Abstract

Aims

The beneficial cardiovascular effects of sodium-glucose cotransporter 2 (SGLT2) inhibitors irrespective of the presence of diabetes mellitus are nowadays well established and they already constitute a significant pillar for the management of heart failure, irrespective of the ejection fraction. The exact underlying mechanisms accountable for these effects, however, remain largely unknown. The direct effect on endothelial function and microcirculation is one of the most well studied. The broad range of studies presented in this review aims to link all available data from the bench to bedside and highlight the existing gaps as well as the future directions in the investigations concerning the effects of SGLT2 inhibitors on the endothelium and the microcirculation.

Methods and results

An extensive search has been conducted using the MEDLINE/PubMed database in order to identify the relevant studies. Preclinical data suggest that SGLT2 inhibitors directly affect endothelial function independently of glucose and specifically via several interplaying molecular pathways, resulting in improved vasodilation, increased NO production, enhanced mitochondrial homeostasis, endothelial cell viability, and angiogenesis as well as attenuation of oxidative stress and inflammation. Clinical data systematically confirm this beneficial effect on the endothelium, whereas the evidence concerning the effect on the microcirculation is conflicting.

Conclusion

Preclinical and clinical studies indicate that SGLT2 inhibitors attenuate endothelial and microvascular dysfunction via a combination of mechanisms, which play a role in their beneficial cardiovascular effect.

Introduction

Sodium-glucose cotransporter 2 (SGLT2) inhibitors are a novel class of anti-diabetic drugs that lower serum glucose levels by blocking its SGLT2 mediated glucose reabsorption in the kidney, regardless of insulin secretion or action.1 It was early recognized from the first clinical trials examining the effects of SGLT2 inhibition in diabetic patients, such as EMPA-REG OUTCOME,2 that treatment with SGLT2 inhibitors reduces the risk of adverse cardiovascular outcomes and death from any cause in diabetic patients with cardiovascular disease. These findings were also consistent among future trials.3,4 Further research examining the cardioprotective role of these drugs in cardiovascular disease, and specifically in heart failure5–7 established that SGLT2 inhibition significantly reduces the risk of worsening heart failure or death from cardiovascular causes, regardless of the presence or absence of type 2 diabetes or HF phenotype. These results led to the recommendation of SGLT2 inhibitors for patients with HF in all phenotypes of the disease in the updated 2022 guidelines8 by the American Heart Association, the American College of Cardiology, and the Heart Failure Society of America.

Given the paramount significance of the positive results in HF, an increasing interest is being developed regarding the pathophysiologic pathways which SGLT2 inhibitors alter, resulting in cardiovascular protection. A handful of mechanisms have been proposed, including improved endothelial and microcirculatory function, reduction in myocardial fibrosis and arterial stiffness, enhancement of the systolic and diastolic cardiac function, weight loss and anti-hypertensive effect, diuresis-induced blood volume reduction, increased red blood cell mass, and improved myocardial bioenergetics.1,9 However, the exact underlying mechanisms are yet to be clarified, while understanding them, especially in endothelial dysfunction, will lead to more effective strategies for treating microvascular dysfunction (MVD) in HF and a broad range of other disorders whose pathophysiology associates with endothelial or MVD, such as diabetes, brain small-vessel disease, and chronic kidney disease (CKD).10,11

This review focuses on the effect of the SGLT2 inhibitors empagliflozin and dapagliflozin on the endothelium and the microcirculation, analyzing a broad range of data derived from research on endothelial cells and animal models, as well as from patient trials.

Pre-clinical data: assessing SGLT2 inhibition in the lab and animal studies

During the past few years, a large number of studies (Table 1) on murine and human endothelial cells have been conducted in order to evaluate the effect of SGLT2 inhibitors on the endothelium, proposing a variety of mechanisms by which they attenuate microvascular and endothelial dysfunction (Figure 1). These mechanisms, extensively analyzed below, mainly include nitric oxide production, mitochondrial function, oxidative stress, inflammation, cell viability, and angiogenesis.

The main mechanisms whereby SGLT2 inhibitors improve endothelial function.
Figure 1

The main mechanisms whereby SGLT2 inhibitors improve endothelial function.

Created with BioRender.com. Drp1, dynamin-related protein 1; EC, endothelial cell; eNOS, endothelial nitric oxide synthase; ICAM-1, intercellular adhesion molecule-1; NO, nitric oxide; ROS, reactive oxygen species; SGLT2 inhibitors, sodium-glucose cotransporter 2 inhibitors; VCAM-1, vascular cell adhesion molecule-1.

