101
|
Čertíková Chábová V, Zakiyanov O. Sodium Glucose Cotransporter-2 Inhibitors: Spotlight on Favorable Effects on Clinical Outcomes beyond Diabetes. Int J Mol Sci 2022; 23:2812. [PMID: 35269954 PMCID: PMC8911473 DOI: 10.3390/ijms23052812] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 12/16/2022] Open
Abstract
Sodium glucose transporter type 2 (SGLT2) molecules are found in proximal tubules of the kidney, and perhaps in the brain or intestine, but rarely in any other tissue. However, their inhibitors, intended to improve diabetes compensation, have many more beneficial effects. They improve kidney and cardiovascular outcomes and decrease mortality. These benefits are not limited to diabetics but were also found in non-diabetic individuals. The pathophysiological pathways underlying the treatment success have been investigated in both clinical and experimental studies. There have been numerous excellent reviews, but these were mostly restricted to limited aspects of the knowledge. The aim of this review is to summarize the known experimental and clinical evidence of SGLT2 inhibitors' effects on individual organs (kidney, heart, liver, etc.), as well as the systemic changes that lead to an improvement in clinical outcomes.
Collapse
Affiliation(s)
- Věra Čertíková Chábová
- Department of Nephrology, 1st Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, 12800 Prague 2, Czech Republic;
| | | |
Collapse
|
102
|
Hesp AC, Smits MM, van Bommel EJM, Muskiet MHA, Tonneijck L, Nieuwdorp M, Kramer MHH, Joles JA, Bjornstad P, van Raalte DH. Kidney hemodynamic profile and systemic vascular function in adults with type 2 diabetes: Analysis of three clinical trials. J Diabetes Complications 2022; 36:108127. [PMID: 35067449 DOI: 10.1016/j.jdiacomp.2022.108127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/27/2021] [Accepted: 01/06/2022] [Indexed: 01/14/2023]
Abstract
AIMS Glomerular hyperfiltration plays a key role in the pathophysiology of diabetic kidney disease (DKD). Mechanisms underlying this adverse hemodynamic profile are incompletely understood. We hypothesized that systemic vascular pathology, including endothelial dysfunction and arterial stiffness, relates to glomerular hyperfiltration indicated by filtration fraction (FF). METHODS Baseline data of three trials of overweight adults with type 2 diabetes (TD2, n = 111) with relatively well preserved kidney function were analyzed. Glomerular filtration rate (GFR), effective renal plasma flow (ERPF), and FF, were assessed with gold-standard clearance techniques. Systemic vascular resistance (SVR), an indicator of endothelial dysfunction, and pulse pressure (PP), a measure of arterial stiffness, were derived from continuous beat-to-beat monitoring. RESULTS SVR related negatively to GFR (β: -0.382, p < 0.001) and ERPF (β: -0.475, p < 0.001), and positively to FF (β:0.369, p < 0.001). Associations between SVR, ERPF and FF persisted after multivariable adjustments.. PP was negatively related to ERPF (β: -0.252, p = 0.008), and positively to FF (β: 0.257, p = 0.006), of which the latter remained significant in multivariable regression. CONCLUSION Parameters of systemic vascular pathology, including endothelial dysfunction and arterial stiffness, relate to an adverse kidney hemodynamic profile characterized by glomerular hyperfiltration, which predisposes to the development of DKD.
Collapse
Affiliation(s)
- Anne C Hesp
- Department of Internal Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands.
| | - Mark M Smits
- Department of Internal Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands.
| | - Erik J M van Bommel
- Department of Internal Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands.
| | - Marcel H A Muskiet
- Department of Internal Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands.
| | - Lennart Tonneijck
- Department of Internal Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands.
| | - Max Nieuwdorp
- Department of Internal Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands.
| | - Mark H H Kramer
- Department of Internal Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands.
| | - Jaap A Joles
- Department of Nephrology & Hypertension, University Medical Center Utrecht, Utrecht, the Netherlands.
| | - Petter Bjornstad
- Department of Pediatrics-Endocrinology, University of Colorado School of Medicine, Aurora, CO, USA.
| | - Daniël H van Raalte
- Department of Internal Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands.
| |
Collapse
|
103
|
Gembillo G, Cernaro V, Giuffrida AE, Russo G, Giandalia A, Siligato R, Longhitano E, Santoro D. Gender differences in new hypoglycemic drug effects on renal outcomes: a systematic review. Expert Rev Clin Pharmacol 2022; 15:323-339. [PMID: 35300556 DOI: 10.1080/17512433.2022.2055546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 01/28/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Lifetime diabetes risk is greater in women than in men. Women with diabetes mellitus (DM) have a greater prevalence of diabetic kidney disease (DKD) risk factors. The diagnosis of DM is often delayed in women, with poorer outcomes and with expected therapeutic goals missed. AREA COVERED A systematic literature review following PRISMA guidelines was conducted in the PubMed gateway of the MEDLINE database and Clinicaltrials.gov. The purpose of our research was to establish the sex differences on renal outcomes in users of the new hypoglycemic drugs: sodium-glucose transport protein 2 inhibitors (SGLT-2i), dipeptidyl peptidase-IV Inhibitors (DPP-IVi) and glucagon-like peptide-1 inhibitors (GLP-1i). EXPERT OPINION New hypoglycemic drugs represent promising tools in the treatment and prevention of severe complications of diabetes, cardiovascular diseases and chronic kidney disease. Even if renal outcomes are investigated in both randomized controlled trials and cardiovascular outcome trials, gender-based analysis is not always performed. Our systematic review demonstrated that the gap among sexes in DKD can be partially filled using new hypoglycemic drugs. Sexual dimorphism analysis could represent a keystone for the development of adequate gender-specific therapies.
Collapse
Affiliation(s)
- Guido Gembillo
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, Messina Italy
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Italy
| | - Valeria Cernaro
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, Messina Italy
| | - Alfio Edoardo Giuffrida
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, Messina Italy
| | - Giuseppina Russo
- Department of Clinical and Experimental Medicine, University of Messina, Messina Italy
| | - Annalisa Giandalia
- Department of Clinical and Experimental Medicine, University of Messina, Messina Italy
| | - Rossella Siligato
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, Messina Italy
| | - Elisa Longhitano
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, Messina Italy
| | - Domenico Santoro
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, Messina Italy
| |
Collapse
|
104
|
Makvandi K, Hockings PD, Jensen G, Unnerstall T, Leonhardt H, Jarl LV, Englund C, Francis S, Sundgren AK, Hulthe J, Baid-Agrawal S. Multiparametric Magnetic Resonance Imaging Allows Non-Invasive Functional and Structural Evaluation of Diabetic Kidney Disease. Clin Kidney J 2022; 15:1387-1402. [PMID: 35756740 PMCID: PMC9217657 DOI: 10.1093/ckj/sfac054] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Indexed: 11/15/2022] Open
Abstract
Background We sought to develop a novel non-contrast multiparametric MRI (mpMRI) protocol employing several complementary techniques in a single scan session for a comprehensive functional and structural evaluation of diabetic kidney disease (DKD). Methods In the cross-sectional part of this prospective observational study, 38 subjects ages 18‒79 years with type 2 diabetes and DKD [estimated glomerular filtration rate (eGFR) 15‒60 mL/min/1.73 m2] and 20 age- and gender-matched healthy volunteers (HVs) underwent mpMRI. Repeat mpMRI was performed on 23 DKD subjects and 10 HVs. By measured GFR (mGFR), 2 DKD subjects had GFR stage G2, 16 stage G3 and 20 stage G4/G5. A wide range of MRI biomarkers associated with kidney haemodynamics, oxygenation and macro/microstructure were evaluated. Their optimal sensitivity, specificity and repeatability to differentiate diabetic versus healthy kidneys and categorize various stages of disease as well as their correlation with mGFR/albuminuria was assessed. Results Several MRI biomarkers differentiated diabetic from healthy kidneys and distinct GFR stages (G3 versus G4/G5); mean arterial flow (MAF) was the strongest predictor (sensitivity 0.94 and 1.0, specificity 1.00 and 0.69; P = .04 and .004, respectively). Parameters significantly correlating with mGFR were specific measures of kidney haemodynamics, oxygenation, microstructure and macrostructure, with MAF being the strongest univariate predictor (r = 0.92; P < .0001). Conclusions A comprehensive and repeatable non-contrast mpMRI protocol was developed that, as a single, non-invasive tool, allows functional and structural assessment of DKD, which has the potential to provide valuable insights into underlying pathophysiology, disease progression and analysis of efficacy/mode of action of therapeutic interventions in DKD.
Collapse
Affiliation(s)
- Kianoush Makvandi
- Department of Molecular and Clinical Medicine/Nephrology, The Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Paul D Hockings
- Antaros Medical, Molndal, Sweden
- MedTech West, Chalmers University of Technology, Gothenburg, Sweden
| | - Gert Jensen
- Department of Molecular and Clinical Medicine/Nephrology, The Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Tim Unnerstall
- Department of Radiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Henrik Leonhardt
- Department of Radiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | | | - Susan Francis
- Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, UK
| | - Anna K Sundgren
- Late-Stage Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Seema Baid-Agrawal
- Department of Molecular and Clinical Medicine/Nephrology, The Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
- Transplant Center, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
105
|
Meta-Analysis Addressing the Effect of Sodium-Glucose Cotransporter 2 Inhibitors on Flow-Mediated Dilation in Patients With Type 2 Diabetes Mellitus. Am J Cardiol 2022; 165:133-135. [PMID: 34857364 DOI: 10.1016/j.amjcard.2021.11.003] [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: 10/30/2021] [Accepted: 11/09/2021] [Indexed: 11/22/2022]
|
106
|
Andreadi A, Bellia A, Di Daniele N, Meloni M, Lauro R, Della-Morte D, Lauro D. The molecular link between oxidative stress, insulin resistance, and type 2 diabetes: A target for new therapies against cardiovascular diseases. Curr Opin Pharmacol 2022; 62:85-96. [PMID: 34959126 DOI: 10.1016/j.coph.2021.11.010] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 02/07/2023]
Abstract
Type 2 Diabetes Mellitus (T2D) is a chronic disease with a pandemic incidence whose pathogenesis has not yet been clarified. Raising evidence highlighted the role of oxidative stress in inducing insulin resistance, pancreatic beta-cell dysfunction, and leading to cardiovascular disease (CVD). Therefore, understanding the link between oxidative stress, T2D and CVD may help to further understand the pathological processes beyond this association, to personalize the algorithm of the cure, and to find new therapeutic targets. Here, we discussed the role of oxidative stress and the decrease of antioxidant defenses in the pathogenesis of T2D. Furthermore, some aspects of hypoglycemic therapies and their potential role as antioxidant agents were examined, which might be pivotal in preventing CVD in T2D patients.
Collapse
Affiliation(s)
- Aikaterini Andreadi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; University Hospital Fondazione Policlinico Tor Vergata, Rome, Italy
| | - Alfonso Bellia
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; University Hospital Fondazione Policlinico Tor Vergata, Rome, Italy
| | - Nicola Di Daniele
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; University Hospital Fondazione Policlinico Tor Vergata, Rome, Italy
| | - Marco Meloni
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; University Hospital Fondazione Policlinico Tor Vergata, Rome, Italy
| | - Renato Lauro
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - David Della-Morte
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; University Hospital Fondazione Policlinico Tor Vergata, Rome, Italy; San Raffaele Rome Open University, Rome, Italy; Department of Neurology, Evelyn F. McKnight Brain Institute, Miller School, Miami, USA
| | - Davide Lauro
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; University Hospital Fondazione Policlinico Tor Vergata, Rome, Italy.
| |
Collapse
|
107
|
El-Shafey M, El-Agawy MSED, Eldosoky M, Ebrahim HA, Elsherbini DMA, El-Sherbiny M, Asseri SM, Elsherbiny NM. Role of Dapagliflozin and Liraglutide on Diabetes-Induced Cardiomyopathy in Rats: Implication of Oxidative Stress, Inflammation, and Apoptosis. Front Endocrinol (Lausanne) 2022; 13:862394. [PMID: 35370937 PMCID: PMC8972060 DOI: 10.3389/fendo.2022.862394] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 02/15/2022] [Indexed: 12/19/2022] Open
Abstract
The current study aims to assess the protective effects of dapagliflozin (Dapa; a sodium-glucose cotransporter-2 inhibitor) and/or liraglutide (Lira; a glucagon-like peptide 1 agonist) in an experimental model of diabetic cardiomyopathy (DCM). A single dose of streptozotocin (STZ) was administrated to male Sprague-Dawley rats by intraperitoneal injection at a dose of 50 mg/kg to induce diabetes mellitus (DM). Dapa (1 mg/kg, orally), Lira (0.4 mg/kg, s.c.), and Dapa-Lira combination were administrated for 8 weeks once-daily. Blood samples were evaluated for glucose level and biochemical markers of cardiac functions. Cardiac tissue was dissected and assessed for redox homeostasis (malondialdehyde (MDA), glutathione (GSH), and catalase (CAT)), pro-inflammatory mediators (NF-κB and tumor necrosis factor-α (TNF-α)), and apoptotic effectors (caspase-3). Moreover, the effect of treatments on the cardiac cellular structure was studied. Dapa and/or Lira administration resulted in significant improvement of biochemical indices of cardiac function. Additionally, all treatment groups demonstrated restoration of oxidant/antioxidant balance. Moreover, inflammation and apoptosis key elements were markedly downregulated in cardiac tissue. Also, histological studies demonstrated attenuation of diabetes-induced cardiac tissue injury. Interestingly, Dapa-Lira combination treatment produced a more favorable protective effect as compared to a single treatment. These data demonstrated that Dapa, Lira, and their combination therapy could be useful in protection against DM-accompanied cardiac tissue injury, shedding the light on their possible utilization as adjuvant therapy for the management of DM patients.
Collapse
Affiliation(s)
- Mohamed El-Shafey
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Physiological Sciences Department, Fakeeh College for Medical Sciences, Jeddah, Saudi Arabia
| | | | - Mohamed Eldosoky
- Department of Neuroscience Technology-College of Applied Sciences, Jubail Imam Abdulraman bin Faisal University, Dammam, Saudi Arabia
| | - Hasnaa Ali Ebrahim
- Department of Basic Medical Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Dalia Mahmoud Abdelmonem Elsherbini
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
- *Correspondence: Mohamed El-Sherbiny, ; Nehal M. Elsherbiny,
| | - Saad Mohamed Asseri
- Department of Clinical Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
| | - Nehal M. Elsherbiny
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
- *Correspondence: Mohamed El-Sherbiny, ; Nehal M. Elsherbiny,
| |
Collapse
|
108
|
Han Z, Kang X, Zhang J, Wang J, Liu Y, Liu J, Wu Z, Li X, Zhao X, Guo X, Chen S, Tao L. Glycated Hemoglobin and Risk of Arterial Stiffness in a Chinese Han Population: A Longitudinal Study. Front Endocrinol (Lausanne) 2022; 13:854875. [PMID: 35574013 PMCID: PMC9098962 DOI: 10.3389/fendo.2022.854875] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND AND AIMS Glycated hemoglobin (HbA1c) associates with the risk of arterial stiffness, and such association can be found between fasting blood glucose (FBG), postprandial blood glucose (PBG), triglyceride-glucose index (TyG index), and arterial stiffness. However, the results were inconsistent, longitudinal studies were sparse, and comparison of these glycemic parameters was less conducted. We aimed to explore the longitudinal relationship between HbA1c and arterial stiffness and compare the effect of the parameters. METHODS Data were collected from 2011 to 2019 in Beijing Health Management Cohort (BHMC) study. Cox proportional hazard models were fitted to investigate the association between the parameters and arterial stiffness. A generalized estimation equation (GEE) analysis was conducted to investigate the effect of repeated measurements of glycemic parameters. A receiver operating characteristic (ROC) analysis was performed to compare the predictive value of glycemic parameters for arterial stiffness. RESULTS Among 3,048 subjects, 591 were diagnosed as arterial stiffness during the follow-up. The adjusted hazard ratio (HR) [95% confidence interval (CI)] for arterial stiffness of the highest quartile group of HbA1c was 1.63 (1.22-2.18), which was higher than those of FBG, PBG, and TyG index. The nonlinear association of arterial stiffness with HbA1c and PBG was proved. The robust results of the sensitivity analysis were obtained. CONCLUSIONS HbA1c is an important risk factor of arterial stiffness compared with PBG, FBG, and TyG index, and has a strong predictive ability for arterial stiffness among non-diabetics and the general population.
