, because there are still some reasons to further explore this route of supplementation. The different definitions of ID applied in HF studies and new doubts regarding possible interactions of intravenous iron with sodium-glucose co-transporter type 2 inhibitors are also emphasized. The experiences of other medical specializations may provide new information on how to optimally replenish iron in patients with HF and ID.
Collapse
Affiliation(s)
- Michał Tkaczyszyn
- Institute of Heart Diseases, Wroclaw Medical University, Borowska 213, 50-556, Wroclaw, Poland.
- Institute of Heart Diseases, University Hospital, Wroclaw, Poland.
| | - Marat Fudim
- Division of Cardiology, Duke University Medical Center, Durham, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
| | - Piotr Ponikowski
- Institute of Heart Diseases, Wroclaw Medical University, Borowska 213, 50-556, Wroclaw, Poland
- Institute of Heart Diseases, University Hospital, Wroclaw, Poland
| | - Jan Biegus
- Institute of Heart Diseases, Wroclaw Medical University, Borowska 213, 50-556, Wroclaw, Poland
- Institute of Heart Diseases, University Hospital, Wroclaw, Poland
| |
Collapse
|
32
|
Jhund PS. Improving heart failure outcomes with sodium-glucose cotransporter 2 inhibitors in different patient groups. Diabetes Obes Metab 2023; 25 Suppl 3:26-32. [PMID: 37334518 PMCID: PMC10946463 DOI: 10.1111/dom.15171] [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: 02/24/2023] [Revised: 05/18/2023] [Accepted: 05/26/2023] [Indexed: 06/20/2023]
Abstract
Sodium-glucose cotransporter 2 inhibitors (SGLT-2is) were originally developed for the treatment of hyperglycaemia in type 2 diabetes. Because of regulatory requirements to show the safety of this new class of drugs, a large randomized cardiovascular (CV) outcomes trial was completed but this showed that instead of having a neutral effect on heart failure (HF) outcomes, that these drugs could reduce HF outcomes in this population. Subsequent trials with SGLT-2is have shown that HF hospitalizations are reduced by 30% and CV death or HF hospitalization by 21% in patients with type 2 diabetes. These findings have extended to patients with HF with reduced and mildly reduced or preserved ejection fraction in whom further HF hospitalizations are reduced by 28% and CV death or HF hospitalizations reduced by 23%, and that it is becoming a central therapy for the treatment of HF. Moreover, the benefit in patients with HF is observed regardless of the presence or absence of type 2 diabetes. Similarly, in patients with chronic kidney disease and albuminuria, with and without type 2 diabetes, the benefit of SGLT-2is is clearly seen with a 44% reduction in HF hospitalization and 25% reduction in CV death or HF hospitalization. These trials support the use of SGLT-2is in improving HF outcomes in a broad range of patients, from those with type 2 diabetes, chronic kidney disease and those with pre-existing HF regardless of ejection fraction.
Collapse
Affiliation(s)
- Pardeep S. Jhund
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic HealthUniversity of GlasgowGlasgowUK
| |
Collapse
|
33
|
Stöllberger C, Finsterer J, Schneider B. Adverse events and drug-drug interactions of sodium glucose co-transporter 2 inhibitors in patients treated for heart failure. Expert Rev Cardiovasc Ther 2023; 21:803-816. [PMID: 37856368 DOI: 10.1080/14779072.2023.2273900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/18/2023] [Indexed: 10/21/2023]
Abstract
INTRODUCTION Sodium glucose co-transporter 2-inhibitors (SGLT2-I), antihyperglycemic agents, are increasingly prescribed in chronic heart failure (CHF). Their risk for drug-drug interactions (DDI) seems low. Safety-data derive mainly from diabetes-patients. This review aims to summarize adverse-events (AE) and DDI of the SGLT2-I dapagliflozin, empagliflozin and sotagliflozin in patients with CHF. AREAS COVERED Literature-search-terms in PubMed were 'adverse event/drug-drug interaction' and 'heart failure AND 'dapagliflozin' OR 'empagliflozin' OR 'sotagliflozin.'AEreported in randomized controlled trials (RCT) comprisegenitaland urinary-tract infections, hypotension, ketoacidosis, renal impairment, hypoglycemia, limb-amputations, Fournier's gangrene, bone-fractures, hepatopathy, pancreatitis, diarrhea, malignancy and venous thromboembolism. Their incidence is largely unknown, since they were not consistently evaluated in RCT of CHF. Further AE from meta-analyses, pharmacovigilance reports, case-series and case-reports include erythrocytosis, hypertriglyceridemia, myopathy, sarcopenia, skin problems, ventricular tachycardia, and urinary retention. The maximal observation period of RCT in CHF was 26 months.DDI were mainly studied in healthy volunteers for 3-8 days. In CHF or diabetes-patients, DDI were reported with interleukin-17-inhibitors, linezolid, lithium, tacrolimus, valproate, angiotensin-receptor-neprilysin-inhibitors and intravenous iron. EXPERT OPINION Guidelines recommend treatment with SGLT2-I for CHF but no data on AE during long-term therapy and only little information on DDI are available, which stresses the need for further research. Evidence-based recommendations for ketoacidosis-prevention are desirable.