Table 1

Preclinical studies regarding the effect of SGLT2 inhibitors in endothelial function and microcirculation

AuthorDrug/DosageExperimental modelEffects of SGLT2 inhibitors on the endothelium
Adingupu et al., 20191Empagliflozin (1,5 mg/kg/day for 10 weeks)55 mice were divided into:
(1) ob/ob−/− obese, diabetic mice treated with
empagliflozin (n = 22)
(2) Untreated ob/ob−/− obese, diabetic mice (n = 21)
(3) Age-matched non-obese littermates as control
(n = 12)
● CFVR ↑
● NO ↑
● L-Arginine/ADMA ratio ↑
Bruckert et al., 202234Empagliflozin (30 mg/kg/day for 5 weeks)48 rats randomly divided into:
(1) Empagliflozin group
(2) Ang II group (hypertensive dose, 0.4 mg/kg/day)
(3) Empagliflozin & Ang II group
(4) Control group
Sub-studies of 3–12 animals per group
● eNOS ↑
● ROS ↓
● ICAM-1, VCAM-1, MCP-1, MMP-2, MMP-9 ↓
Cai et al., 202220Empagliflozin (10 mg/kg/day) for 7 daysCardiac microvascular endothelial cells (CMECs) of mice subjected to IRI
The mice were divided into:
(1) Sham operation group
(2) Sham operation & empagliflozin group
(3) I/R injury group
(4) I/R injury & empagliflozin group
Sub-studies of six animals per group
● eNOS ↑
● ROS ↓
● Mitochondrial fission ↓
● Mitochondrial apoptosis ↓
Campeau et al., 202241Empagliflozin (50 μM for 24 h)Human abdominal aortic endothelial cells (HAAECs) subjected to sustained GCX disruption● Inflammation caused by GCX disruption ↓
● Leukocyte (NB4 cells)—endothelium adhesion ↓
Cappetta et al., 202036Dapagliflozin (0,1 mg/kg/day for 6 weeks)67 rats were divided into:
(1) High-salt diet & dapagliflozin group (n = 25)
(2) High-salt diet & vehicle group (n = 30)
(3) Low-salt diet group (n = 12)
Sub-studies of 2–13 animals per group
● NHE1 activity ↓
● NF-kB, galectin-3 ↓
● VCAM-1, E-selectin, MCP-1, IL-6 ↓
Cooper et al., 201940Empagliflozin (50 μM for 24 h)Human abdominal aortic endothelial cells (HAAECs) exposed to TNF-α and wall shear stress● Glycocalyx integrity ↑
● Leukocyte (NB4 cells)—endothelium adhesion ↓
El-Daly et al., 201829Empagliflozin (25 nM up to 10 μM for 24 h)
Dapagliflozin (25 nM up to 10 μM for 24 h)
Mice aortic rings incubated in high glucose medium● NO-dependent relaxation through activation of PAR2 ↑
● EGFR-kinase/Src/Rho-kinase and PKC ↓
● ↓ ROS
Ganbaatar et al., 202038Empagliflozin (20 mg/kg/day for 8 or 12 weeks)Mice were randomly divided into:
(1) Control group
(2) Diabetic mice on vehicle
(3) Diabetic mice on empagliflozin
Sub-studies of 6–14 animals per group
● NOX-2, p22phox, MCP-1, ICAM-1, VCAM-1 ↓
● Vasoconstrictive eicosanoids (PGE2/TXB2) ↓
● Macrophage accumulation ↓
Gaspari et al., 201739Dapagliflozin (1 mg/kg/day for 4 weeks)






Dapagliflozin (1 nM to 1 mM for 24 h)
Aortic sections of Apo E−/− mice, which were divided into:
(1) Normal fat diet & vehicle
(2) High fat diet & vehicle
(3) Normal fat diet & dapagliflozin
(4) High fat diet & dapagliflozin
Sub-studies of 3–9 animals per group
HUVECs stimulated by TNF-α or hyperglycemia
● ICAM-1, VCAM ↓
● NF-kB ↓
He et al., 202230Dapagliflozin (1.0 mg/kg/day for 16 weeks)




Dapagliflozin (1 μΜ for 24 h)
Mice divided into:
(1) Normal diet group
(2) High-fat diet group
(3) High-fat diet & dapagliflozin group
Sub-studies of three animals per group
HUVECs stimulated with palmitic acid (PA)
● Angiogenesis ↑
● Cell viability ↑
● Mitochondrial homeostasis (SIRT1/PGC-1a pathway) ↑
Juni et al., 201925Empagliflozin (1 μM for 6 h)Human cardiac microvascular endothelial cells (CMECs) co-cultured with Cardiomyocytes (CMs) and exposed to TNF-α● Mitochondrial ROS ↓
● Cytoplasmic ROS ↓
● NO ↑
● Preserved CM contraction and relaxation
Juni et al., 202026Empagliflozin (1 μM for 6 h)Human cardiac microvascular endothelial cells (CMECs) co-cultured with Cardiomyocytes (CMs) and exposed to uraemic serum● Mitochondrial ROS ↓
● Cytoplasmic ROS ↓
● NO ↑
● Preserved CM contraction and relaxation
● Mitochondrial fragmentation ↓
Lee et al., 201819Dapagliflozin (60 mg/kg diet; 0.006%, for 8 weeks)47 Mice divided into:
(1) Control mice with standard diet (n = 11)
(2) Control mice with standard diet & dapagliflozin
(n = 12)
(3) Diabetic mice with standard diet (n = 12)
(4) Diabetic mice with standard diet & dapagliflozin
(n = 12)
Sub-studies of 3–12 animals per group
● Endothelium-dependent and -independent relaxation ↑
● PWV ↓
● IL-1β, IL-6, IL-10, IL-17, MCP-1, CCL-5 ↓
Li et al., 202133Empagliflozin (1 μΜ for 24 h)
Dapagliflozin (1 μΜ for 24 h)
Human coronary artery endothelial cells (HCAECs) subjected to cyclic stretch● NHE-1 and NOX-mediated ROS ↓
● Cell permeability ↓
● VE-cadherin ↑
Ma et al., 202224Dapagliflozin (40 mg/kg/day for 7 days)