Collapse
Affiliation(s)
- Ze Han
- School of Public Health, Capital Medical University, Beijing, China
- Department of Epidemiology and Health Statistics, Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Xiaoping Kang
- Rehabilitation Centre, Beijing Xiaotangshan Hospital, Beijing, China
| | - Jie Zhang
- School of Public Health, Capital Medical University, Beijing, China
- Department of Epidemiology and Health Statistics, Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Jinqi Wang
- School of Public Health, Capital Medical University, Beijing, China
- Department of Epidemiology and Health Statistics, Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Yue Liu
- School of Public Health, Capital Medical University, Beijing, China
- Department of Epidemiology and Health Statistics, Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Jia Liu
- School of Public Health, Capital Medical University, Beijing, China
- Department of Epidemiology and Health Statistics, Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Zhiyuan Wu
- School of Public Health, Capital Medical University, Beijing, China
- Department of Public Health, School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
| | - Xia Li
- Department of Mathematics and Statistics, La Trobe University, Melbourne, VIC, Australia
| | - Xiaoyu Zhao
- School of Public Health, Capital Medical University, Beijing, China
- Department of Epidemiology and Health Statistics, Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Xiuhua Guo
- School of Public Health, Capital Medical University, Beijing, China
- Department of Epidemiology and Health Statistics, Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Shuo Chen
- Information Department, Beijing Physical Examination Center, Beijing, China
- *Correspondence: Lixin Tao, ; Shuo Chen,
| | - Lixin Tao
- School of Public Health, Capital Medical University, Beijing, China
- Department of Epidemiology and Health Statistics, Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
- *Correspondence: Lixin Tao, ; Shuo Chen,
| |
Collapse
|
109
|
Pelusi C. The Effects of the New Therapeutic Treatments for Diabetes Mellitus on the Male Reproductive Axis. Front Endocrinol (Lausanne) 2022; 13:821113. [PMID: 35518937 PMCID: PMC9065269 DOI: 10.3389/fendo.2022.821113] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
One of the complications of chronic hyperglycemia and insulin resistance due to type 2 diabetes mellitus (T2DM) on the hypothalamic-pituitary-gonadal axis in men, is the high prevalence of hypogonadotropic hypogonadism, which has been recently defined as functional hypogonadism, characterized by low testosterone associated with inappropriately normal gonadotropin levels. Although the pathophysiology of this hormonal imbalance may be related to several factors, including glycemic control, concomitant sleep apnea, insulin resistance, the main role is determined by the degree of central or visceral obesity and the consequent inflammatory state. Several drugs have been developed to treat T2DM such as glucagon-like peptide-1 receptor agonists, dipeptidyl peptidase 4 inhibitors, and sodium-glucose co-transporter 2 inhibitors. All appear to be effective in ameliorating blood glucose control, by lowering inflammation and body weight, and most seem to reduce the risk of micro- and macrovascular damage as a consequence of uncontrolled diabetes. A few studies have evaluated the impact of these drugs on gonadal function in T2DM patients with hypogonadism, with promising results. This review summarizes the main current knowledge of the effects of these new antidiabetic drugs on the hypothalamus-pituitary-gonadal axis, showing their potential future application in addition to glucose control in dysmetabolic male patients.
Collapse
Affiliation(s)
- Carla Pelusi
- Division of Endocrinology and Diabetes Prevention and Care, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
- *Correspondence: Carla Pelusi,
| |
Collapse
|
110
|
Liu H, Sridhar VS, Boulet J, Dharia A, Khan A, Lawler PR, Cherney DZI. Cardiorenal protection with SGLT2 inhibitors in patients with diabetes mellitus: from biomarkers to clinical outcomes in heart failure and diabetic kidney disease. Metabolism 2022; 126:154918. [PMID: 34699838 DOI: 10.1016/j.metabol.2021.154918] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 12/23/2022]
Abstract
Type 2 diabetes (T2D) is one of the most common causes of chronic kidney disease (CKD) and cardiovascular (CV) disease. Until recently, glycemic and BP control were the cornerstones for preventing progression of CKD and CV disease associated with T2D. However, there has been a paradigm shift in treatment since the publication of the first clinical trial demonstrating benefits of sodium glucose cotransporter 2 (SGLT2) inhibitors in 2015. SGLT2 inhibitors have been shown to reduce the risk of major adverse CV events and progression of kidney disease in the setting of T2D. However, the elucidation of mechanisms of underlying these clinical benefits is the subject of ongoing investigation. Experimental studies have shown that SGLT2 inhibitors have diverse pleiotropic effects such as modulation of neurohormones such as the renin-angiotensin-aldosterone system, increasing hematocrit, altering energy substrate use, and attenuating systemic inflammation and oxidative stress, all of which have been implicated in the CV and kidney protective effects of SGLT2 inhibitors. In this review, we highlight biomarkers linked with diabetic kidney disease and heart failure and discuss how SGLT2 inhibitor-associated changes potentially mediate the cardiorenal protection observed with these therapies.
Collapse
Affiliation(s)
- Hongyan Liu
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Vikas S Sridhar
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, Ontario, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jacinthe Boulet
- Department of Medicine, Division of Cardiology, Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
| | - Atit Dharia
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, Ontario, Canada
| | - Abid Khan
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, Ontario, Canada
| | - Patrick R Lawler
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada; Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada; Division of Cardiology and Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON, Canada
| | - David Z I Cherney
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
111
|
Wei R, Wang W, Pan Q, Guo L. Effects of SGLT-2 Inhibitors on Vascular Endothelial Function and Arterial Stiffness in Subjects With Type 2 Diabetes: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Front Endocrinol (Lausanne) 2022; 13:826604. [PMID: 35250882 PMCID: PMC8889103 DOI: 10.3389/fendo.2022.826604] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/19/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE This systematic review and meta-analysis aimed to evaluate the effects of SGLT-2 inhibitors (SGLT-2i) on endothelial function and arteriosclerosis in diabetic patients. METHODS Randomized controlled trials (RCTs) were retrieved from PubMed, Embase, Cochrane Library, and Web of Science databases to evaluate the effects of SGLT-2i on endothelial function and atherosclerosis in type 2 diabetic patients. RESULTS We selected 9 RCTs and 2 cohort studys involving 868 patients. Of these, six studies provided flow-mediated dilation (FMD) levels before and after the intervention. The pooled analysis showed that SGLT-2i could significantly improve the FMD compared to the control group (SMD: 0.18, 95% CI: 0.02 ~ 0.34, P = 0.03). Three studies provided the change in FMD before and after the intervention. Pooled analysis showed no significant differences in FMD change between the SGLT-2i group and the control group. (MD: 2.1, 95%-CI: -0.11~4.31, P = 0.06). Five studies showed pulse wave velocity (PWV) results. Pooled analysis showed no significant differences in the change in PWV between the SGLT-2i group and the control group (SMD: 0.11, 95%-CI: - 0.15 ~ 0.37, P = 0.4). CONCLUSIONS The ability of SGLT-2 inhibitors to improve FMD was significant, but there was no significant effect on PWV levels. SGLT-2i was superior to other antidiabetic agents in improving arterial endothelial function.
Collapse
Affiliation(s)
- Ran Wei
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Fifth School of Clinical Medicine, Peking University, Beijing, China
| | - Weihao Wang
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Qi Pan
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Lixin Guo
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Fifth School of Clinical Medicine, Peking University, Beijing, China
- *Correspondence: Lixin Guo,
| |
Collapse
|
112
|
Cignarelli A, Genchi VA, Le Grazie G, Caruso I, Marrano N, Biondi G, D’Oria R, Sorice GP, Natalicchio A, Perrini S, Laviola L, Giorgino F. Mini Review: Effect of GLP-1 Receptor Agonists and SGLT-2 Inhibitors on the Growth Hormone/IGF Axis. Front Endocrinol (Lausanne) 2022; 13:846903. [PMID: 35265043 PMCID: PMC8899086 DOI: 10.3389/fendo.2022.846903] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/01/2022] [Indexed: 11/13/2022] Open
Abstract
Accumulating evidence supports the early use of glucagon-like peptide-1 receptor agonists (GLP-1RAs) and sodium glucose transporter-2 inhibitors (SGLT-2is) for the treatment of type 2 diabetes. Indeed, these compounds exert numerous pleiotropic actions that favorably affect metabolism and diabetes comorbidities, showing an additional effect beyond glucose control. Although a substantial amount of knowledge has been generated regarding the mechanism of action of both drug classes, much remains to be understood. Growth hormone (GH) is an important driver for multiple endocrine responses involving changes in glucose and lipid metabolism, and affects several tissues and organs (e.g., bone, heart). It acts directly on several target tissues, including skeletal muscle and bone, but several effects are mediated indirectly by circulating (liver-derived) or locally produced IGF-1. In consideration of the multiple metabolic and cardiovascular effects seen in subjects treated with GLP-1RAs and SGLT-2is (e.g., reduction of hyperglycemia, weight loss, free/fat mass and bone remodeling, anti-atherosclerosis, natriuresis), it is reasonable to speculate that GH and IGF-1 may play a about a relevant role in this context. This narrative mini-review aims to describe the involvement of the GH/IGF-1/IGF-1R axis in either mediating or responding to the effects of each of the two drug classes.
Collapse
|
113
|
Kolwelter J, Bosch A, Jung S, Stabel L, Kannenkeril D, Ott C, Bramlage P, Schiffer M, Achenbach S, Schmieder RE. Effects of the sodium-glucose cotransporter 2 inhibitor empagliflozin on vascular function in patients with chronic heart failure. ESC Heart Fail 2021; 8:5327-5337. [PMID: 34544205 PMCID: PMC8712907 DOI: 10.1002/ehf2.13622] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 09/04/2021] [Indexed: 01/01/2023] Open
Abstract
AIMS Impairment of vascular function contributes to the progression of chronic heart failure (HF) by increasing the afterload. Treatment with selective sodium-glucose cotransporter 2 (SGLT2) inhibitors improves the prognosis of HF, but the precise mechanisms remain unclear. The aim of this study was to analyse the effect of empagliflozin on vascular function in patients with HF. METHODS AND RESULTS In an investigator initiated, double-blind, randomized, placebo-controlled, parallel-group, clinical study, patients with HF NYHA II-III and an ejection fraction of 49% or less were randomized 2:1 to receive empagliflozin 10 mg once daily or placebo for 3 months. A total of 74 patients (15% female), aged 66 ± 9 years, with a mean ejection fraction of 39 ± 8% and a median NTproBNP of 558 pg/mL (IQR 219-1051 pg/mL), were included. Vascular parameters such as central systolic blood pressure (cSBP), central pulse pressure (cPP), forward (FPH), and reflected pressure pulse height (RPH) decreased under resting conditions after 1 and 3 months (1 month: cSBP -6.4 ± 8.3 mmHg, P < 0.001, cPP -3.0 ± 6.6 mmHg, P = 0.004, FPH -2.5 ± 4.5 mmHg, P = 0.001, RPH -1.6 ± 3.0 mmHg, P = 0.001; 3 months: cSBP -4.6 ± 8.4 mmHg, P = 0.001, cPP -3.1 ± 4.8 mmHg, P < 0.001, FPH -1.7 ± 3.7 mmHg, P = 0.004, RPH -1.4 ± 2.5 mmHg, P = 0.001) in patients treated with empagliflozin (n = 45). In accordance, cSBP and cPP decreased in patients with empagliflozin treatment under 24 h ambulatory conditions after 1 and 3 months (1 month: cSBP -4.8 ± 10.1 mmHg, P = 0.003, cPP -2.0 ± 5.7 mmHg, P = 0.026; 3 months: cSBP -4.7 ± 9.0 mmHg, P = 0.002, cPP -2.1 ± 6.4 mmHg, P = 0.044). In the placebo group, there was no significant change after 1 and 3 months. The decrease in cSBP under resting conditions (-5.7 ± 2.4 mmHg, P = 0.019) after 1 month and in cSBP (-6.0 ± 2.6, P = 0.027) as well as in pulse wave velocity (-0.5 ± 0.2 m/s, P = 0.021) under 24 h ambulatory conditions after 3 months was greater in the empagliflozin group than in the placebo group. CONCLUSIONS We found an improvement of vascular function after treatment with empagliflozin that indicates decreased afterload of the left ventricle and may contribute to the beneficial effects of SGLT2 inhibition in HF.