Collapse
|
34
|
Sato T, Kouzu H, Yano T, Sakuma I, Furuhashi M, Tohse N. Potential favorable action of sodium-glucose cotransporter-2 inhibitors on sudden cardiac death: a brief overview. Front Cardiovasc Med 2023; 10:1159953. [PMID: 37252114 PMCID: PMC10214280 DOI: 10.3389/fcvm.2023.1159953] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/21/2023] [Indexed: 05/31/2023] Open
Abstract
The primary pharmacological action of sodium-glucose co-transporter 2 (SGLT2) inhibitors is to inhibit the reabsorption of glucose and sodium ions from the proximal tubules of the kidney and to promote urinary glucose excretion. Notably, several clinical trials have recently demonstrated potent protective effects of SGLT2 inhibitors in patients with heart failure (HF) or chronic kidney disease (CKD), regardless of the presence or absence of diabetes. However, the impact of SGLT2 inhibitors on sudden cardiac death (SCD) or fatal ventricular arrhythmias (VAs), the pathophysiology of which is partly similar to that of HF and CKD, remains undetermined. The cardiorenal protective effects of SGLT2 inhibitors have been reported to include hemodynamic improvement, reverse remodeling of the failing heart, amelioration of sympathetic hyperactivity, correction of anemia and impaired iron metabolism, antioxidative effects, correction of serum electrolyte abnormalities, and antifibrotic effects, which may lead to prevent SCD and/or VAs. Recently, as possible direct cardiac effects of SGLT2 inhibitors, not only inhibition of Na+/H+ exchanger (NHE) activity, but also suppression of late Na+ current have been focused on. In addition to the indirect cardioprotective mechanisms of SGLT2 inhibitors, suppression of aberrantly increased late Na+ current may contribute to preventing SCD and/or VAs via restoration of the prolonged repolarization phase in the failing heart. This review summarizes the results of previous clinical trials of SGLT2 inhibitors for prevention of SCD, their impact on the indices of electrocardiogram, and the possible molecular mechanisms of their anti-arrhythmic effects.
Collapse
Affiliation(s)
- Tatsuya Sato
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Cardiovascular, Renal, and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hidemichi Kouzu
- Department of Cardiovascular, Renal, and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toshiyuki Yano
- Department of Cardiovascular, Renal, and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Ichiro Sakuma
- Caress Sapporo Hokko Memorial Clinic, Sapporo, Japan
| | - Masato Furuhashi
- Department of Cardiovascular, Renal, and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Noritsugu Tohse
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
35
|
Conte F, Sam JE, Lefeber DJ, Passier R. Metabolic Cardiomyopathies and Cardiac Defects in Inherited Disorders of Carbohydrate Metabolism: A Systematic Review. Int J Mol Sci 2023; 24:ijms24108632. [PMID: 37239976 DOI: 10.3390/ijms24108632] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/25/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
Heart failure (HF) is a progressive chronic disease that remains a primary cause of death worldwide, affecting over 64 million patients. HF can be caused by cardiomyopathies and congenital cardiac defects with monogenic etiology. The number of genes and monogenic disorders linked to development of cardiac defects is constantly growing and includes inherited metabolic disorders (IMDs). Several IMDs affecting various metabolic pathways have been reported presenting cardiomyopathies and cardiac defects. Considering the pivotal role of sugar metabolism in cardiac tissue, including energy production, nucleic acid synthesis and glycosylation, it is not surprising that an increasing number of IMDs linked to carbohydrate metabolism are described with cardiac manifestations. In this systematic review, we offer a comprehensive overview of IMDs linked to carbohydrate metabolism presenting that present with cardiomyopathies, arrhythmogenic disorders and/or structural cardiac defects. We identified 58 IMDs presenting with cardiac complications: 3 defects of sugar/sugar-linked transporters (GLUT3, GLUT10, THTR1); 2 disorders of the pentose phosphate pathway (G6PDH, TALDO); 9 diseases of glycogen metabolism (GAA, GBE1, GDE, GYG1, GYS1, LAMP2, RBCK1, PRKAG2, G6PT1); 29 congenital disorders of glycosylation (ALG3, ALG6, ALG9, ALG12, ATP6V1A, ATP6V1E1, B3GALTL, B3GAT3, COG1, COG7, DOLK, DPM3, FKRP, FKTN, GMPPB, MPDU1, NPL, PGM1, PIGA, PIGL, PIGN, PIGO, PIGT, PIGV, PMM2, POMT1, POMT2, SRD5A3, XYLT2); 15 carbohydrate-linked lysosomal storage diseases (CTSA, GBA1, GLA, GLB1, HEXB, IDUA, IDS, SGSH, NAGLU, HGSNAT, GNS, GALNS, ARSB, GUSB, ARSK). With this systematic review we aim to raise awareness about the cardiac presentations in carbohydrate-linked IMDs and draw attention to carbohydrate-linked pathogenic mechanisms that may underlie cardiac complications.