Dapagliflozin (10 μΜ for 24 h)
Mice divided into:
(1) Sham operation group
(2) IRI group
(3) IRI & dapagliflozin group
Sub-studies of 6 or 10 animals per group
Human coronary artery endothelial cells (HCAECs) subjected to H/R injury
● eNOS ↑
● ICAM-1, IL-6, MCP1, TNF-α ↓
● VEGF ↑
● Mitochondrial fission ↓
● Cell survival ↑
● F-actin, Angiogenesis ↑
Mone et al., 202231Empagliflozin (1 μM for 24 h)Human umbilical vein endothelial cells (HUVECs) stimulated with high glucose levels
Human brain microvascular endothelial cells (hBMECs) stimulated with high glucose levels or H2O2
● Mitochondrial Ca2+ overload ↓
● Mitochondrial ROS ↓
● Endothelial leakage ↓ (claudin-5 and occluding mRNA ↑)
● Cell viability ↑
Nakao et al., 202112Empagliflozin (0,03% w/w)Murine model of left ventricular pressure overload generated by transverse aortic constriction (TAC)
Mice were divided into:
(1) TAC & empagliflozin group
(2) TAC—control group
(3) Sham operation & empagliflozin group
(4) Sham operation—control group
Sub-studies of 3–31 animals per group
● NO ↑
● eNOS ↑
Nikolaou et al., 2019 A35Empagliflozin (10 mg/kg/day for 6 weeks)IRI murine model
130 mice were divided into cohorts, with each cohort consisting of 2–4 groups with 5–8 animals each
● VEGF ↑
● Superoxide dismutase 2 ↑
● Cell survival ↑
Nikolaou et al., 2019 B35Empagliflozin (500 nM for 24 h)Human cardiac microvascular endothelial cells (CMECs) subjected to H/R injury● ROS ↓
● Cell survival ↑
Oelze et al., 201418Empagliflozin (10 and 30 mg/kg/day for 7 weeks)48 mice divided into:
(1) Untreated controls
(2) Diabetic mice & placebo
(3) Diabetic mice & low dose empagliflozin
(4) Diabetic mice & high dose empagliflozin
Three sub-studies were performed with 3,4, and 5 animals per group
● NO-dependent vasodilation ↑
● eNOSSer1177 phosphorylation ↑
● ROS, NOX-1, NOX-2, HO-1 ↓
● CCL-2, CD68, IL-6, IFN-γ, TNF-α, ICAM-1 ↓
Park et al., 202016Empagliflozin (30 mg/kg/day for 6 weeks)40 mice were equally divided into:
(1) Lean control
(2) Lean control & empagliflozin
(3) Obese, diabetic, and hypertensive mice with HF
(4) Obese, diabetic, and hypertensive mice with HF &
empagliflozin
Sub-studies of 3–10 animals per group
● NO-mediated relaxation ↑
● COX-derived prostanoids ↓
● eNOS ↑
● senescence markers (p53,p21,p16), tissue factor, VCAM-1 ↓
Soares et al., 20229Empagliflozin (14 mg/kg/day for 6 weeks)Aged mice with confirmed vascular dysfunction were divided into:
(1) Empagliflozin group (n = 29)
(2) Control group (n = 29)
Sub-studies of 5–10 animals per group
● eNOS ↑
● ROS, MDA ↓
● F-actin, P-cofilin, arterial stiffness ↓
Steven et al., 201717Empagliflozin (10 and 30 mg/kg/day for 6 weeks)





Empagliflozin (1 or 10 μM for 6 days)
35 diabetic ZDF rats and 16 lean controls divided into:
(1) Lean controls
(2) Diabetic rats
(3) Diabetic rats on low dose empagliflozin
(4) Diabetic rats on high dose empagliflozin
Sub-studies of 4–16 animals per group
Human umbilical vein endothelial cells (HUVECs) exposed to Ach after cultured in high glucose medium
● NO-dependent vasodilation ↑
● eNOSThr495 phosphorylation ↓
● ROS ↓
● ECs viability ↑
● IFN-γ, COX2, NOS-2, P-selectin, ICAM-1 ↓
Uthman et al., 201922Empagliflozin (1 μM for 4–24 h)

Dapagliflozin (1 μM for 4–24 h)
Human coronary artery endothelial cells (HCAECs) exposed to TNF-α
Human umbilical vein endothelial cells (HUVECs) exposed to TNF-α
● NO ↑
● ROS ↓
Zhou et al., 201821Empagliflozin (10 mg/kg/day for 20 weeks)Streptozotocin (STZ)-induced diabetic mice were divided into:
(1) Control group (n = 6)
(2) Diabetic group (n = 6)
(3) Diabetic & empagliflozin group (n = 6)
Their cardiac microvascular endothelial cells (CMECs) were also used
● Endothelium-dependent relaxation ↑
● eNOSSer1177 phosphorylation ↑
● Cytoplasmic ROS ↓
● Mitochondrial ROS ↓
● Mitochondrial fission ↓
● F-actin, Angiogenesis ↑
● ICAM-1, VCAM-1 ↓
Zou et al., 202223Empagliflozin (10 mg/kg/day for 7 days)