Collapse
Affiliation(s)
- Julie Kolwelter
- Department of Nephrology and HypertensionUniversity Hospital Erlangen, Friedrich‐Alexander University Erlangen‐Nuremberg (FAU)Ulmenweg 18Erlangen91054Germany
- Department of CardiologyUniversity Hospital Erlangen, Friedrich‐Alexander University Erlangen‐Nuremberg (FAU)ErlangenGermany
| | - Agnes Bosch
- Department of Nephrology and HypertensionUniversity Hospital Erlangen, Friedrich‐Alexander University Erlangen‐Nuremberg (FAU)Ulmenweg 18Erlangen91054Germany
| | - Susanne Jung
- Department of CardiologyUniversity Hospital Erlangen, Friedrich‐Alexander University Erlangen‐Nuremberg (FAU)ErlangenGermany
| | - Lena Stabel
- Department of Nephrology and HypertensionUniversity Hospital Erlangen, Friedrich‐Alexander University Erlangen‐Nuremberg (FAU)Ulmenweg 18Erlangen91054Germany
| | - Dennis Kannenkeril
- Department of Nephrology and HypertensionUniversity Hospital Erlangen, Friedrich‐Alexander University Erlangen‐Nuremberg (FAU)Ulmenweg 18Erlangen91054Germany
| | - Christian Ott
- Department of Nephrology and HypertensionUniversity Hospital Erlangen, Friedrich‐Alexander University Erlangen‐Nuremberg (FAU)Ulmenweg 18Erlangen91054Germany
| | - Peter Bramlage
- Institute for Pharmacology and Preventive MedicineCloppenburgGermany
| | - Mario Schiffer
- Department of Nephrology and HypertensionUniversity Hospital Erlangen, Friedrich‐Alexander University Erlangen‐Nuremberg (FAU)Ulmenweg 18Erlangen91054Germany
| | - Stephan Achenbach
- Department of CardiologyUniversity Hospital Erlangen, Friedrich‐Alexander University Erlangen‐Nuremberg (FAU)ErlangenGermany
| | - Roland E. Schmieder
- Department of Nephrology and HypertensionUniversity Hospital Erlangen, Friedrich‐Alexander University Erlangen‐Nuremberg (FAU)Ulmenweg 18Erlangen91054Germany
| |
Collapse
|
114
|
Madonna R, Barachini S, Moscato S, Ippolito C, Mattii L, Lenzi C, Balistreri CR, Zucchi R, De Caterina R. Sodium-glucose cotransporter type 2 inhibitors prevent ponatinib-induced endothelial senescence and disfunction: A potential rescue strategy. Vascul Pharmacol 2021; 142:106949. [PMID: 34843980 DOI: 10.1016/j.vph.2021.106949] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 11/23/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Ponatinib (PON), a third-generation tyrosine kinase inhibitor (TKI), has proven cardiovascular toxicity, with no known preventing agents usable to limit such side effect. Sodium-glucose cotransporter type 2 (SGLT2) inhibitors are a new class of glucose-lowering agents, featuring favorable cardiac and vascular effects. AIMS We assessed the effects of the SGLT2 inhibitors empagliflozin (EMPA) and dapagliflozin (DAPA) on human aortic endothelial cells (HAECs) and underlying vasculo-protective mechanisms in an in vitro model of PON-induced endothelial toxicity. METHODS AND RESULTS We exposed HAECs to PON or vehicle (DMSO) in the presence or absence of EMPA (100 and 500 nmol/L) or dapagliflozin (DAPA) for 0-48 h exposure times. Compared with vehicle, incubations of HAECs with PON significantly reduced cell viability (0.56 ± 0.11 vs 0.23 ± 0.05 absorbance units, p < 0.01), increased the number of senescent cells at β-gal-assay (PON 9 ± 4 vs basal DMSO 3 ± 1 β-Gal+ cells/field, p < 0.01), decreased tubulization in Matrigel (PON PON: 6 ± 1 vs basal DMSO 12 ± 1 tubuli number/field, p < 0.05) with a non-statistically significant trend of PON to decrease the number of autophagic cells at immunofluorescence assay and flow cytometry. EMPA reverted the effects of PON on cell viability (E 500 + PON 0.24 ± 0.05 vs PON 0.56 ± 0.11 absorbance units, p < 0.01) and induced autophagy (E 500 7 ± 4.3 vs basal DMSO 2.6 ± 2.3 mean fluorescence vs PON 2.6 ± 2.4 mean fluorescence, p < 0.05). EMPA and DAPA also reversed the effects of PON on cell senescence (E 500 + PON 4 ± 1 and DAPA 100 4 ± 2 vs PON 9 ± 4 β-Gal+ cells/field, p < 0.01) and improved cell tubulization (E 500 + PON 21 ± 3 vs PON 6 ± 1 tubuli number/field, p < 0.05; DAPA 100 + PON 16 ± 2 vs PON 6 ± 1 tubuli number/field, p < 0.05). CONCLUSION EMPA and DAPA attenuate the vasculo-toxic effect exerted by PON by reverting endothelial cell senescence and dysfunction. These findings support the design of clinical studies exploring the vasculo-protective effects of EMPA or DAPA on PON-induced vascular toxicity.
Collapse
Affiliation(s)
- Rosalinda Madonna
- Department of Pathology, Cardiology Division, University of Pisa, Pisa, Italy.
| | - Serena Barachini
- Department of Clinical and Experimental Medicine, Hematology Division, University of Pisa, Pisa, Italy
| | - Stefania Moscato
- Department of Clinical and Experimental Medicine, Histology Division, University of Pisa, Pisa, Italy
| | - Chiara Ippolito
- Department of Clinical and Experimental Medicine, Histology Division, University of Pisa, Pisa, Italy
| | - Letizia Mattii
- Department of Clinical and Experimental Medicine, Histology Division, University of Pisa, Pisa, Italy
| | - Chiara Lenzi
- Department of Histopathology, Pisa University Hospital, Italy
| | - Carmela Rita Balistreri
- Department of Biomedicine, Neuroscience and Advanced, Diagnostics (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Riccardo Zucchi
- Department of Pathology, Laboratory of Biochemistry, University of Pisa, Italy
| | | |
Collapse
|
115
|
Rey-García J, Townsend RR. Large Artery Stiffness: A Companion to the 2015 AHA Science Statement on Arterial Stiffness. Pulse (Basel) 2021; 9:1-10. [PMID: 34722350 DOI: 10.1159/000518613] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 07/15/2021] [Indexed: 11/19/2022] Open
Abstract
Large artery stiffness (LAS) has proven to be an independent risk factor for cardiovascular disease and mortality. Nevertheless, the position of current hypertension guidelines regarding the usefulness of assessing LAS differs across different continents. In general, European Guidelines recognize pulse wave velocity (PWV) as a marker of target organ damage but do not recommend its systematic use in general population. Asian guidelines consider PWV as a recommended test at diagnosis of hypertension, in contrast to North American guidelines that do not state any position about its usefulness. However, PWV predicts cardiovascular events, and several studies have shown that it improves risk classification adjusting for established risk factors especially for intermediate-risk patients. Finally, some advances have been made related to treatments affecting LAS. Dietary interventions such as sodium restriction and exercise-based interventions have a modest effect in reducing LAS. Pharmacological interventions, such as statins, or more recent advances with mineralocorticoid blocker seem to have a beneficial effect. Last, controversial effects of renal denervation on LAS have been found. Our goal here is to update the reader on LAS on these areas since the 2015 American Heart Association Scientific Statement.
Collapse
Affiliation(s)
- Jimena Rey-García
- Department of Preventive Medicine and Public Health, Universidad Autónoma de Madrid, Madrid, Spain.,Internal Medicine Department, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Raymond R Townsend
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
116
|
Yamaji T, Harada T, Hashimoto Y, Nakano Y, Kajikawa M, Yoshimura K, Aoki G, Chayama K, Goto C, Mizobuchi A, Han Y, Yusoff FM, Kishimoto S, Maruhashi T, Nakashima A, Higashi Y. Relationship between hemoglobin A1c level and flow-mediated vasodilation in patients with type 2 diabetes mellitus receiving antidiabetic drugs. J Diabetes Investig 2021; 13:677-686. [PMID: 34725937 PMCID: PMC9017617 DOI: 10.1111/jdi.13705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 10/13/2021] [Accepted: 10/31/2021] [Indexed: 12/05/2022] Open
Abstract
Aims/Introduction Diabetes mellitus is associated with endothelial dysfunction. However, it is still controversial as to whether antidiabetic drug treatment affects endothelial function. The purpose of this study was to evaluate the relationships of the hemoglobin A1c (HbA1c) level with flow‐mediated vasodilation (FMD) and nitroglycerine‐induced vasodilation (NID) in patients with type 2 diabetes mellitus who are receiving antidiabetic drugs. Materials and Methods The FMD was measured in 866 patients with type 2 diabetes mellitus who were receiving antidiabetic drugs (625 men and 241 women; mean age: 62 ± 10 years). The patients were divided into four groups according to HbA1c levels: <6.5, 6.5–6.9, 7.0–7.9, and ≥8.0%. Results There was an inverted U‐shaped pattern of association of the HbA1c level with the FMD at an HbA1c level of about 7% of the peak of FMD in patients with type 2 diabetes mellitus who were receiving antidiabetic drugs. The FMD was significantly smaller in the HbA1c <6.5% group than in the HbA1c 6.5–6.9% group and the HbA1c 7.0–7.9% group (P < 0.001 and P < 0.001, respectively). The FMD values were similar in the HbA1c <6.5% group and HbA1c ≥8.0% group (P = 0.10). There were no significant differences in NID among the four groups (P = 0.98). Conclusions These findings suggest that a low HbA1c <6.5% as well as a high HbA1c ≥8.0% is associated with endothelial dysfunction in patients with type 2 diabetes mellitus who are receiving antidiabetic drugs and that vascular smooth muscle function is similar in such patients regardless of the HbA1c level.
Collapse
Affiliation(s)
- Takayuki Yamaji
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Takahiro Harada
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Yu Hashimoto
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Yukiko Nakano
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Masato Kajikawa
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Kenichi Yoshimura
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan.,Department of Biostatistics, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Gaku Aoki
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan.,Department of Biostatistics, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University Hiroshima, Japan
| | - Chikara Goto
- Hiroshima International University, Hiroshima, Japan
| | - Aya Mizobuchi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Yiming Han
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Farina Mohamad Yusoff
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Shinji Kishimoto
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Tatsuya Maruhashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Ayumu Nakashima
- Department of Stem Cell Biology and Medicine, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Yukihito Higashi
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan.,Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
117
|
Love KM, Barrett EJ, Malin SK, Reusch JEB, Regensteiner JG, Liu Z. Diabetes pathogenesis and management: the endothelium comes of age. J Mol Cell Biol 2021; 13:500-512. [PMID: 33787922 PMCID: PMC8530521 DOI: 10.1093/jmcb/mjab024] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/10/2021] [Accepted: 02/25/2021] [Indexed: 12/03/2022] Open
Abstract
Endothelium, acting as a barrier, protects tissues against factors that provoke insulin resistance and type 2 diabetes and itself responds to the insult of insulin resistance inducers with altered function. Endothelial insulin resistance and vascular dysfunction occur early in the evolution of insulin resistance-related disease, can co-exist with and even contribute to the development of metabolic insulin resistance, and promote vascular complications in those affected. The impact of endothelial insulin resistance and vascular dysfunction varies depending on the blood vessel size and location, resulting in decreased arterial plasticity, increased atherosclerosis and vascular resistance, and decreased tissue perfusion. Women with insulin resistance and diabetes are disproportionately impacted by cardiovascular disease, likely related to differential sex-hormone endothelium effects. Thus, reducing endothelial insulin resistance and improving endothelial function in the conduit arteries may reduce atherosclerotic complications, in the resistance arteries lead to better blood pressure control, and in the microvasculature lead to less microvascular complications and more effective tissue perfusion. Multiple diabetes therapeutic modalities, including medications and exercise training, improve endothelial insulin action and vascular function. This action may delay the onset of type 2 diabetes and/or its complications, making the vascular endothelium an attractive therapeutic target for type 2 diabetes and potentially type 1 diabetes.
Collapse
MESH Headings
- Age Factors
- Cardiovascular Diseases/epidemiology
- Cardiovascular Diseases/ethnology
- Cardiovascular Diseases/metabolism
- Cardiovascular Diseases/physiopathology
- Comorbidity
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/epidemiology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/physiopathology
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/epidemiology
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/physiopathology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Exercise
- Female
- Humans
- Hypoglycemic Agents/pharmacology
- Hypoglycemic Agents/therapeutic use
- Insulin Resistance
- Male
- Racial Groups
- Risk Factors
- Sex Factors
Collapse
Affiliation(s)
- Kaitlin M Love
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Eugene J Barrett
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Steven K Malin
- Department of Kinesiology and Health, Rutgers University, New Brunswick, NJ, USA
- Division of Endocrinology, Metabolism and Nutrition, Rutgers University, New Brunswick, NJ, USA
- New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, NJ, USA
- Institute of Translational Medicine and Research, Rutgers University, New Brunswick, NJ, USA
| | - Jane E B Reusch
- Center for Women’s Health Research, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, USA
| | - Judith G Regensteiner
- Center for Women’s Health Research, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Zhenqi Liu
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908, USA
| |
Collapse
|
118
|
Yu H, Basu S, Tang W, Penland RC, Greasley PJ, Oscarsson J, Boulton DW, Hallow KM. Predicted Cardiac Functional Responses to Renal Actions of SGLT2i in the DAPACARD Trial Population: A Mathematical Modeling Analysis. J Clin Pharmacol 2021; 62:541-554. [PMID: 34657303 DOI: 10.1002/jcph.1987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/11/2021] [Indexed: 11/07/2022]
Abstract
Sodium-glucose cotransporter-2 inhibitors (SGLT2is) have been shown to reduce the risk of worsening heart failure (HF) in subjects with HF and a reduced ejection fraction (HFrEF) in multiple clinical trials. The DAPACARD clinical trial was conducted to examine the effects of DAPAgliflozin on CARDiac substrate uptake, myocardial efficiency, and myocardial contractile work in type 2 diabetes mellitus (T2DM) subjects. As a complement to the clinical study, a mechanistic mathematical model of cardiorenal physiology was used to quantify the influence of established natriuretic/diuretic effects of SGLT2i on cardiac function (myocardial efficiency and global longitudinal strain). Virtual participants reflecting the participant-level characteristics in the DAPACARD trial were produced by varying model parameters over physiologically plausible ranges. A second virtual population was generated by inducing a state of HFrEF in the DAPACARD T2DM virtual participants (DAPACARD-HFrEF virtual participants) for comparison. Cardiac responses to placebo and SGLT2i were simulated over 42 days. Cardiac hemodynamic improvements were predicted in DAPACARD-HFrEF virtual participants but not in DAPACARD virtual participants. In particular, the natriuresis/diuresis induced by SGLT2i improved the global longitudinal strain and myocardial efficiency in DAPACARD-HFrEF virtual participants within the first 14 days (change from baseline: global longitudinal strain: -0.95% and myocardial efficiency: 0.34%), whereas the global longitudinal strain and myocardial efficiency in DAPACARD virtual participants were slightly worse (change from baseline: global longitudinal strain: 0.35% and myocardial efficiency: -0.01%). The results of the DAPACARD virtual participants modeling were in line with the clinical data but do not preclude additional effects from other mechanisms of SGLT2i. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Hongtao Yu
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, Georgia, USA
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Sanchita Basu
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, Georgia, USA
| | - Weifeng Tang
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Robert C Penland
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Boston, Massachusetts, USA
| | - Peter J Greasley
- Early Clinical Development, Research, and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceutical R&D, AstraZeneca, Gothenburg, Sweden
| | - Jan Oscarsson
- Late Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceutical R&D, AstraZeneca, Gothenburg, Sweden
| | - David W Boulton
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - K Melissa Hallow
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, Georgia, USA
- Department of Epidemiology and Biostatistics, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
119
|
Barraclough JY, Patel S, Yu J, Neal B, Arnott C. The Role of Sodium Glucose Cotransporter-2 Inhibitors in Atherosclerotic Cardiovascular Disease: A Narrative Review of Potential Mechanisms. Cells 2021; 10:cells10102699. [PMID: 34685677 PMCID: PMC8534746 DOI: 10.3390/cells10102699] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/30/2021] [Accepted: 09/30/2021] [Indexed: 12/16/2022] Open
Abstract
Sodium glucose cotransporter 2 (SGLT2) inhibitors are a class of medication with broad cardiovascular benefits in those with type 2 diabetes, chronic kidney disease, and heart failure. These include reductions in major adverse cardiac events and cardiovascular death. The mechanisms that underlie their benefits in atherosclerotic cardiovascular disease (ASCVD) are not well understood, but they extend beyond glucose lowering. This narrative review summarises the ASCVD benefits of SGLT2 inhibitors seen in large human outcome trials, as well as the mechanisms of action explored in rodent and small human studies. Potential pathways include favourable alterations in lipid metabolism, inflammation, and endothelial function. These all require further investigation in large human clinical trials with mechanistic endpoints, to further elucidate the disease modifying benefits of this drug class and those who will benefit most from it.
Collapse
Affiliation(s)
- Jennifer Y. Barraclough
- The George Institute for Global Health, University of New South Wales, Sydney, NSW 2042, Australia; (J.Y.B.); (J.Y.); (B.N.)
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia;
| | - Sanjay Patel
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia;
- Sydney Medical School, University of Sydney, Sydney, NSW 2042, Australia
| | - Jie Yu
- The George Institute for Global Health, University of New South Wales, Sydney, NSW 2042, Australia; (J.Y.B.); (J.Y.); (B.N.)
| | - Bruce Neal
- The George Institute for Global Health, University of New South Wales, Sydney, NSW 2042, Australia; (J.Y.B.); (J.Y.); (B.N.)
| | - Clare Arnott
- The George Institute for Global Health, University of New South Wales, Sydney, NSW 2042, Australia; (J.Y.B.); (J.Y.); (B.N.)