Collapse
Affiliation(s)
- Federica Conte
- Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, 7522 NH Enschede, The Netherlands
| | - Juda-El Sam
- Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Dirk J Lefeber
- Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Robert Passier
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, 7522 NH Enschede, The Netherlands
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
36
|
Shi JF, Liu Y, Wang Y, Gao R, Wang Y, Liu J. Targeting ferroptosis, a novel programmed cell death, for the potential of alcohol-related liver disease therapy. Front Pharmacol 2023; 14:1194343. [PMID: 37214434 PMCID: PMC10196366 DOI: 10.3389/fphar.2023.1194343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 04/27/2023] [Indexed: 05/24/2023] Open
Abstract
Ferroptosis is a new iron-dependent cell death mode, which is different from the other types of programmed cell death, such as apoptosis, necrosis, and autophagy. Ferroptosis is characterized by a process in which fatal lipids from lipid peroxidation accumulate in cells and eventually lead to cell death. Alcohol-related liver disease (ALD) is a type of liver injury caused by excessive alcohol intake. Alcohol-related liver disease is a broad-spectrum disease category, which includes fatty liver, steatohepatitis, hepatitis, cirrhosis, and hepatocellular tumors. Recent studies have found that ferroptosis is involved in the pathological development of non-viral liver diseases. Therefore, ferroptosis may be an ideal target for the treatment of non-viral liver diseases. In this review article, we will elaborate the molecular mechanism and regulatory mechanism of ferroptosis, explore the key role of ferroptosis in the Alcohol-related liver disease process, and summarize the existing targeted ferroptosis drugs and their feasibility for the treatment of Alcohol-related liver disease.
Collapse
Affiliation(s)
- Jing-Fen Shi
- Institute for Health Policy and Hospital Management, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Wenjiang District People’s Hospital of Chengdu, Chengdu, China
| | - Yu’e Liu
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital of Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Yan Wang
- Wenjiang District People’s Hospital of Chengdu, Chengdu, China
| | - Ru Gao
- Wenjiang District People’s Hospital of Chengdu, Chengdu, China
| | - Yi Wang
- Department of Critical Care Medicine, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jun Liu
- Wenjiang District People’s Hospital of Chengdu, Chengdu, China
- Department of Ultrasound Medicine, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
37
|
Packer M. Dosing of iron supplementation for iron-deficient patients with heart failure: should we prefer more intensive or less intensive repletion targets? Eur J Heart Fail 2023; 25:454-456. [PMID: 36853087 DOI: 10.1002/ejhf.2816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/20/2023] [Accepted: 02/26/2023] [Indexed: 03/01/2023] Open
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Dallas, TX, USA.,Imperial College, London, UK
| |
Collapse
|
38
|
Packer M. Alleviation of functional iron deficiency by SGLT2 inhibition in patients with type 2 diabetes. Diabetes Obes Metab 2022; 25:1143-1146. [PMID: 36583283 DOI: 10.1111/dom.14963] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/10/2022] [Accepted: 12/25/2022] [Indexed: 12/31/2022]
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Dallas, TX, USA
- Imperial College, London, UK
| |
Collapse
|