Empagliflozin (10 µM for 12 h)
Mice were divided into:
(1) Sham operation group
(2) I/R group
(3) I/R & empagliflozin group
Sub-studies of 6 or 10 animals per group
Human coronary artery endothelial cells (HCAECs) subjected to IRI
● eNOS ↑
● Mitochondrial fission ↓
● Cytoplasmic ROS ↓
● Mitochondrial ROS ↓
● VE-cadherin ↑
● ICAM-1 ↓
AuthorDrug/DosageExperimental modelEffects of SGLT2 inhibitors on the endothelium
Adingupu et al., 20191Empagliflozin (1,5 mg/kg/day for 10 weeks)55 mice were divided into:
(1) ob/ob−/− obese, diabetic mice treated with
empagliflozin (n = 22)
(2) Untreated ob/ob−/− obese, diabetic mice (n = 21)
(3) Age-matched non-obese littermates as control
(n = 12)
● CFVR ↑
● NO ↑
● L-Arginine/ADMA ratio ↑
Bruckert et al., 202234Empagliflozin (30 mg/kg/day for 5 weeks)48 rats randomly divided into:
(1) Empagliflozin group
(2) Ang II group (hypertensive dose, 0.4 mg/kg/day)
(3) Empagliflozin & Ang II group
(4) Control group
Sub-studies of 3–12 animals per group
● eNOS ↑
● ROS ↓
● ICAM-1, VCAM-1, MCP-1, MMP-2, MMP-9 ↓
Cai et al., 202220Empagliflozin (10 mg/kg/day) for 7 daysCardiac microvascular endothelial cells (CMECs) of mice subjected to IRI
The mice were divided into:
(1) Sham operation group
(2) Sham operation & empagliflozin group
(3) I/R injury group
(4) I/R injury & empagliflozin group
Sub-studies of six animals per group
● eNOS ↑
● ROS ↓
● Mitochondrial fission ↓
● Mitochondrial apoptosis ↓
Campeau et al., 202241Empagliflozin (50 μM for 24 h)Human abdominal aortic endothelial cells (HAAECs) subjected to sustained GCX disruption● Inflammation caused by GCX disruption ↓
● Leukocyte (NB4 cells)—endothelium adhesion ↓
Cappetta et al., 202036Dapagliflozin (0,1 mg/kg/day for 6 weeks)67 rats were divided into:
(1) High-salt diet & dapagliflozin group (n = 25)
(2) High-salt diet & vehicle group (n = 30)
(3) Low-salt diet group (n = 12)
Sub-studies of 2–13 animals per group
● NHE1 activity ↓
● NF-kB, galectin-3 ↓
● VCAM-1, E-selectin, MCP-1, IL-6 ↓
Cooper et al., 201940Empagliflozin (50 μM for 24 h)Human abdominal aortic endothelial cells (HAAECs) exposed to TNF-α and wall shear stress● Glycocalyx integrity ↑
● Leukocyte (NB4 cells)—endothelium adhesion ↓
El-Daly et al., 201829Empagliflozin (25 nM up to 10 μM for 24 h)
Dapagliflozin (25 nM up to 10 μM for 24 h)
Mice aortic rings incubated in high glucose medium● NO-dependent relaxation through activation of PAR2 ↑
● EGFR-kinase/Src/Rho-kinase and PKC ↓
● ↓ ROS
Ganbaatar et al., 202038Empagliflozin (20 mg/kg/day for 8 or 12 weeks)Mice were randomly divided into:
(1) Control group
(2) Diabetic mice on vehicle
(3) Diabetic mice on empagliflozin
Sub-studies of 6–14 animals per group
● NOX-2, p22phox, MCP-1, ICAM-1, VCAM-1 ↓
● Vasoconstrictive eicosanoids (PGE2/TXB2) ↓
● Macrophage accumulation ↓
Gaspari et al., 201739Dapagliflozin (1 mg/kg/day for 4 weeks)






Dapagliflozin (1 nM to 1 mM for 24 h)
Aortic sections of Apo E−/− mice, which were divided into:
(1) Normal fat diet & vehicle
(2) High fat diet & vehicle
(3) Normal fat diet & dapagliflozin
(4) High fat diet & dapagliflozin
Sub-studies of 3–9 animals per group
HUVECs stimulated by TNF-α or hyperglycemia
● ICAM-1, VCAM ↓
● NF-kB ↓
He et al., 202230Dapagliflozin (1.0 mg/kg/day for 16 weeks)