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia;
- Sydney Medical School, University of Sydney, Sydney, NSW 2042, Australia
- Correspondence: ; Tel.: +61-2-8052-4300
| |
Collapse
|
120
|
Salvatore T, Caturano A, Galiero R, Di Martino A, Albanese G, Vetrano E, Sardu C, Marfella R, Rinaldi L, Sasso FC. Cardiovascular Benefits from Gliflozins: Effects on Endothelial Function. Biomedicines 2021; 9:biomedicines9101356. [PMID: 34680473 PMCID: PMC8533063 DOI: 10.3390/biomedicines9101356] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/15/2021] [Accepted: 09/26/2021] [Indexed: 12/23/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a known independent risk factor for atherosclerotic cardiovascular disease (CVD) and solid epidemiological evidence points to heart failure (HF) as one of the most common complications of diabetes. For this reason, it is imperative to consider the prevention of CV outcomes as an effective goal for the management of diabetic patients, as important as lowering blood glucose. Endothelial dysfunction (ED) is an early event of atherosclerosis involving adhesion molecules, chemokines, and leucocytes to enhance low-density lipoprotein oxidation, platelet activation, and vascular smooth muscle cell proliferation and migration. This abnormal vascular phenotype represents an important risk factor for the genesis of any complication of diabetes, contributing to the pathogenesis of not only macrovascular disease but also microvascular damage. Gliflozins are a novel class of anti-hyperglycemic agents used for the treatment of Type 2 diabetes mellitus (T2DM) that selectively inhibit the sodium glucose transporter 2 (SGLT2) in the kidneys and have provoked large interest in scientific community due to their cardiovascular beneficial effects, whose underlying pathophysiology is still not fully understood. This review aimed to analyze the cardiovascular protective mechanisms of SGLT2 inhibition in patients T2DM and their impact on endothelial function.
Collapse
Affiliation(s)
- Teresa Salvatore
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via De Crecchio 7, I-80138 Naples, Italy;
| | - Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (A.C.); (R.G.); (A.D.M.); (G.A.); (E.V.); (C.S.); (R.M.); (L.R.)
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (A.C.); (R.G.); (A.D.M.); (G.A.); (E.V.); (C.S.); (R.M.); (L.R.)
| | - Anna Di Martino
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (A.C.); (R.G.); (A.D.M.); (G.A.); (E.V.); (C.S.); (R.M.); (L.R.)
| | - Gaetana Albanese
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (A.C.); (R.G.); (A.D.M.); (G.A.); (E.V.); (C.S.); (R.M.); (L.R.)
| | - Erica Vetrano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (A.C.); (R.G.); (A.D.M.); (G.A.); (E.V.); (C.S.); (R.M.); (L.R.)
| | - Celestino Sardu
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (A.C.); (R.G.); (A.D.M.); (G.A.); (E.V.); (C.S.); (R.M.); (L.R.)
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (A.C.); (R.G.); (A.D.M.); (G.A.); (E.V.); (C.S.); (R.M.); (L.R.)
| | - Luca Rinaldi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (A.C.); (R.G.); (A.D.M.); (G.A.); (E.V.); (C.S.); (R.M.); (L.R.)
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (A.C.); (R.G.); (A.D.M.); (G.A.); (E.V.); (C.S.); (R.M.); (L.R.)
- Correspondence: ; Tel.: +39-081-566-5010
| |
Collapse
|
121
|
Natali A, Nesti L, Tricò D, Ferrannini E. Effects of GLP-1 receptor agonists and SGLT-2 inhibitors on cardiac structure and function: a narrative review of clinical evidence. Cardiovasc Diabetol 2021; 20:196. [PMID: 34583699 PMCID: PMC8479881 DOI: 10.1186/s12933-021-01385-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/17/2021] [Indexed: 02/08/2023] Open
Abstract
The impressive results of recent clinical trials with glucagon-like peptide-1 receptor agonists (GLP-1Ra) and sodium glucose transporter 2 inhibitors (SGLT-2i) in terms of cardiovascular protection prompted a huge interest in these agents for heart failure (HF) prevention and treatment. While both classes show positive effects on composite cardiovascular endpoints (i.e. 3P MACE), their actions on the cardiac function and structure, as well as on volume regulation, and their impact on HF-related events have not been systematically evaluated and compared. In this narrative review, we summarize and critically interpret the available evidence emerging from clinical studies. While chronic exposure to GLP-1Ra appears to be essentially neutral on both systolic and diastolic function, irrespective of left ventricular ejection fraction (LVEF), a beneficial impact of SGLT-2i is consistently detectable for both systolic and diastolic function parameters in subjects with diabetes with and without HF, with a gradient proportional to the severity of baseline dysfunction. SGLT-2i have a clinically significant impact in terms of HF hospitalization prevention in subjects at high and very high cardiovascular risk both with and without type 2 diabetes (T2D) or HF, while GLP-1Ra have been proven to be safe (and marginally beneficial) in subjects with T2D without HF. We suggest that the role of the kidney is crucial for the effect of SGLT-2i on the clinical outcomes not only because these drugs slow-down the time-dependent decline of kidney function and enhance the response to diuretics, but also because they attenuate the meal-related anti-natriuretic pressure (lowering postprandial hyperglycemia and hyperinsulinemia and preventing proximal sodium reabsorption), which would reduce the individual sensitivity to day-to-day variations in dietary sodium intake.
Collapse
Affiliation(s)
- Andrea Natali
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56100, Pisa, Italy.
| | - Lorenzo Nesti
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56100, Pisa, Italy
| | - Domenico Tricò
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56100, Pisa, Italy
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | | |
Collapse
|
122
|
Gager GM, von Lewinski D, Sourij H, Jilma B, Eyileten C, Filipiak K, Hülsmann M, Kubica J, Postula M, Siller-Matula JM. Effects of SGLT2 Inhibitors on Ion Homeostasis and Oxidative Stress associated Mechanisms in Heart Failure. Biomed Pharmacother 2021; 143:112169. [PMID: 34560555 DOI: 10.1016/j.biopha.2021.112169] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 12/21/2022] Open
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors present a class of antidiabetic drugs, which inhibit renal glucose reabsorption resulting in the elevation of urinary glucose levels. Within the past years, SGLT2 inhibitors have become increasingly relevant due to their effects beyond glycemic control in patients with type 2 diabetes (T2DM). Although dedicated large trials demonstrated cardioprotective effects of SGLT2 inhibitors, the exact mechanisms responsible for those benefits have not been fully identified. Alterations in Ca2+ signaling and oxidative stress accompanied by excessive reactive oxygen species (ROS) production, fibrosis and inflammatory processes form cornerstones of potential molecular targets for SGLT2 inhibitors. This review focused on three hypotheses for SGLT2 inhibitor-mediated cardioprotection: ion homeostasis, oxidative stress and endothelial dysfunction.
Collapse
Affiliation(s)
- Gloria M Gager
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Austria; Department of Clinical Pharmacology, Medical University of Vienna, Austria
| | - Dirk von Lewinski
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Harald Sourij
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Interdisciplinary Metabolic Medicine Trials Unit, Medical University of Graz, Graz, Austria
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Austria
| | - Ceren Eyileten
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland
| | - Krzysztof Filipiak
- First Chair and Department of Cardiology, Medical University of Warsaw, Poland
| | - Martin Hülsmann
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Austria
| | - Jacek Kubica
- Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Marek Postula
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland
| | - Jolanta M Siller-Matula
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Austria; Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland.
| |
Collapse
|
123
|
Pabel S, Hamdani N, Luedde M, Sossalla S. SGLT2 Inhibitors and Their Mode of Action in Heart Failure-Has the Mystery Been Unravelled? Curr Heart Fail Rep 2021; 18:315-328. [PMID: 34523061 PMCID: PMC8484236 DOI: 10.1007/s11897-021-00529-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/09/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW SGLT2 inhibitors (SGLT2i) are new drugs for patients with heart failure (HF) irrespective of diabetes. However, the mechanisms of SGLT2i in HF remain elusive. This article discusses the current clinical evidence for using SGLT2i in different types of heart failure and provides an overview about the possible underlying mechanisms. RECENT FINDINGS Clinical and basic data strongly support and extend the use of SGLT2i in HF. Improvement of conventional secondary risk factors is unlikely to explain the prognostic benefits of these drugs in HF. However, different multidirectional mechanisms of SGLT2i could improve HF status including volume regulation, cardiorenal mechanisms, metabolic effects, improved cardiac remodelling, direct effects on cardiac contractility and ion-homeostasis, reduction of inflammation and oxidative stress as well as an impact on autophagy and adipokines. Further translational studies are needed to determine the mechanisms of SGLT2i in HF. However, basic and clinical evidence encourage the use of SGLT2i in HFrEF and possibly HFpEF.
Collapse
Affiliation(s)
- Steffen Pabel
- Department of Internal Medicine II, University Medical Centre Regensburg, Regensburg, Germany
| | - Nazha Hamdani
- Department of Molecular and Experimental Cardiology and Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Mark Luedde
- Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Samuel Sossalla
- Department of Internal Medicine II, University Medical Centre Regensburg, Regensburg, Germany. .,Clinic for Cardiology and Pneumology, Georg-August University Göttingen, and DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.
| |
Collapse
|
124
|
Latosinska A, Siwy J, Cherney DZ, Perkins BA, Mischak H, Beige J. SGLT2-Inhibition reverts urinary peptide changes associated with severe COVID-19: An in-silico proof-of-principle of proteomics-based drug repurposing. Proteomics 2021; 21:e2100160. [PMID: 34477316 PMCID: PMC8646299 DOI: 10.1002/pmic.202100160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/31/2021] [Indexed: 01/08/2023]
Abstract
Severe COVID‐19 is reflected by significant changes in urine peptides. Based on this observation, a clinical test predicting COVID‐19 severity, CoV50, was developed and registered as in vitro diagnostic in Germany. We have hypothesized that molecular changes displayed by CoV50, likely reflective of endothelial damage, may be reversed by specific drugs. Such an impact by a drug could indicate potential benefits in the context of COVID‐19. To test this hypothesis, urinary peptide data from patients without COVID‐19 prior to and after drug treatment were collected from the human urinary proteome database. The drugs chosen were selected based on availability of sufficient number of participants in the dataset (n > 20) and potential value of drug therapies in the treatment of COVID‐19 based on reports in the literature. In these participants without COVID‐19, spironolactone did not demonstrate a significant impact on CoV50 scoring. Empagliflozin treatment resulted in a significant change in CoV50 scoring, indicative of a potential therapeutic benefit. The study serves as a proof‐of‐principle for a drug repurposing approach based on human urinary peptide signatures. The results support the initiation of a randomized control trial testing a potential positive effect of empagliflozin for severe COVID‐19, possibly via endothelial protective mechanisms.
Collapse
Affiliation(s)
| | | | - David Z Cherney
- Division of Nephrology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Bruce A Perkins
- Division of Endocrinology and Metabolism, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Joachim Beige
- Department of Nephrology and Kuratorium for Dialysis and Transplantation (KfH) Renal Unit, Hospital St. Georg, Leipzig, Germany.,Department of Internal Medicine 2 (Nephrology, Rheumatology, Endocrinology), Martin-Luther-University Halle/Wittenberg, Halle, Germany
| |
Collapse
|
125
|
Kosiborod MN, Esterline R, Furtado RHM, Oscarsson J, Gasparyan SB, Koch GG, Martinez F, Mukhtar O, Verma S, Chopra V, Buenconsejo J, Langkilde AM, Ambery P, Tang F, Gosch K, Windsor SL, Akin EE, Soares RVP, Moia DDF, Aboudara M, Hoffmann Filho CR, Feitosa ADM, Fonseca A, Garla V, Gordon RA, Javaheri A, Jaeger CP, Leaes PE, Nassif M, Pursley M, Silveira FS, Barroso WKS, Lazcano Soto JR, Nigro Maia L, Berwanger O. Dapagliflozin in patients with cardiometabolic risk factors hospitalised with COVID-19 (DARE-19): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Diabetes Endocrinol 2021; 9:586-594. [PMID: 34302745 PMCID: PMC8294807 DOI: 10.1016/s2213-8587(21)00180-7] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 06/27/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND COVID-19 can lead to multiorgan failure. Dapagliflozin, a SGLT2 inhibitor, has significant protective benefits for the heart and kidney. We aimed to see whether this agent might provide organ protection in patients with COVID-19 by affecting processes dysregulated during acute illness. METHODS DARE-19 was a randomised, double-blind, placebo-controlled trial of patients hospitalised with COVID-19 and with at least one cardiometabolic risk factor (ie, hypertension, type 2 diabetes, atherosclerotic cardiovascular disease, heart failure, and chronic kidney disease). Patients critically ill at screening were excluded. Patients were randomly assigned 1:1 to dapagliflozin (10 mg daily orally) or matched placebo for 30 days. Dual primary outcomes were assessed in the intention-to-treat population: the outcome of prevention (time to new or worsened organ dysfunction or death), and the hierarchial composite outcome of recovery (change in clinical status by day 30). Safety outcomes, in patients who received at least one study medication dose, included serious adverse events, adverse events leading to discontinuation, and adverse events of interest. This study is registered with ClinicalTrials.gov, NCT04350593. FINDINGS Between April 22, 2020 and Jan 1, 2021, 1250 patients were randomly assigned with 625 in each group. The primary composite outcome of prevention showed organ dysfunction or death occurred in 70 patients (11·2%) in the dapagliflozin group, and 86 (13·8%) in the placebo group (hazard ratio [HR] 0·80, 95% CI 0·58-1·10; p=0·17). For the primary outcome of recovery, 547 patients (87·5%) in the dapagliflozin group and 532 (85·1%) in the placebo group showed clinical status improvement, although this was not statistically significant (win ratio 1·09, 95% CI 0·97-1·22; p=0·14). There were 41 deaths (6·6%) in the dapagliflozin group, and 54 (8·6%) in the placebo group (HR 0·77, 95% CI 0·52-1·16). Serious adverse events were reported in 65 (10·6%) of 613 patients treated with dapagliflozin and in 82 (13·3%) of 616 patients given the placebo. INTERPRETATION In patients with cardiometabolic risk factors who were hospitalised with COVID-19, treatment with dapagliflozin did not result in a statistically significant risk reduction in organ dysfunction or death, or improvement in clinical recovery, but was well tolerated. FUNDING AstraZeneca.