Dapagliflozin (1 μΜ for 24 h)
Mice divided into:
(1) Normal diet group
(2) High-fat diet group
(3) High-fat diet & dapagliflozin group
Sub-studies of three animals per group
HUVECs stimulated with palmitic acid (PA)
● Angiogenesis ↑
● Cell viability ↑
● Mitochondrial homeostasis (SIRT1/PGC-1a pathway) ↑
Juni et al., 201925Empagliflozin (1 μM for 6 h)Human cardiac microvascular endothelial cells (CMECs) co-cultured with Cardiomyocytes (CMs) and exposed to TNF-α● Mitochondrial ROS ↓
● Cytoplasmic ROS ↓
● NO ↑
● Preserved CM contraction and relaxation
Juni et al., 202026Empagliflozin (1 μM for 6 h)Human cardiac microvascular endothelial cells (CMECs) co-cultured with Cardiomyocytes (CMs) and exposed to uraemic serum● Mitochondrial ROS ↓
● Cytoplasmic ROS ↓
● NO ↑
● Preserved CM contraction and relaxation
● Mitochondrial fragmentation ↓
Lee et al., 201819Dapagliflozin (60 mg/kg diet; 0.006%, for 8 weeks)47 Mice divided into:
(1) Control mice with standard diet (n = 11)
(2) Control mice with standard diet & dapagliflozin
(n = 12)
(3) Diabetic mice with standard diet (n = 12)
(4) Diabetic mice with standard diet & dapagliflozin
(n = 12)
Sub-studies of 3–12 animals per group
● Endothelium-dependent and -independent relaxation ↑
● PWV ↓
● IL-1β, IL-6, IL-10, IL-17, MCP-1, CCL-5 ↓
Li et al., 202133Empagliflozin (1 μΜ for 24 h)
Dapagliflozin (1 μΜ for 24 h)
Human coronary artery endothelial cells (HCAECs) subjected to cyclic stretch● NHE-1 and NOX-mediated ROS ↓
● Cell permeability ↓
● VE-cadherin ↑
Ma et al., 202224Dapagliflozin (40 mg/kg/day for 7 days)




Dapagliflozin (10 μΜ for 24 h)
Mice divided into:
(1) Sham operation group
(2) IRI group
(3) IRI & dapagliflozin group
Sub-studies of 6 or 10 animals per group
Human coronary artery endothelial cells (HCAECs) subjected to H/R injury
● eNOS ↑
● ICAM-1, IL-6, MCP1, TNF-α ↓
● VEGF ↑
● Mitochondrial fission ↓
● Cell survival ↑
● F-actin, Angiogenesis ↑
Mone et al., 202231Empagliflozin (1 μM for 24 h)Human umbilical vein endothelial cells (HUVECs) stimulated with high glucose levels
Human brain microvascular endothelial cells (hBMECs) stimulated with high glucose levels or H2O2
● Mitochondrial Ca2+ overload ↓
● Mitochondrial ROS ↓
● Endothelial leakage ↓ (claudin-5 and occluding mRNA ↑)
● Cell viability ↑
Nakao et al., 202112Empagliflozin (0,03% w/w)Murine model of left ventricular pressure overload generated by transverse aortic constriction (TAC)
Mice were divided into:
(1) TAC & empagliflozin group
(2) TAC—control group
(3) Sham operation & empagliflozin group
(4) Sham operation—control group
Sub-studies of 3–31 animals per group
● NO ↑
● eNOS ↑
Nikolaou et al., 2019 A35Empagliflozin (10 mg/kg/day for 6 weeks)IRI murine model
130 mice were divided into cohorts, with each cohort consisting of 2–4 groups with 5–8 animals each
● VEGF ↑
● Superoxide dismutase 2 ↑
● Cell survival ↑
Nikolaou et al., 2019 B35Empagliflozin (500 nM for 24 h)Human cardiac microvascular endothelial cells (CMECs) subjected to H/R injury● ROS ↓
● Cell survival ↑
Oelze et al., 201418Empagliflozin (10 and 30 mg/kg/day for 7 weeks)48 mice divided into:
(1) Untreated controls
(2) Diabetic mice & placebo
(3) Diabetic mice & low dose empagliflozin
(4) Diabetic mice & high dose empagliflozin
Three sub-studies were performed with 3,4, and 5 animals per group
● NO-dependent vasodilation ↑
● eNOSSer1177 phosphorylation ↑
● ROS, NOX-1, NOX-2, HO-1 ↓
● CCL-2, CD68, IL-6, IFN-γ, TNF-α, ICAM-1 ↓
Park et al., 202016Empagliflozin (30 mg/kg/day for 6 weeks)40 mice were equally divided into:
(1) Lean control
(2) Lean control & empagliflozin
(3) Obese, diabetic, and hypertensive mice with HF
(4) Obese, diabetic, and hypertensive mice with HF &
empagliflozin
Sub-studies of 3–10 animals per group
● NO-mediated relaxation ↑
● COX-derived prostanoids ↓
● eNOS ↑
● senescence markers (p53,p21,p16), tissue factor, VCAM-1 ↓
Soares et al., 20229Empagliflozin (14 mg/kg/day for 6 weeks)Aged mice with confirmed vascular dysfunction were divided into:
(1) Empagliflozin group (n = 29)
(2) Control group (n = 29)
Sub-studies of 5–10 animals per group
● eNOS ↑
● ROS, MDA ↓
● F-actin, P-cofilin, arterial stiffness ↓
Steven et al., 201717Empagliflozin (10 and 30 mg/kg/day for 6 weeks)





Empagliflozin (1 or 10 μM for 6 days)
35 diabetic ZDF rats and 16 lean controls divided into:
(1) Lean controls
(2) Diabetic rats
(3) Diabetic rats on low dose empagliflozin
(4) Diabetic rats on high dose empagliflozin
Sub-studies of 4–16 animals per group
Human umbilical vein endothelial cells (HUVECs) exposed to Ach after cultured in high glucose medium
● NO-dependent vasodilation ↑
● eNOSThr495 phosphorylation ↓
● ROS ↓
● ECs viability ↑
● IFN-γ, COX2, NOS-2, P-selectin, ICAM-1 ↓
Uthman et al., 201922Empagliflozin (1 μM for 4–24 h)

Dapagliflozin (1 μM for 4–24 h)
Human coronary artery endothelial cells (HCAECs) exposed to TNF-α
Human umbilical vein endothelial cells (HUVECs) exposed to TNF-α
● NO ↑
● ROS ↓
Zhou et al., 201821Empagliflozin (10 mg/kg/day for 20 weeks)Streptozotocin (STZ)-induced diabetic mice were divided into:
(1) Control group (n = 6)
(2) Diabetic group (n = 6)
(3) Diabetic & empagliflozin group (n = 6)
Their cardiac microvascular endothelial cells (CMECs) were also used
● Endothelium-dependent relaxation ↑
● eNOSSer1177 phosphorylation ↑
● Cytoplasmic ROS ↓
● Mitochondrial ROS ↓
● Mitochondrial fission ↓
● F-actin, Angiogenesis ↑
● ICAM-1, VCAM-1 ↓
Zou et al., 202223Empagliflozin (10 mg/kg/day for 7 days)




Empagliflozin (10 µM for 12 h)
Mice were divided into:
(1) Sham operation group
(2) I/R group
(3) I/R & empagliflozin group
Sub-studies of 6 or 10 animals per group
Human coronary artery endothelial cells (HCAECs) subjected to IRI
● eNOS ↑
● Mitochondrial fission ↓
● Cytoplasmic ROS ↓
● Mitochondrial ROS ↓
● VE-cadherin ↑
● ICAM-1 ↓

Abbreviations: ADMA ratio, asymmetric dimethylarginine ratio; Apo E, apolipoprotein E; CCL-5, chemokine (C-C motif) ligand-5; CD68, cluster of differentiation 68; CFVR, coronary flow velocity reserve; cGMP, cyclic guanosine monophosphate; CMECs, cardiac microvascular endothelial cells; CMs, cardiomyocytes; COX, cyclooxygenase; COX-2 = cyclooxygenase-2; EGFR, epidermal growth factor receptor; eNOS, endothelial nitric oxide synthase; GCX, glycocalyx; HAAECs, human abdominal aortic endothelial cells; hBMECs, human brain microvascular endothelial cells; HCAECs, human coronary artery endothelial cells; HO-1, heme oxygenase-1; H/R injury, hypoxia/reoxygenation injury; HUVECs, human umbilical vein endothelial cells; ICAM-1, intercellular adhesion molecule-1; IFN-γ, interferon-γ; IL-1β, interleukin-1β; IL-6, interleukin-6; IL-10, interleukin-10; IL-17, interleukin-17; IRI, ischemia/reperfusion injury; MCP-1, monocyte chemotactic protein-1; MDA, malondialdehyde; MMP-2, matrix metalloproteinase-2; MMP-9, matrix metalloproteinase-9; NF-kB, nuclear factor-kB; NHE1, Na+/H+ exchanger 1; NO, nitric oxide; NOS-2, inducible NO synthase-2; NOX-1, NADPH oxidase-1; NOX-2, NADPH oxidase-2; Ob, obese; p22phox, 22 kilodalton phagocyte oxidase; PAR2, proteinase-activated receptor 2; PGC-1a, peroxisome proliferator-activated receptor-gamma coactivator -1a; PGE2, prostaglandin E2; PKC, protein kinase C; PKG, protein kinase G; PWV, pulse wave velocity; Rho, Ras homologous; ROS, reactive oxygen species; SIRT1, silent information regulator 1; Src, sarcoma virus tyrosine kinase; TNF-α ,tumor necrosis factor-α; TXB2, thromboxane B2; VCAM-1, vascular cell adhesion molecule-1; VE-cadherin, vascular endothelial-cadherin; VEGF, vascular endothelial growth factor; VSMCs, vascular smooth muscle cells; ZSF1, Zucker diabetic fatty/spontaneously hypertensive heart failure F1 hybrid; ZDF rats, Zucker Diabetic fatty rats.