Collapse
Affiliation(s)
- Mikhail N Kosiborod
- Saint Luke's Mid America Heart Institute, Kansas City, MO, USA; School of Medicine, University of Missouri-Kansas City, Kansas City, MO, USA; The George Institute for Global Health, University of New South Wales, Sydney, NSW, Australia.
| | - Russell Esterline
- Late-stage Development, CVRM, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Remo H M Furtado
- Academic Research Organization-Hospital Israelita Albert Einstein, Sao Paulo, Brazil; Instituto do Coracao do Hospital das Clinicas da FMUSP, Sao Paulo, Brazil
| | - Jan Oscarsson
- Late-stage Development, CVRM, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Samvel B Gasparyan
- Late-stage Development, CVRM, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Gary G Koch
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Omar Mukhtar
- Experimental Medicine and Immunotherapeutics Division, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Subodh Verma
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute of St Michael's Hospital, Toronto, ON, Canada; Department of Surgery and Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | | | - Joan Buenconsejo
- Late-stage Development, CVRM, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Anna Maria Langkilde
- Late-stage Development, CVRM, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Philip Ambery
- Late-stage Development, CVRM, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Fengming Tang
- Saint Luke's Mid America Heart Institute, Kansas City, MO, USA
| | - Kensey Gosch
- Saint Luke's Mid America Heart Institute, Kansas City, MO, USA
| | | | | | - Ronaldo V P Soares
- Academic Research Organization-Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | - Diogo D F Moia
- Academic Research Organization-Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | - Matthew Aboudara
- Division of Pulmonary and Critical Care, Saint Luke's Health System, Kansas City, MO, USA
| | | | | | | | - Vishnu Garla
- Department of Endocrinology, Diabetes and Metabolism, Internal Medicine, University of Mississippi Medical Center, Jackson, MS, USA; Mississippi Center for Clinical and Translational Research, Jackson, MI, USA
| | | | - Ali Javaheri
- Washington University School of Medicine, St Louis, MO, USA
| | | | - Paulo E Leaes
- Irmandade Da Santa Casa de Misericórdia de Porto Alegre, Brazil
| | - Michael Nassif
- Saint Luke's Mid America Heart Institute, Kansas City, MO, USA
| | | | | | - Weimar Kunz Sebba Barroso
- Liga de Hipertensão Arterial -Universidade Federal de Goiás, Brazil; HCAMP-Secretaria Estadual de Saúde, Goiás, Brazil
| | | | - Lilia Nigro Maia
- Centro Integrado de Pesquisas, Hospital de Base, São José do Rio Preto, Brazil
| | - Otavio Berwanger
- Academic Research Organization-Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| |
Collapse
|
126
|
Durante W, Behnammanesh G, Peyton KJ. Effects of Sodium-Glucose Co-Transporter 2 Inhibitors on Vascular Cell Function and Arterial Remodeling. Int J Mol Sci 2021; 22:ijms22168786. [PMID: 34445519 PMCID: PMC8396183 DOI: 10.3390/ijms22168786] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular disease is the leading cause of morbidity and mortality in diabetes. Recent clinical studies indicate that sodium-glucose co-transporter 2 (SGLT2) inhibitors improve cardiovascular outcomes in patients with diabetes. The mechanism underlying the beneficial effect of SGLT2 inhibitors is not completely clear but may involve direct actions on vascular cells. SGLT2 inhibitors increase the bioavailability of endothelium-derived nitric oxide and thereby restore endothelium-dependent vasodilation in diabetes. In addition, SGLT2 inhibitors favorably regulate the proliferation, migration, differentiation, survival, and senescence of endothelial cells (ECs). Moreover, they exert potent antioxidant and anti-inflammatory effects in ECs. SGLT2 inhibitors also inhibit the contraction of vascular smooth muscle cells and block the proliferation and migration of these cells. Furthermore, studies demonstrate that SGLT2 inhibitors prevent postangioplasty restenosis, maladaptive remodeling of the vasculature in pulmonary arterial hypertension, the formation of abdominal aortic aneurysms, and the acceleration of arterial stiffness in diabetes. However, the role of SGLT2 in mediating the vascular actions of these drugs remains to be established as important off-target effects of SGLT2 inhibitors have been identified. Future studies distinguishing drug- versus class-specific effects may optimize the selection of specific SGLT2 inhibitors in patients with distinct cardiovascular pathologies.
Collapse
|
127
|
Ilyas F, Jones L, Tee SL, Horsfall M, Swan A, Wollaston F, Hecker T, De Pasquale C, Thomas S, Chong W, Stranks S, Mangoni AA, Selvanayagam JB, Chew DP, De Pasquale CG. Acute pleiotropic effects of dapagliflozin in type 2 diabetic patients with heart failure with reduced ejection fraction: a crossover trial. ESC Heart Fail 2021; 8:4346-4352. [PMID: 34382353 PMCID: PMC8497349 DOI: 10.1002/ehf2.13553] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/21/2021] [Accepted: 08/02/2021] [Indexed: 11/09/2022] Open
Abstract
Aims This study aimed to explore the rapid effects of dapagliflozin in heart failure with reduced ejection fraction (HFrEF). Methods and results We studied the functional, echocardiographic, electrophysiological, lung ultrasound, ambulatory blood pressure (BP), microvascular and macrovascular function, and biochemical effects of 2 week treatment with dapagliflozin in 19 type 2 diabetic HFrEF patients in a double‐blind, crossover, placebo‐controlled trial. Dapagliflozin had no significant effect on clinical, functional, or quality of life parameters. Dapagliflozin reduced systolic BP [114 (105, 131) vs. 106 (98, 113) mmHg, P < 0.01] and diastolic BP [71 (61, 78) vs. 62 (55, 70) mmHg, P < 0.01]. There was no effect on cardiac chamber size, ventricular systolic function, lung ultrasound, or arterial wave reflection. Dapagliflozin increased creatinine [117 (92, 129) vs. 122 (107, 135) μmol/L, P < 0.05] and haemoglobin [135 (118, 138) vs. 136 (123, 144) g/L, P < 0.05]. There was a reduction in ventricular ectopy [1.4 (0.1, 2.9) vs. 0.2 (0.1, 1.4) %, P < 0.05] and an increase in standard deviation of normal heart beat intervals [70 (58, 90) vs. 74 (62, 103), P < 0.05]. Unexpectedly, dapagliflozin increased high‐sensitivity troponin T [25 (19, 37) vs. 28 (20, 42) ng/L, P < 0.01] and reduced reactive hyperaemia index [1.29 (1.21, 1.56) vs. 1.40 (1.23, 1.84), P < 0.05]. Conclusions After 2 weeks, while multiple parameters supported BP reduction and haemoconcentration with dapagliflozin, reduction in cardiac filling pressure, lung water, and functional improvement was not shown. Reduced ventricular ectopic burden suggests an early antiarrhythmic benefit. The small increase in troponin T and the reduction in the reactive hyperaemia index warrant further mechanistic exploration in this treatment of proven mortality benefit in HFrEF.
Collapse
Affiliation(s)
- Fahmida Ilyas
- Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Lynette Jones
- Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Su Ling Tee
- Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Matthew Horsfall
- Flinders Medical Centre, Bedford Park, South Australia, Australia.,Flinders University, Bedford Park, South Australia, Australia
| | - Amy Swan
- Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Fiona Wollaston
- Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Tracy Hecker
- Flinders Medical Centre, Bedford Park, South Australia, Australia
| | | | - Simeoni Thomas
- Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - William Chong
- Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Steve Stranks
- Flinders Medical Centre, Bedford Park, South Australia, Australia.,Flinders University, Bedford Park, South Australia, Australia
| | - Arduino A Mangoni
- Flinders Medical Centre, Bedford Park, South Australia, Australia.,Flinders University, Bedford Park, South Australia, Australia
| | - Joseph B Selvanayagam
- Flinders Medical Centre, Bedford Park, South Australia, Australia.,Flinders University, Bedford Park, South Australia, Australia
| | - Derek P Chew
- Flinders Medical Centre, Bedford Park, South Australia, Australia.,Flinders University, Bedford Park, South Australia, Australia
| | - Carmine G De Pasquale
- Flinders Medical Centre, Bedford Park, South Australia, Australia.,Flinders University, Bedford Park, South Australia, Australia
| |
Collapse
|
128
|
Abstract
Alterations of endothelial function, inflammatory activation, and nitric oxide-cyclic guanosine monophosphate (NO-cGMP) pathway are involved in the pathophysiology of heart failure. Metabolic alterations have been studied in the myocardium of heart failure (HF) patients; alterations in ketone body and amino acid/protein metabolism have been described in patients affected by HF, as well as mitochondrial dysfunction and other modified metabolic signaling. However, their possible contributions toward cardiac function impairment in HF patients are not completely known. Recently, sodium-glucose co-transporter 2 inhibitors (SGLT2i) and glucagon-like peptide-1 (GLP-1) receptor agonists (RAs) have emerged as a new class of drugs designed to treat patients with type 2 diabetes (T2D), but have also been shown to be protective against HF-related events and CV mortality. To date, the protective cardiovascular effects of these drugs in patients with and without T2D are not completely understood and several mechanisms have been proposed. In this review, we discuss on vascular and metabolic effects of SGLT2i and GLP-1 in HF patients.
Collapse
|
129
|
Gitto M, Vrachatis DA, Condorelli G, Papathanasiou K, Reimers B, Deftereos S, Stefanini GG. Potential Therapeutic Benefits of Sodium-Glucose Cotransporter 2 Inhibitors in the Context of Ischemic Heart Failure: A State-Of-The-Art Review. Cardiovasc Hematol Agents Med Chem 2021; 20:90-102. [PMID: 34370645 DOI: 10.2174/1871525719666210809121016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/25/2021] [Accepted: 07/15/2021] [Indexed: 11/22/2022]
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors are a class of anti-diabetic agents that block the reabsorption of glucose in the proximal convoluted tubule of the nephron, thereby contributing to glycosuria and lowering blood glucose levels. SGLT2 inhibitors have been associated with improved cardiovascular outcomes in patients with diabetes, including a reduced risk of cardiovascular death and hospitalizations for heart failure. Recently, DAPA-HF and EMPEROR REDUCED trials showed the beneficial cardiovascular effect of SGLT2 inhibitors in patients with heart failure with consistently reduced ejection fraction (HFrEF) regardless of the presence of diabetes. Moreover, some exploratory studies suggested that these drugs improve Left Ventricular (LV) systolic function and oppose LV adverse remodeling in patients with HFrEF. However, the exact mechanisms that mediated for this benefit are not fully understood. Beyond glycemic control, enhanced natriuresis, increased erythropoiesis, improved endothelial function, changes in myocardial metabolism, anti-inflammatory and anti-oxidative properties may all play an active role in SGLT2 inhibitors' cardiovascular benefits. A deep understanding of the pathophysiological interplay is key to define which HF phenotype could benefit more from SGLT2 inhibitors. Current clinical evidence on the comparison of different HF etiologies is limited to posthoc subgroup analysis of DAPA-HF and EMPEROR-REDUCED, which showed similar outcomes in patients with or without ischemic HF. On the other hand, in earlier studies of patients suffering from diabetes, rates of classic ischemic endpoints, such as myocardial infarction, stroke or coronary revascularization, did not differ between patients treated with SGLT2 inhibitors or placebo. The aim of this review is to discuss whether SGLT2 inhibitors may improve prognosis in patients with ischemic HF, not only in terms of reducing re-hospitalizations and improving left ventricular function but also by limiting coronary artery disease progression and ischemic burden.
Collapse
Affiliation(s)
- Mauro Gitto
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele-Milan, Italy
| | - Dimitrios A Vrachatis
- 2nd Department of Cardiology, National and Kapodistrian University of Athens, Athens, Greece
| | - Gianluigi Condorelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele-Milan, Italy
| | | | - Bernhard Reimers
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele-Milan, Italy
| | - Spyridon Deftereos
- 2nd Department of Cardiology, National and Kapodistrian University of Athens, Athens, Greece
| | - Giulio G Stefanini
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele-Milan, Italy
| |
Collapse
|
130
|
Liu L, Ni YQ, Zhan JK, Liu YS. The Role of SGLT2 Inhibitors in Vascular Aging. Aging Dis 2021; 12:1323-1336. [PMID: 34341711 PMCID: PMC8279525 DOI: 10.14336/ad.2020.1229] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 12/29/2020] [Indexed: 12/19/2022] Open
Abstract
Vascular aging is defined as organic and functional changes in blood vessels, in which decline in autophagy levels, DNA damage, MicroRNA (miRNA), oxidative stress, sirtuin, and apoptosis signal-regulated kinase 1 (ASK1) are integral thereto. With regard to vascular morphology, the increase in arterial stiffness, atherosclerosis, vascular calcification and high amyloid beta levels are closely related to vascular aging. Further closely related thereto, at the cellular level, is the aging of vascular endothelial cells (ECs) and vascular smooth muscle cells (VSMCs). Vascular aging seriously affects the health, economy and life of patients, but can be delayed by SGLT2 inhibitors through the improvement of vascular function. In the present article, a review is conducted of recent domestic and international progress in research on SGLT2 inhibitors,vascular aging and diseases related thereto, thereby providing theoretical support and guidance for further revealing the relationship between SGLT2 inhibitors and diseases related to vascular aging.
Collapse
Affiliation(s)
- Le Liu
- 1Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,2Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan 410011, China
| | - Yu-Qing Ni
- 1Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,2Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan 410011, China
| | - Jun-Kun Zhan
- 1Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,2Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan 410011, China
| | - You-Shuo Liu
- 1Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,2Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
131
|
Hoong CWS, Chua MWJ. SGLT2 Inhibitors as Calorie Restriction Mimetics: Insights on Longevity Pathways and Age-Related Diseases. Endocrinology 2021; 162:6226811. [PMID: 33857309 DOI: 10.1210/endocr/bqab079] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Indexed: 02/08/2023]
Abstract
Sodium-glucose cotransporter-2 (SGLT2) inhibitors induce glycosuria, reduce insulin levels, and promote fatty acid oxidation and ketogenesis. By promoting a nutrient deprivation state, SGLT2 inhibitors upregulate the energy deprivation sensors AMPK and SIRT1, inhibit the nutrient sensors mTOR and insulin/IGF1, and modulate the closely linked hypoxia-inducible factor (HIF)-2α/HIF-1α pathways. Phosphorylation of AMPK and upregulation of adiponectin and PPAR-α favor a reversal of the metabolic syndrome which have been linked to suppression of chronic inflammation. Downregulation of insulin/IGF1 pathways and mTOR signaling from a reduction in glucose and circulating amino acids promote cellular repair mechanisms, including autophagy and proteostasis which confer cellular stress resistance and attenuate cellular senescence. SIRT1, another energy sensor activated by NAD+ in nutrient-deficient states, is reciprocally activated by AMPK, and can deacetylate and activate transcription factors, such as PCG-1α, mitochondrial transcription factor A (TFAM), and nuclear factor E2-related factor (NRF)-2, that regulate mitochondrial biogenesis. FOXO3 transcription factor which target genes in stress resistance, is also activated by AMPK and SIRT1. Modulation of these pathways by SGLT2 inhibitors have been shown to alleviate metabolic diseases, attenuate vascular inflammation and arterial stiffness, improve mitochondrial function and reduce oxidative stress-induced tissue damage. Compared with other calorie restriction mimetics such as metformin, rapamycin, resveratrol, and NAD+ precursors, SGLT2 inhibitors appear to be the most promising in the treatment of aging-related diseases, due to their regulation of multiple longevity pathways that closely resembles that achieved by calorie restriction and their established efficacy in reducing cardiovascular events and all-cause mortality. Evidence is compelling for the role of SGLT2 inhibitors as a calorie restriction mimetic in anti-aging therapeutics.
Collapse
Affiliation(s)
- Caroline W S Hoong
- Division of Endocrinology, Department of General Medicine, Woodlands Health Campus, National Healthcare Group Singapore, Woodlands Health Campus Singapore, 768024, Singapore
| | - Marvin W J Chua
- Endocrinology Service, Department of General Medicine, Sengkang General Hospital, SingHealth Group Singapore, Sengkang General Hospital Singapore, 544886, Singapore
| |
Collapse
|
132
|
Tsai KF, Chen YL, Chiou TTY, Chu TH, Li LC, Ng HY, Lee WC, Lee CT. Emergence of SGLT2 Inhibitors as Powerful Antioxidants in Human Diseases. Antioxidants (Basel) 2021; 10:1166. [PMID: 34439414 PMCID: PMC8388972 DOI: 10.3390/antiox10081166] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/10/2021] [Accepted: 07/19/2021] [Indexed: 12/14/2022] Open
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors are a new class of oral glucose-lowering agents. Apart from their glucose-lowering effects, large clinical trials assessing certain SGLT2 inhibitors have revealed cardiac and renal protective effects in non-diabetic patients. These excellent outcomes motivated scientists and clinical professionals to revisit their underlying mechanisms. In addition to the heart and kidney, redox homeostasis is crucial in several human diseases, including liver diseases, neural disorders, and cancers, with accumulating preclinical studies demonstrating the therapeutic benefits of SGLT2 inhibitors. In the present review, we aimed to update recent advances in the antioxidant roles of SGLT2 inhibitors in common but debilitating human diseases. We anticipate that this review will guide new research directions and novel therapeutic strategies for diabetes, cardiovascular diseases, nephropathies, liver diseases, neural disorders, and cancers in the era of SGLT2 inhibitors.