Table 1

Preclinical studies regarding the effect of SGLT2 inhibitors in endothelial function and microcirculation

AuthorDrug/DosageExperimental modelEffects of SGLT2 inhibitors on the endothelium
Adingupu et al., 20191Empagliflozin (1,5 mg/kg/day for 10 weeks)55 mice were divided into:
(1) ob/ob−/− obese, diabetic mice treated with
empagliflozin (n = 22)
(2) Untreated ob/ob−/− obese, diabetic mice (n = 21)
(3) Age-matched non-obese littermates as control
(n = 12)
● CFVR ↑
● NO ↑
● L-Arginine/ADMA ratio ↑
Bruckert et al., 202234Empagliflozin (30 mg/kg/day for 5 weeks)48 rats randomly divided into:
(1) Empagliflozin group
(2) Ang II group (hypertensive dose, 0.4 mg/kg/day)
(3) Empagliflozin & Ang II group
(4) Control group
Sub-studies of 3–12 animals per group
● eNOS ↑
● ROS ↓
● ICAM-1, VCAM-1, MCP-1, MMP-2, MMP-9 ↓
Cai et al., 202220Empagliflozin (10 mg/kg/day) for 7 daysCardiac microvascular endothelial cells (CMECs) of mice subjected to IRI
The mice were divided into:
(1) Sham operation group
(2) Sham operation & empagliflozin group
(3) I/R injury group
(4) I/R injury & empagliflozin group
Sub-studies of six animals per group
● eNOS ↑
● ROS ↓
● Mitochondrial fission ↓
● Mitochondrial apoptosis ↓
Campeau et al., 202241Empagliflozin (50 μM for 24 h)Human abdominal aortic endothelial cells (HAAECs) subjected to sustained GCX disruption● Inflammation caused by GCX disruption ↓
● Leukocyte (NB4 cells)—endothelium adhesion ↓
Cappetta et al., 202036Dapagliflozin (0,1 mg/kg/day for 6 weeks)67 rats were divided into:
(1) High-salt diet & dapagliflozin group (n = 25)
(2) High-salt diet & vehicle group (n = 30)
(3) Low-salt diet group (n = 12)
Sub-studies of 2–13 animals per group
● NHE1 activity ↓
● NF-kB, galectin-3 ↓
● VCAM-1, E-selectin, MCP-1, IL-6 ↓
Cooper et al., 201940Empagliflozin (50 μM for 24 h)Human abdominal aortic endothelial cells (HAAECs) exposed to TNF-α and wall shear stress● Glycocalyx integrity ↑
● Leukocyte (NB4 cells)—endothelium adhesion ↓
El-Daly et al., 201829Empagliflozin (25 nM up to 10 μM for 24 h)
Dapagliflozin (25 nM up to 10 μM for 24 h)
Mice aortic rings incubated in high glucose medium● NO-dependent relaxation through activation of PAR2 ↑
● EGFR-kinase/Src/Rho-kinase and PKC ↓
● ↓ ROS
Ganbaatar et al., 202038Empagliflozin (20 mg/kg/day for 8 or 12 weeks)Mice were randomly divided into:
(1) Control group
(2) Diabetic mice on vehicle
(3) Diabetic mice on empagliflozin
Sub-studies of 6–14 animals per group
● NOX-2, p22phox, MCP-1, ICAM-1, VCAM-1 ↓
● Vasoconstrictive eicosanoids (PGE2/TXB2) ↓
● Macrophage accumulation ↓
Gaspari et al., 201739Dapagliflozin (1 mg/kg/day for 4 weeks)






Dapagliflozin (1 nM to 1 mM for 24 h)
Aortic sections of Apo E−/− mice, which were divided into:
(1) Normal fat diet & vehicle
(2) High fat diet & vehicle
(3) Normal fat diet & dapagliflozin
(4) High fat diet & dapagliflozin
Sub-studies of 3–9 animals per group
HUVECs stimulated by TNF-α or hyperglycemia
● ICAM-1, VCAM ↓
● NF-kB ↓
He et al., 202230Dapagliflozin (1.0 mg/kg/day for 16 weeks)




Dapagliflozin (1 μΜ for 24 h)
Mice divided into:
(1) Normal diet group
(2) High-fat diet group
(3) High-fat diet & dapagliflozin group
Sub-studies of three animals per group
HUVECs stimulated with palmitic acid (PA)
● Angiogenesis ↑
● Cell viability ↑
● Mitochondrial homeostasis (SIRT1/PGC-1a pathway) ↑
Juni et al., 201925Empagliflozin (1 μM for 6 h)Human cardiac microvascular endothelial cells (CMECs) co-cultured with Cardiomyocytes (CMs) and exposed to TNF-α● Mitochondrial ROS ↓
● Cytoplasmic ROS ↓
● NO ↑
● Preserved CM contraction and relaxation
Juni et al., 202026Empagliflozin (1 μM for 6 h)Human cardiac microvascular endothelial cells (CMECs) co-cultured with Cardiomyocytes (CMs) and exposed to uraemic serum● Mitochondrial ROS ↓
● Cytoplasmic ROS ↓
● NO ↑
● Preserved CM contraction and relaxation
● Mitochondrial fragmentation ↓
Lee et al., 201819Dapagliflozin (60 mg/kg diet; 0.006%, for 8 weeks)47 Mice divided into:
(1) Control mice with standard diet (n = 11)
(2) Control mice with standard diet & dapagliflozin
(n = 12)
(3) Diabetic mice with standard diet (n = 12)
(4) Diabetic mice with standard diet & dapagliflozin
(n = 12)
Sub-studies of 3–12 animals per group
● Endothelium-dependent and -independent relaxation ↑
● PWV ↓
● IL-1β, IL-6, IL-10, IL-17, MCP-1, CCL-5 ↓
Li et al., 202133Empagliflozin (1 μΜ for 24 h)
Dapagliflozin (1 μΜ for 24 h)
Human coronary artery endothelial cells (HCAECs) subjected to cyclic stretch● NHE-1 and NOX-mediated ROS ↓
● Cell permeability ↓
● VE-cadherin ↑
Ma et al., 202224Dapagliflozin (40 mg/kg/day for 7 days)