Collapse
Affiliation(s)
- Kai-Fan Tsai
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (K.-F.T.); (T.T.-Y.C.); (L.-C.L.); (H.-Y.N.)
| | - Yung-Lung Chen
- Section of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Terry Ting-Yu Chiou
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (K.-F.T.); (T.T.-Y.C.); (L.-C.L.); (H.-Y.N.)
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Tian-Huei Chu
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
- Biobank and Tissue Bank, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Lung-Chih Li
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (K.-F.T.); (T.T.-Y.C.); (L.-C.L.); (H.-Y.N.)
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Hwee-Yeong Ng
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (K.-F.T.); (T.T.-Y.C.); (L.-C.L.); (H.-Y.N.)
| | - Wen-Chin Lee
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (K.-F.T.); (T.T.-Y.C.); (L.-C.L.); (H.-Y.N.)
| | - Chien-Te Lee
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (K.-F.T.); (T.T.-Y.C.); (L.-C.L.); (H.-Y.N.)
| |
Collapse
|
133
|
The Potential Roles of Osmotic and Nonosmotic Sodium Handling in Mediating the Effects of Sodium-Glucose Cotransporter 2 Inhibitors on Heart Failure. J Card Fail 2021; 27:1447-1455. [PMID: 34289398 PMCID: PMC8759453 DOI: 10.1016/j.cardfail.2021.07.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 06/08/2021] [Accepted: 07/02/2021] [Indexed: 11/24/2022]
Abstract
Concomitant type 2 diabetes and chronic kidney disease increases the risk of heart failure. Recent studies demonstrate beneficial effects of sodium-glucose cotransporter 2 (SGLT2) inhibitors on chronic kidney disease progression and heart failure hospitalization in patients with and without diabetes. In addition to inhibiting glucose reabsorption, SGLT2 inhibitors decrease proximal tubular sodium reabsorption, possibly leading to transient natriuresis. We review the hypothesis that SGLT2 inhibitor’s natriuretic and osmotic diuretic effects mediate their cardioprotective effects. The degree to which these benefits are related to changes in sodium, independent of the kidney, is currently unknown. Aside from effects on osmotically active sodium, we explore the intriguing possibility that SGLT2 inhibitors could also modulate nonosmotic sodium storage. This alternative hypothesis is based on emerging literature that challenges the traditional 2-compartment model of sodium balance to provide support for a 3-compartment model that includes the binding of sodium to glycosaminoglycans, such as those in muscles and skin. This recent research on nonosmotic sodium storage, as well as direct cardiac effects of SGLT2 inhibitors, provides possibilities for other ways in which SGLT2 inhibitors might mitigate heart failure risk. Overall, we review the effects of SGLT2 inhibitors on sodium balance and sensitivity, cardiac tissue, interstitial fluid and plasma volume, and nonosmotic sodium storage. SGLT2 inhibitors have cardiovascular benefits that include HF outcomes in patients with and without diabetes. Because the underlying mechanisms are only partly explained by improvements in BP, body weight, or glucose control, other mechanisms have been proposed. We focus here on a central role for effects on sodium as underlying the positive benefits of SGLT2 inhibitors in HF. We explore the new (although still unconfirmed) idea that SGLT2 inhibitors exert some of their positive effects by affecting nonosmotic sodium (ie, sodium bound to muscles and skin and not dissolved in the blood). SGLT2 inhibitors have emerged as a class of drugs, previously prescribed for patients with T2D, that have in more recent years been shown to have substantial heart and kidney clinical benefits in patients with and without T2D. The degree to which these benefits are related to kidney-independent changes in sodium homeostasis is currently unknown. A better understanding of the nonosmotic mechanisms underpinning the benefits of SGLT2 inhibition on HF (with reduced or preserved left ventricular ejection fraction) may allow researchers to assess the effects of SGLT2 inhibitors in combination with other treatments that affect sodium balance.
Collapse
|
134
|
Xiao L, Nie X, Cheng Y, Wang N. Sodium-Glucose Cotransporter-2 Inhibitors in Vascular Biology: Cellular and Molecular Mechanisms. Cardiovasc Drugs Ther 2021; 35:1253-1267. [PMID: 34273091 DOI: 10.1007/s10557-021-07216-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/07/2021] [Indexed: 12/16/2022]
Abstract
Sodium-glucose cotransporter-2 (SGLT2) inhibitors are new antidiabetic drugs that reduce hyperglycemia by inhibiting the glucose reabsorption in renal proximal tubules. Clinical studies have shown that SGLT2 inhibitors not only improve glycemic control but also reduce major adverse cardiovascular events (MACE, cardiovascular and total mortality, fatal or nonfatal myocardial infarction or stroke) and hospitalization for heart failure (HF), and improve outcome in chronic kidney disease. These cardiovascular and renal benefits have now been confirmed in both diabetes and non-diabetes patients. The precise mechanism(s) responsible for the protective effects are under intensive investigation. This review examines current evidence on the cardiovascular benefits of SGLT2 inhibitors, with a special emphasis on the vascular actions and their potential mechanisms.
Collapse
Affiliation(s)
- Lei Xiao
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xin Nie
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Yanyan Cheng
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Nanping Wang
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Institute of Cardiovascular Science, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
135
|
Trujillo H, Caravaca-Fontán F, Caro J, Morales E, Praga M. The Forgotten Antiproteinuric Properties of Diuretics. Am J Nephrol 2021; 52:435-449. [PMID: 34233330 DOI: 10.1159/000517020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/30/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Although diuretics are one of the most widely used drugs by nephrologists, their antiproteinuric properties are not generally taken into consideration. SUMMARY Thiazide diuretics have been shown to reduce proteinuria by >35% in several prospective controlled studies, and these values are markedly increased when combined with a low-salt diet. Thiazide-like diuretics (indapamide and chlorthalidone) have shown similar effectiveness. The antiproteinuric effect of mineralocorticoid receptor antagonists (spironolactone, eplerenone, and finerenone) has been clearly established through prospective and controlled studies, and treatment with finerenone reduces the risk of chronic kidney disease progression in type-2 diabetic patients. The efficacy of other diuretics such as amiloride, triamterene, acetazolamide, or loop diuretics has been less explored, but different investigations suggest that they might share the same antiproteinuric properties of other diuretics that should be evaluated through controlled studies. Although the inclusion of sodium-glucose cotransporter-2 inhibitors (SGLT2i) among diuretics is a controversial issue, their renoprotective and cardioprotective properties, confirmed in various landmark trials, constitute a true revolution in the treatment of patients with kidney disease. Recent subanalyses of these trials have shown that the early antiproteinuric effect induced by SGLT2i predicts long-term preservation of kidney function. Key Message: Whether the early reduction in proteinuria induced by diuretics other than finerenone and SGLT2i, as summarized in this review, also translates into long-term renoprotection requires further prospective and observational studies. In any case, it is important for the clinician to be aware of the antiproteinuric properties of drugs so often used in daily clinical practice.
Collapse
Affiliation(s)
- Hernando Trujillo
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain,
| | | | - Jara Caro
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
| | - Enrique Morales
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
- Department of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Manuel Praga
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
- Department of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
136
|
Wu VCC, Li YR, Wang CY. Impact of Sodium-Glucose Co-Transporter 2 Inhibitors on Cardiac Protection. Int J Mol Sci 2021; 22:ijms22137170. [PMID: 34281221 PMCID: PMC8268177 DOI: 10.3390/ijms22137170] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/12/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023] Open
Abstract
Sodium–glucose co-transporter 2 (SGLT2) inhibitors have been approved as a new class of anti-diabetic drugs for type 2 diabetes mellitus (T2DM). The SGLT2 inhibitors reduce glucose reabsorption through renal systems, thus improving glycemic control in all stages of diabetes mellitus, independent of insulin. This class of drugs has the advantages of no clinically relevant hypoglycemia and working in synergy when combined with currently available anti-diabetic drugs. While improving sugar level control in these patients, SGLT2 inhibitors also have the advantages of blood-pressure improvement and bodyweight reduction, with potential cardiac and renal protection. In randomized control trials for patients with diabetes, SGLT2 inhibitors not only improved cardiovascular and renal outcomes, but also hospitalization for heart failure, with this effect extending to those without diabetes mellitus. Recently, dynamic communication between autophagy and the innate immune system with Beclin 1-TLR9-SIRT3 complexes in response to SGLT2 inhibitors that may serve as a potential treatment strategy for heart failure was discovered. In this review, the background molecular pathways leading to the clinical benefits are examined in this new class of anti-diabetic drugs, the SGLT2 inhibitors.
Collapse
Affiliation(s)
- Victor Chien-Chia Wu
- Division of Cardiology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City 33305, Taiwan;
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan;
| | - Yan-Rong Li
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan;
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan City 33305, Taiwan
| | - Chao-Yung Wang
- Division of Cardiology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City 33305, Taiwan;
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan;
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 35053, Taiwan
- Department of Medical Science, National Tsing Hua University, Hsinchu 30013, Taiwan
- Correspondence:
| |
Collapse
|
137
|
Lamacchia O, Sorrentino MR. Diabetes Mellitus, Arterial Stiffness and Cardiovascular Disease: Clinical Implications and the Influence of SGLT2i. Curr Vasc Pharmacol 2021; 19:233-240. [PMID: 32183678 DOI: 10.2174/1570161118666200317150359] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 12/20/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a rapidly evolving global health issue associated with a markedly increased risk of cardiovascular (CV) morbidity and mortality. The hyperglycaemic milieu contributes to the development of CV complications via several pathological pathways, leading to increased arterial stiffness (AS), that can be considered as a predictor of CV events in patients with diabetes. The measurement of AS is increasingly used for the clinical assessment of patients. Several methodologies were used in extensive population studies to assess AS; the most commonly used is the pulse wave velocity (PWV). The cardio-ankle vascular index (CAVI) was developed to measure AS; it is not affected by blood pressure at the time of measurement and shows stable values in healthy persons for years. There are several potential pharmacological and non-pharmacological interventions aiming to reduce AS. Recent evidence from clinical trials suggests that newer antidiabetic drugs do not only exert glycaemic-lowering properties but also decrease CV risk. In this context, sodium glucose cotransporter- 2 inhibitors (SGLT2i) ( empagliflozin, canagliflozin and dapagliflozin) significantly reduced the risk of CV and all-cause mortality (only EMPA-REG OUTCOME study) and hospitalization for heart failure in patients with T2DM with established CV disease and/or with CV risk factors. Improved endothelial function and AS probably represents one of the mechanisms by which these drugs exert their beneficial effects. The present review aimed both to describe the association between AS and T2DM and to discuss the effectiveness of SGLT2i on vascular endothelial dysfunction and AS.
Collapse
Affiliation(s)
- Olga Lamacchia
- Unit of Endocrinology, Department of Medical and Surgical Sciences, University of Foggia, via Luigi Pinto, 1, 71122 Foggia, Italy
| | - Maria Rosaria Sorrentino
- Unit of Endocrinology, Department of Medical and Surgical Sciences, University of Foggia, via Luigi Pinto, 1, 71122 Foggia, Italy
| |
Collapse
|
138
|
Cignarelli A, Genchi VA, D’Oria R, Giordano F, Caruso I, Perrini S, Natalicchio A, Laviola L, Giorgino F. Role of Glucose-Lowering Medications in Erectile Dysfunction. J Clin Med 2021; 10:jcm10112501. [PMID: 34198786 PMCID: PMC8201035 DOI: 10.3390/jcm10112501] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/31/2021] [Accepted: 06/03/2021] [Indexed: 01/11/2023] Open
Abstract
Erectile dysfunction (ED) is a long-term complication of type 2 diabetes (T2D) widely known to affect the quality of life. Several aspects of altered metabolism in individuals with T2D may help to compromise the penile vasculature structure and functions, thus exacerbating the imbalance between smooth muscle contractility and relaxation. Among these, advanced glycation end-products and reactive oxygen species derived from a hyperglycaemic state are known to accelerate endothelial dysfunction by lowering nitric oxide bioavailability, the essential stimulus of relaxation. Although several studies have explained the pathogenetic mechanisms involved in the generation of erectile failure, few studies to date have described the efficacy of glucose-lowering medications in the restoration of normal sexual activity. Herein, we will present current knowledge about the main starters of the pathophysiology of diabetic ED and explore the role of different anti-diabetes therapies in the potential remission of ED, highlighting specific pathways whose activation or inhibition could be fundamental for sexual care in a diabetes setting.
Collapse
|
139
|
Provenzano M, Pelle MC, Zaffina I, Tassone B, Pujia R, Ricchio M, Serra R, Sciacqua A, Michael A, Andreucci M, Arturi F. Sodium-Glucose Co-transporter-2 Inhibitors and Nephroprotection in Diabetic Patients: More Than a Challenge. Front Med (Lausanne) 2021; 8:654557. [PMID: 34150796 PMCID: PMC8212983 DOI: 10.3389/fmed.2021.654557] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 05/11/2021] [Indexed: 12/16/2022] Open
Abstract
Diabetic nephropathy is the most common cause of end-stage renal disease worldwide. Control of blood glucose and blood pressure (BP) reduces the risk of developing this complication, but once diabetic nephropathy is established, it is then only possible to slow its progression. Sodium-glucose cotransporter-2 inhibitors (SGLT2is) are a novel class of oral hypoglycemic agents that increase urinary glucose excretion by suppressing glucose reabsorption at the renal proximal tubule. SGLT2is lower glycated hemoglobin (HbA1c) without increasing the risk of hypoglycemia, induce weight loss and improve various metabolic parameters including BP, lipid profile, albuminuria and uric acid. Several clinical trials have shown that SGLT2is (empagliflozin, dapagliflozin canagliflozin, and ertugliflozin) improve cardiovascular and renal outcomes and mortality in patients with type 2 diabetes. Effects of SGLT2is on the kidney can be explained by multiple pathways. SGLT2is may improve renal oxygenation and intra-renal inflammation thereby slowing the progression of kidney function decline. Additionally, SGLT2is are associated with a reduction in glomerular hyperfiltration, an effect which is mediated by the increase in natriuresis, the re-activation of tubule-glomerular feedback and independent of glycemic control. In this review, we will focus on renal results of major cardiovascular and renal outcome trials and we will describe direct and indirect mechanisms through which SGLT2is confer renal protection.
Collapse
Affiliation(s)
- Michele Provenzano
- Chair of Nephrology, Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Maria Chiara Pelle
- Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Isabella Zaffina
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Bruno Tassone
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Roberta Pujia
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Marco Ricchio
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Raffaele Serra
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology at the Department of Surgical and Medical Sciences University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Angela Sciacqua
- Unit of Geriatric, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Ashour Michael
- Chair of Nephrology, Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Michele Andreucci
- Chair of Nephrology, Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Franco Arturi
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| |
Collapse
|
140
|
Roush GC, Messerli FH. Chlorthalidone versus hydrochlorothiazide: major cardiovascular events, blood pressure, left ventricular mass, and adverse effects. J Hypertens 2021; 39:1254-1260. [PMID: 33470735 DOI: 10.1097/hjh.0000000000002771] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND There is continuous debate whether chlorthalidone (CTD) and hydrochlorothiazide (HCTZ) differ in reducing major cardiovascular events (MACE). HCTZ is prescribed 10 times more commonly than CTD. METHOD A systematic literature search yielded 14 references, including two network meta-analyses of randomized trials with MACE and left ventricular mass as outcomes. RESULTS The network meta-analysis of randomized trials showed CTD reducing MACE more than HCTZ, hazard ratio = 0.79 (0.72-0.88), P < 0.0001, and an observational cohort study gave an identical point estimate: hazard ratio = 0.79 (0.68-0.92), P = 0.002. In contrast, two observational cohort studies reported no differences between CTD and HCTZ. However, in the studies showing the superiority of CTD median follow-up was 4.3 and 7.0 years, respectively, whereas in the latter studies showing no difference between the two drugs follow-up was only 0.95 and 0.25 years. As differences in outcomes for MACE in hypertension trials with various interventions only emerge after prolonged (>1 year) therapy, differences in follow-up explain these discrepant results. CTD also more effectively reduced left ventricular mass in observational data and network analysis of trials. These advantages of CTD over HCTZ are consistent with greater reductions in night-time blood pressure, greater reductions in oxidative stress and platelet aggregation, and greater improvements in endothelial function. CONCLUSION Over the short-term there were no differences in the net clinical benefit between HCTZ and CTD. However, long-term available data document CTD to be significantly more effective in reducing MACE than HCTZ. The Veterans Administration's trial in progress may provide definitive answer to these questions.
Collapse
Affiliation(s)
| | - Franz H Messerli
- Swiss Cardiovascular Center, Bern, Switzerland
- Mount Sinai Icahn School of Medicine, New York City, New York, USA
- Jagiellonian University Krakow, Poland
| |
Collapse
|
141
|
Impact of SGLT2 Inhibitors on Heart Failure: From Pathophysiology to Clinical Effects. Int J Mol Sci 2021; 22:ijms22115863. [PMID: 34070765 PMCID: PMC8199383 DOI: 10.3390/ijms22115863] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) affects up to over 20% of patients with type 2 diabetes (T2DM), even more in the elderly. Although, in T2DM, both hyperglycemia and the proinflammatory status induced by insulin resistance are crucial in cardiac function impairment, SGLT2i cardioprotective mechanisms against HF are several. In particular, these beneficial effects seem attributable to the significant reduction of intracellular sodium levels, well-known to exert a cardioprotective role in the prevention of oxidative stress and consequent cardiomyocyte death. From a molecular perspective, patients’ exposure to gliflozins’ treatment mimics nutrient and oxygen deprivation, with consequent autophagy stimulation. This allows to maintain the cellular homeostasis through different degradative pathways. Thus, since their introduction in the clinical practice, the hypotheses on SGLT2i mechanisms of action have changed: from simple glycosuric drugs, with consequent glucose lowering, erythropoiesis enhancing and ketogenesis stimulating, to intracellular sodium-lowering molecules. This provides their consequent cardioprotective effect, which justifies its significant reduction in CV events, especially in populations at higher risk. Finally, the updated clinical evidence of SGLT2i benefits on HF was summarized. Thus, this review aimed to analyze the cardioprotective mechanisms of sodium glucose transporter 2 inhibitors (SGLT2i) in patients with HF, as well as their clinical impact on cardiovascular events.
Collapse
|
142
|
Thirunavukarasu S, Brown LAE, Chowdhary A, Jex N, Swoboda P, Greenwood JP, Plein S, Levelt E. Rationale and design of the randomised controlled cross-over trial: Cardiovascular effects of empaglifozin in diabetes mellitus. Diab Vasc Dis Res 2021; 18:14791641211021585. [PMID: 34182806 PMCID: PMC8481726 DOI: 10.1177/14791641211021585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) is associated with an increased risk of cardiovascular (CV) disease. In patients with T2D and established CV disease, selective inhibitors of sodium-glucose cotransporter 2 (SGLT2) have been shown to decrease CV and all-cause mortality, and heart failure (HF) admissions. Utilising CV magnetic resonance imaging (CMR) and continuous glucose monitoring (CGM) by FreeStyle Libre Pro Sensor, we aim to explore the mechanisms of action which give Empagliflozin, an SGLT2 inhibitor, its beneficial CV effects and compare these to the effects of dipeptidyl peptidase-4 inhibitor Sitagliptin. METHODS This is a single centre, open-label, cross-over trial conducted at the Leeds Teaching Hospitals NHS Trust. Participants are randomised for the order of treatment and receive 3 months therapy with Empagliflozin, and 3 months therapy with Sitagliptin sequentially. Twenty-eight eligible T2D patients with established ischaemic heart disease will be recruited. Patients undergo serial CMR scans on three visits. DISCUSSION The primary outcome measure is the myocardial perfusion reserve in remote myocardium. We hypothesise that Empaglifozin treatment is associated with improvements in myocardial blood flow and reductions in myocardial interstitial fibrosis, independent of CGM measured glycemic control in patients with T2D and established CV disease. TRIAL REGISTRATION This study has full research ethics committee approval (REC: 18/YH/0190) and data collection is anticipated to finish in December 2021. This study was retrospectively registered at https://doi.org/10.1186/ISRCTN82391603 and monitored by the University of Leeds. The study results will be submitted for publication within 6 months of completion.
Collapse
Affiliation(s)
- Sharmaine Thirunavukarasu
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Louise AE Brown
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Amrit Chowdhary
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Nicholas Jex
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Peter Swoboda
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - John P Greenwood
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Sven Plein
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Eylem Levelt
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| |
Collapse
|
143
|
Ferrannini G, Savarese G, Rydén L. Sodium-glucose transporter inhibition in heart failure: from an unexpected side effect to a novel treatment possibility. Diabetes Res Clin Pract 2021; 175:108796. [PMID: 33845051 DOI: 10.1016/j.diabres.2021.108796] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 03/30/2021] [Accepted: 04/02/2021] [Indexed: 12/17/2022]
Abstract
Sodium-glucose transporter-2 inhibitors (SGLT2i), originally launched as glucose-lowering drugs, have been studied in large cardiovascular outcome trials to ascertain safety. Surprisingly, these compounds reduced the risk of cardiovascular events (cardiovascular death, non-fatal myocardial and non-fatal stroke) and total mortality. The mechanisms behind this benefit are only partly understood, but a major contributor is the reduction of heart failure hospitalisations, evident already within weeks after the initiation of the SGLT2i. SGLT2 inhibition increases urinary glucose excretion, thereby improving glycaemic control in an insulin-independent manner. Moreover, SGLT2i potentially impact the cardiovascular system both indirectly via weight loss and blood pressure lowering and directly through osmotic diuresis and increased sodium excretion and presumably by improving myocardial energetics. The aim of this review is to summarise evidence from all major outcome trials investigating SGLT2i in patients with diabetes, as well as recent evidence from trials in heart failure patients without glucose perturbations, which pave the way for novel treatment of large groups of patients. The results of these studies have been taken into account in recently issued guidelines for the management of diabetes and cardiovascular disease. An important task for diabetologists, cardiologists and general practitioners is to incorporate them into clinical practice to the benefit of many patients.
Collapse
Affiliation(s)
- Giulia Ferrannini
- Department of Medicine K2, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Gianluigi Savarese
- Department of Medicine K2, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Lars Rydén
- Department of Medicine K2, Karolinska Institutet, 171 76 Stockholm, Sweden.
| |
Collapse
|
144
|
Endothelin antagonism and sodium glucose Co-transporter 2 inhibition. A potential combination therapeutic strategy for COVID-19. Pulm Pharmacol Ther 2021; 69:102035. [PMID: 33933611 PMCID: PMC8084922 DOI: 10.1016/j.pupt.2021.102035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 03/30/2021] [Accepted: 04/22/2021] [Indexed: 02/08/2023]
Abstract
The novel coronavirus 2019 (COVID-19) infection caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a global pandemic that requires a multi-faceted approach to tackle this unprecedent health crisis. Therapeutics to treat COVID-19 are an integral part of any such management strategy and there is a substantial unmet need for treatments for individuals most at risk of severe disease. This perspective review provides rationale of a combined therapeutic regimen of selective endothelin-A (ET-A) receptor antagonism and sodium glucose co-transporter-2 (SGLT-2) inhibition to treat COVID-19. Endothelin is a potent vasoconstrictor with pro-inflammatory and atherosclerotic effects. It is upregulated in a number of conditions including acute respiratory distress syndrome and cardiovascular disease. Endothelin mediates vasocontractility via endothelin (ET-A and ET-B) receptors on vascular smooth muscle cells (VSMCs). ET-B receptors regulate endothelin clearance and are present on endothelial cells, where in contrast to their role on VSMCs, mediate vasodilation. Therefore, selective endothelin-A (ET-A) receptor inhibition is likely the optimal approach to attenuate the injurious effects of endothelin and may reduce ventilation-perfusion mismatch and pulmonary inflammation, whilst improving pulmonary haemodynamics and oxygenation. SGLT-2 inhibition may dampen inflammatory cytokines, reduce hyperglycaemia if present, improve endothelial function, cardiovascular haemodynamics and cellular bioenergetics. This combination therapeutic approach may therefore have beneficial effects to mitigate both the pulmonary, metabolic and cardiorenal manifestations of COVID-19. Given these drug classes include medicines licensed to treat heart failure, diabetes and pulmonary hypertension respectively, information regarding their safety profile is established. Randomised controlled clinical trials are the best way to determine efficacy and safety of these medicines in COVID-19.
Collapse
|
145
|
Cardiovascular benefits of sodium-glucose cotransporter 2 inhibitors in diabetic and nondiabetic patients. Cardiovasc Diabetol 2021; 20:78. [PMID: 33827579 PMCID: PMC8028072 DOI: 10.1186/s12933-021-01266-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 03/19/2021] [Indexed: 12/25/2022] Open
Abstract
Sodium-glucose cotransporter 2 inhibitors (SGLT2i) were developed as antidiabetic agents, but accumulating evidence has shown their beneficial effects on the cardiovascular system. Analyses of the EMPA-REG OUTCOME trial (Empagliflozin Cardiovascular Outcome Event Trial in Type 2 Diabetes Mellitus Patients) suggested that these benefits are independent of glycemic control. Several large-scale outcome trials of SGLT2i also showed cardiovascular benefits in nondiabetic patients, strengthening this perspective. Extensive animal and clinical studies have likewise shown that mechanisms other than the antihyperglycemic effect underlie the cardiovascular benefits. Recent clinical guidelines recommend the use of SGLT2i in patients with type 2 diabetes mellitus and cardiovascular diseases because of the proven cardiovascular protective effects. Since the cardiovascular benefits are independent of glycemic control, the therapeutic spectrum of SGLT2i will likely be extended to nondiabetic patients.
Collapse
|
146
|
Abstract
Arterial stiffness, a leading marker of risk in hypertension, can be measured at material or structural levels, with the latter combining effects of the geometry and composition of the wall, including intramural organization. Numerous studies have shown that structural stiffness predicts outcomes in models that adjust for conventional risk factors. Elastic arteries, nearer to the heart, are most sensitive to effects of blood pressure and age, major determinants of stiffness. Stiffness is usually considered as an index of vascular aging, wherein individuals excessively affected by risk factor exposure represent early vascular aging, whereas those resistant to risk factors represent supernormal vascular aging. Stiffness affects the function of the brain and kidneys by increasing pulsatile loads within their microvascular beds, and the heart by increasing left ventricular systolic load; excessive pressure pulsatility also decreases diastolic pressure, necessary for coronary perfusion. Stiffness promotes inward remodeling of small arteries, which increases resistance, blood pressure, and in turn, central artery stiffness, thus creating an insidious feedback loop. Chronic antihypertensive treatments can reduce stiffness beyond passive reductions due to decreased blood pressure. Preventive drugs, such as lipid-lowering drugs and antidiabetic drugs, have additional effects on stiffness, independent of pressure. Newer anti-inflammatory drugs also have blood pressure independent effects. Reduction of stiffness is expected to confer benefit beyond the lowering of pressure, although this hypothesis is not yet proven. We summarize different steps for making arterial stiffness measurement a keystone in hypertension management and cardiovascular prevention as a whole.
Collapse
Affiliation(s)
- Pierre Boutouyrie
- Faculté de Médecine, Université de Paris, INSERM U970, Hôpital Européen Georges Pompidou, Assistance Publique Hôpitaux de Paris, France (P.B.)
| | - Phil Chowienczyk
- King's College London British Heart Foundation Centre, Department of Clinical Pharmacology, St Thomas' Hospital, London, United Kingdom (P.C.)
| | - Jay D Humphrey
- Department of Biomedical Engineering and Vascular Biology and Therapeutics Program, Yale University, New Haven, CT (J.D.H.)
| | | |
Collapse
|
147
|
Yu J, Arnott C, Neuen BL, Heersprink HL, Mahaffey KW, Cannon CP, Khan SS, Baldridge AS, Shah SJ, Huang Y, Li C, Figtree GA, Perkovic V, Jardine MJ, Neal B, Huffman MD. Cardiovascular and renal outcomes with canagliflozin according to baseline diuretic use: a post hoc analysis from the CANVAS Program. ESC Heart Fail 2021; 8:1482-1493. [PMID: 33595905 PMCID: PMC8006652 DOI: 10.1002/ehf2.13236] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/09/2020] [Accepted: 01/19/2021] [Indexed: 01/10/2023] Open
Abstract
AIMS The CANVAS Program identified the effect of canagliflozin on major adverse cardiovascular events (MACE) differed according to whether participants were using diuretics at study commencement. We sought to further evaluate this finding related to baseline differences, treatment effects, safety, and risk factor changes. METHODS AND RESULTS The CANVAS Program enrolled 10 142 participants with type 2 diabetes mellitus and high cardiovascular risk. Participants were randomized to canagliflozin or placebo and followed for a mean of 188 weeks. The primary outcome was major cardiovascular events, a composite of cardiovascular death, nonfatal myocardial infarction, or nonfatal stroke. Secondary outcomes included multiple cardiovascular, renal, and safety events. In this post hoc subgroup analysis, participants were categorized according to baseline use of any diuretic. The effect on outcomes was compared using Cox proportional hazards models, while risk factor changes were compared using mixed-effect models. At baseline, 4490 (44.3%) participants were using a diuretic. Compared with those not using a diuretic, participants using a diuretic were more likely to be older (mean age ± standard deviation, 64.3 ± 8.0 vs. 62.5 ± 8.3), be female (38.9% vs. 33.4%), and have heart failure (19.6% vs. 10.3%) (all Pdifference < 0.0001). The effect of canagliflozin on major cardiovascular events was greater for those using diuretic at baseline than for those who were not [adjusted hazard ratio 0.65 (95% confidence interval 0.54-0.78) vs. adjusted hazard ratio 1.13 (95% confidence interval 0.93-1.36), Pheterogeneity < 0.0001]. Changes in most risk factors, including blood pressure, body weight, and urine albumin-to-creatinine ratio, were similar between groups (all Pdifference > 0.11), although the effect of canagliflozin on haemoglobin A1c reduction was slightly weaker in participants using compared with not using diuretics at baseline (-0.52% vs. -0.64%, Pheterogeneity = 0.0007). Overall serious adverse events and key safety outcomes, including adverse renal events, were also similar (all Pheterogeneity > 0.07). CONCLUSIONS Participants on baseline diuretics derived a greater benefit for major cardiovascular events from canagliflozin, which was not fully explained by differences in participant characteristics nor risk factor changes.
Collapse
Affiliation(s)
- Jie Yu
- The George Institute for Global HealthUNSW SydneySydneyAustralia
- Department of CardiologyPeking University Third HospitalBeijingChina
- Faculty of MedicineUniversity of New South Wales, SydneySydneyAustralia
| | - Clare Arnott
- The George Institute for Global HealthUNSW SydneySydneyAustralia
- Department of CardiologyRoyal Prince Alfred HospitalSydneyAustralia
- Sydney Medical SchoolUniversity of SydneySydneyAustralia
- Faculty of MedicineUniversity of New South Wales, SydneySydneyAustralia
| | - Brendon L. Neuen
- The George Institute for Global HealthUNSW SydneySydneyAustralia
- Faculty of MedicineUniversity of New South Wales, SydneySydneyAustralia
| | - Hiddo L. Heersprink
- The George Institute for Global HealthUNSW SydneySydneyAustralia
- Department of Clinical Pharmacy and PharmacologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Kenneth W. Mahaffey
- Stanford Center for Clinical Research, Department of MedicineStanford University School of MedicineStanfordCAUSA
| | | | - Sadiya S. Khan
- Center for Global Cardiovascular HealthNorthwestern University Feinberg School of Medicine710 N. Lake Shore Drive, Suite 800ChicagoIL60611USA
| | - Abigail S. Baldridge
- Center for Global Cardiovascular HealthNorthwestern University Feinberg School of Medicine710 N. Lake Shore Drive, Suite 800ChicagoIL60611USA
| | - Sanjiv J. Shah
- Center for Global Cardiovascular HealthNorthwestern University Feinberg School of Medicine710 N. Lake Shore Drive, Suite 800ChicagoIL60611USA
| | - Yuli Huang
- The George Institute for Global HealthUNSW SydneySydneyAustralia
| | - Chao Li
- The George Institute for Global HealthUNSW SydneySydneyAustralia
| | - Gemma A. Figtree
- The George Institute for Global HealthUNSW SydneySydneyAustralia
- Sydney Medical SchoolUniversity of SydneySydneyAustralia
- Kolling InstituteRoyal North Shore Hospital and University of SydneySydneyAustralia
| | - Vlado Perkovic
- The George Institute for Global HealthUNSW SydneySydneyAustralia
- Faculty of MedicineUniversity of New South Wales, SydneySydneyAustralia
| | - Meg J. Jardine
- The George Institute for Global HealthUNSW SydneySydneyAustralia
- Faculty of MedicineUniversity of New South Wales, SydneySydneyAustralia
| | - Bruce Neal
- The George Institute for Global HealthUNSW SydneySydneyAustralia
- The Charles Perkins CentreUniversity of SydneySydneyAustralia
- Faculty of Clinical EpidemiologyImperial College LondonLondonUK
| | - Mark D. Huffman
- The George Institute for Global HealthUNSW SydneySydneyAustralia
- Center for Global Cardiovascular HealthNorthwestern University Feinberg School of Medicine710 N. Lake Shore Drive, Suite 800ChicagoIL60611USA
| |
Collapse
|
148
|
Kosiborod M, Berwanger O, Koch GG, Martinez F, Mukhtar O, Verma S, Chopra V, Javaheri A, Ambery P, Gasparyan SB, Buenconsejo J, Sjöström CD, Langkilde AM, Oscarsson J, Esterline R. Effects of dapagliflozin on prevention of major clinical events and recovery in patients with respiratory failure because of COVID-19: Design and rationale for the DARE-19 study. Diabetes Obes Metab 2021; 23:886-896. [PMID: 33319454 PMCID: PMC8049025 DOI: 10.1111/dom.14296] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023]
Abstract
AIMS Coronavirus disease 2019 (COVID-19) is caused by a novel severe acute respiratory syndrome coronavirus 2. It can lead to multiorgan failure, including respiratory and cardiovascular decompensation, and kidney injury, with significant associated morbidity and mortality, particularly in patients with underlying metabolic, cardiovascular, respiratory or kidney disease. Dapagliflozin, a sodium-glucose cotransporter-2 inhibitor, has shown significant cardio- and renoprotective benefits in patients with type 2 diabetes (with and without atherosclerotic cardiovascular disease), heart failure and chronic kidney disease, and may provide similar organ protection in high-risk patients with COVID-19. MATERIALS AND METHODS DARE-19 (NCT04350593) is an investigator-initiated, collaborative, international, multicentre, randomized, double-blind, placebo-controlled study testing the dual hypotheses that dapagliflozin can reduce the incidence of cardiovascular, kidney and/or respiratory complications or all-cause mortality, or improve clinical recovery, in adult patients hospitalized with COVID-19 but not critically ill on admission. Eligible patients will have ≥1 cardiometabolic risk factor for COVID-19 complications. Patients will be randomized 1:1 to dapagliflozin 10 mg or placebo. Primary efficacy endpoints are time to development of new or worsened organ dysfunction during index hospitalization, or all-cause mortality, and the hierarchical composite endpoint of change in clinical status through day 30 of treatment. Safety of dapagliflozin in individuals with COVID-19 will be assessed. CONCLUSIONS DARE-19 will evaluate whether dapagliflozin can prevent COVID-19-related complications and all-cause mortality, or improve clinical recovery, and assess the safety profile of dapagliflozin in this patient population. Currently, DARE-19 is the first large randomized controlled trial investigating use of sodium-glucose cotransporter 2 inhibitors in patients with COVID-19.
Collapse
Affiliation(s)
- Mikhail Kosiborod
- Saint Luke's Mid America Heart InstituteUniversity of MissouriKansas CityMissouriUSA
- The George Institute for Global HealthUniversity of New South WalesSydneyNew South WalesAustralia
| | - Otavio Berwanger
- Academic Research Organization ‐ Hospital Israelita Albert EinsteinSão PauloBrazil
| | - Gary G. Koch
- The University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | | | - Omar Mukhtar
- Experimental Medicine & Immunotherapeutics Division, Department of MedicineUniversity of CambridgeCambridgeUK
| | - Subodh Verma
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical ScienceLi Ka Shing Knowledge Institute of St. Michael's HospitalTorontoOntarioCanada
- Department of SurgeryUniversity of TorontoTorontoOntarioCanada
- Department of Pharmacology and ToxicologyUniversity of TorontoTorontoOntarioCanada
| | | | - Ali Javaheri
- Washington University School of MedicineSt LouisMissouriUSA
| | - Philip Ambery
- Late‐Stage Development, CVRM, BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Samvel B. Gasparyan
- Late‐Stage Development, CVRM, BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Joan Buenconsejo
- Late‐Stage Development, CVRM, BioPharmaceuticals R&DAstraZenecaGaithersburgMarylandUSA
| | - C. David Sjöström
- Late‐Stage Development, CVRM, BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | | | - Jan Oscarsson
- Late‐Stage Development, CVRM, BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Russell Esterline
- Late‐Stage Development, CVRM, BioPharmaceuticals R&DAstraZenecaGaithersburgMarylandUSA
| |
Collapse
|
149
|
Koo BK. Letter: Effects of 6 Months of Dapagliflozin Treatment on Metabolic Profile and Endothelial Cell Dysfunction for Obese Type 2 Diabetes Mellitus Patients without Atherosclerotic Cardiovascular Disease (J Obes Metab Syndr 2020;29:215-21). J Obes Metab Syndr 2021; 30:72-73. [PMID: 33518535 PMCID: PMC8017332 DOI: 10.7570/jomes20133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 12/21/2020] [Accepted: 12/21/2020] [Indexed: 01/07/2023] Open
Affiliation(s)
- Bo Kyung Koo
- Department of Internal Medicine, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
150
|
Sposito AC, Breder I, Soares AAS, Kimura-Medorima ST, Munhoz DB, Cintra RMR, Bonilha I, Oliveira DC, Breder JC, Cavalcante P, Moreira C, Moura FA, de Lima-Junior JC, do Carmo HRP, Barreto J, Nadruz W, Carvalho LSF, Quinaglia T. Dapagliflozin effect on endothelial dysfunction in diabetic patients with atherosclerotic disease: a randomized active-controlled trial. Cardiovasc Diabetol 2021; 20:74. [PMID: 33771149 PMCID: PMC8004411 DOI: 10.1186/s12933-021-01264-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/16/2021] [Indexed: 12/30/2022] Open
Abstract
Background The glucose-lowering independent effect of sodium glucose cotransporter-2 inhibitors (SGLT2i) on arterial wall function has not yet been clarified. This study aims to assess whether SGLT2i treatment can attenuate endothelial dysfunction related to type 2 diabetes mellitus (T2D) compared with glucose-lowering equivalent therapy. Methods In a prospective, open-label, single-center, randomized clinical trial, 98 patients with T2DM and carotid intima-media thickness above the 75th percentile were randomized 1:1 to 12 weeks of therapy with dapagliflozin or glibenclamide in addition to metformin in glucose-lowering equivalent regimens. The coprimary endpoints were 1-min flow-mediated dilation (FMD) at rest and 1-min FMD after 15 min of ischemia followed by 15 min of reperfusion time (I/R). Results Ninety-seven patients (61% males, 57 ± 7 years) completed the study. The median HbA1c decreased by − 0.8 (0.7)% and -0.7 (0.95)% following dapagliflozin and glibenclamide, respectively. The first coprimary endpoint, i.e., rest FMD changed by + 3.3(8.2)% and − 1.2(7.5)% for the dapagliflozin and glibenclamide arms, respectively (p = 0.0001). Differences between study arms in the second coprimary endpoint were not significant. Plasma nitrite 1 min after rest FMD was higher for dapagliflozin [308(220) nmol/L] than for glibenclamide (258[110] nmol/L; p = 0.028). The resistive indices at 1 min [0.90 (0.11) vs. 0.93 (0.07); p = 0.03] and 5 min [0.93 (0.07) vs. 0.95 (0.05); p = 0.02] were higher for the glibenclamide group than for the dapagliflozin group. Plasma biomarkers for inflammation and oxidative stress did not differ between the treatments. Conclusions Dapagliflozin improved micro- and macrovascular endothelial function compared to glibenclamide, regardless of glycemic control in patients with T2DM and subclinical carotid atherosclerotic disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12933-021-01264-z.
Collapse
Affiliation(s)
- Andrei C Sposito
- Aterosclerose and Vascular Biology Laboratory (Aterolab), Cardiology Division, State University of Campinas Medical School, Rua Tessalia Vieira de Camargo 126, Cidade Universitaria Zeferino Vaz, Campinas, SP, 13084-971, Brazil.
| | - Ikaro Breder
- Aterosclerose and Vascular Biology Laboratory (Aterolab), Cardiology Division, State University of Campinas Medical School, Rua Tessalia Vieira de Camargo 126, Cidade Universitaria Zeferino Vaz, Campinas, SP, 13084-971, Brazil
| | - Alexandre A S Soares
- Aterosclerose and Vascular Biology Laboratory (Aterolab), Cardiology Division, State University of Campinas Medical School, Rua Tessalia Vieira de Camargo 126, Cidade Universitaria Zeferino Vaz, Campinas, SP, 13084-971, Brazil
| | - Sheila T Kimura-Medorima
- Aterosclerose and Vascular Biology Laboratory (Aterolab), Cardiology Division, State University of Campinas Medical School, Rua Tessalia Vieira de Camargo 126, Cidade Universitaria Zeferino Vaz, Campinas, SP, 13084-971, Brazil
| | - Daniel B Munhoz
- Aterosclerose and Vascular Biology Laboratory (Aterolab), Cardiology Division, State University of Campinas Medical School, Rua Tessalia Vieira de Camargo 126, Cidade Universitaria Zeferino Vaz, Campinas, SP, 13084-971, Brazil
| | - Riobaldo M R Cintra
- Aterosclerose and Vascular Biology Laboratory (Aterolab), Cardiology Division, State University of Campinas Medical School, Rua Tessalia Vieira de Camargo 126, Cidade Universitaria Zeferino Vaz, Campinas, SP, 13084-971, Brazil
| | - Isabella Bonilha
- Aterosclerose and Vascular Biology Laboratory (Aterolab), Cardiology Division, State University of Campinas Medical School, Rua Tessalia Vieira de Camargo 126, Cidade Universitaria Zeferino Vaz, Campinas, SP, 13084-971, Brazil
| | - Daniela C Oliveira
- Aterosclerose and Vascular Biology Laboratory (Aterolab), Cardiology Division, State University of Campinas Medical School, Rua Tessalia Vieira de Camargo 126, Cidade Universitaria Zeferino Vaz, Campinas, SP, 13084-971, Brazil
| | - Jessica Cunha Breder
- Aterosclerose and Vascular Biology Laboratory (Aterolab), Cardiology Division, State University of Campinas Medical School, Rua Tessalia Vieira de Camargo 126, Cidade Universitaria Zeferino Vaz, Campinas, SP, 13084-971, Brazil
| | - Pamela Cavalcante
- Aterosclerose and Vascular Biology Laboratory (Aterolab), Cardiology Division, State University of Campinas Medical School, Rua Tessalia Vieira de Camargo 126, Cidade Universitaria Zeferino Vaz, Campinas, SP, 13084-971, Brazil
| | - Camila Moreira
- Aterosclerose and Vascular Biology Laboratory (Aterolab), Cardiology Division, State University of Campinas Medical School, Rua Tessalia Vieira de Camargo 126, Cidade Universitaria Zeferino Vaz, Campinas, SP, 13084-971, Brazil
| | - Filipe A Moura
- Aterosclerose and Vascular Biology Laboratory (Aterolab), Cardiology Division, State University of Campinas Medical School, Rua Tessalia Vieira de Camargo 126, Cidade Universitaria Zeferino Vaz, Campinas, SP, 13084-971, Brazil
| | - Jose Carlos de Lima-Junior
- Aterosclerose and Vascular Biology Laboratory (Aterolab), Cardiology Division, State University of Campinas Medical School, Rua Tessalia Vieira de Camargo 126, Cidade Universitaria Zeferino Vaz, Campinas, SP, 13084-971, Brazil
| | - Helison R P do Carmo
- Aterosclerose and Vascular Biology Laboratory (Aterolab), Cardiology Division, State University of Campinas Medical School, Rua Tessalia Vieira de Camargo 126, Cidade Universitaria Zeferino Vaz, Campinas, SP, 13084-971, Brazil
| | - Joaquim Barreto
- Aterosclerose and Vascular Biology Laboratory (Aterolab), Cardiology Division, State University of Campinas Medical School, Rua Tessalia Vieira de Camargo 126, Cidade Universitaria Zeferino Vaz, Campinas, SP, 13084-971, Brazil
| | - Wilson Nadruz
- Aterosclerose and Vascular Biology Laboratory (Aterolab), Cardiology Division, State University of Campinas Medical School, Rua Tessalia Vieira de Camargo 126, Cidade Universitaria Zeferino Vaz, Campinas, SP, 13084-971, Brazil
| | - Luiz Sergio F Carvalho
- Aterosclerose and Vascular Biology Laboratory (Aterolab), Cardiology Division, State University of Campinas Medical School, Rua Tessalia Vieira de Camargo 126, Cidade Universitaria Zeferino Vaz, Campinas, SP, 13084-971, Brazil
| | - Thiago Quinaglia
- Aterosclerose and Vascular Biology Laboratory (Aterolab), Cardiology Division, State University of Campinas Medical School, Rua Tessalia Vieira de Camargo 126, Cidade Universitaria Zeferino Vaz, Campinas, SP, 13084-971, Brazil
| | | |
Collapse
|