Dapagliflozin (10 μΜ for 24 h)
Mice divided into:
(1) Sham operation group
(2) IRI group
(3) IRI & dapagliflozin group
Sub-studies of 6 or 10 animals per group
Human coronary artery endothelial cells (HCAECs) subjected to H/R injury
● eNOS ↑
● ICAM-1, IL-6, MCP1, TNF-α ↓
● VEGF ↑
● Mitochondrial fission ↓
● Cell survival ↑
● F-actin, Angiogenesis ↑
Mone et al., 202231Empagliflozin (1 μM for 24 h)Human umbilical vein endothelial cells (HUVECs) stimulated with high glucose levels
Human brain microvascular endothelial cells (hBMECs) stimulated with high glucose levels or H2O2
● Mitochondrial Ca2+ overload ↓
● Mitochondrial ROS ↓
● Endothelial leakage ↓ (claudin-5 and occluding mRNA ↑)
● Cell viability ↑
Nakao et al., 202112Empagliflozin (0,03% w/w)Murine model of left ventricular pressure overload generated by transverse aortic constriction (TAC)
Mice were divided into:
(1) TAC & empagliflozin group
(2) TAC—control group
(3) Sham operation & empagliflozin group
(4) Sham operation—control group
Sub-studies of 3–31 animals per group
● NO ↑
● eNOS ↑
Nikolaou et al., 2019 A35Empagliflozin (10 mg/kg/day for 6 weeks)IRI murine model
130 mice were divided into cohorts, with each cohort consisting of 2–4 groups with 5–8 animals each
● VEGF ↑
● Superoxide dismutase 2 ↑
● Cell survival ↑
Nikolaou et al., 2019 B35Empagliflozin (500 nM for 24 h)Human cardiac microvascular endothelial cells (CMECs) subjected to H/R injury● ROS ↓
● Cell survival ↑
Oelze et al., 201418Empagliflozin (10 and 30 mg/kg/day for 7 weeks)48 mice divided into:
(1) Untreated controls
(2) Diabetic mice & placebo
(3) Diabetic mice & low dose empagliflozin
(4) Diabetic mice & high dose empagliflozin
Three sub-studies were performed with 3,4, and 5 animals per group
● NO-dependent vasodilation ↑
● eNOSSer1177 phosphorylation ↑
● ROS, NOX-1, NOX-2, HO-1 ↓
● CCL-2, CD68, IL-6, IFN-γ, TNF-α, ICAM-1 ↓
Park et al., 202016Empagliflozin (30 mg/kg/day for 6 weeks)40 mice were equally divided into:
(1) Lean control
(2) Lean control & empagliflozin
(3) Obese, diabetic, and hypertensive mice with HF
(4) Obese, diabetic, and hypertensive mice with HF &
empagliflozin
Sub-studies of 3–10 animals per group
● NO-mediated relaxation ↑
● COX-derived prostanoids ↓
● eNOS ↑
● senescence markers (p53,p21,p16), tissue factor, VCAM-1 ↓
Soares et al., 20229Empagliflozin (14 mg/kg/day for 6 weeks)Aged mice with confirmed vascular dysfunction were divided into:
(1) Empagliflozin group (n = 29)
(2) Control group (n = 29)
Sub-studies of 5–10 animals per group
● eNOS ↑
● ROS, MDA ↓
● F-actin, P-cofilin, arterial stiffness ↓
Steven et al., 201717Empagliflozin (10 and 30 mg/kg/day for 6 weeks)





Empagliflozin (1 or 10 μM for 6 days)
35 diabetic ZDF rats and 16 lean controls divided into:
(1) Lean controls
(2) Diabetic rats
(3) Diabetic rats on low dose empagliflozin
(4) Diabetic rats on high dose empagliflozin
Sub-studies of 4–16 animals per group
Human umbilical vein endothelial cells (HUVECs) exposed to Ach after cultured in high glucose medium
● NO-dependent vasodilation ↑
● eNOSThr495 phosphorylation ↓
● ROS ↓
● ECs viability ↑
● IFN-γ, COX2, NOS-2, P-selectin, ICAM-1 ↓
Uthman et al., 201922Empagliflozin (1 μM for 4–24 h)

Dapagliflozin (1 μM for 4–24 h)
Human coronary artery endothelial cells (HCAECs) exposed to TNF-α
Human umbilical vein endothelial cells (HUVECs) exposed to TNF-α
● NO ↑
● ROS ↓
Zhou et al., 201821Empagliflozin (10 mg/kg/day for 20 weeks)Streptozotocin (STZ)-induced diabetic mice were divided into:
(1) Control group (n = 6)
(2) Diabetic group (n = 6)
(3) Diabetic & empagliflozin group (n = 6)
Their cardiac microvascular endothelial cells (CMECs) were also used
● Endothelium-dependent relaxation ↑
● eNOSSer1177 phosphorylation ↑
● Cytoplasmic ROS ↓
● Mitochondrial ROS ↓
● Mitochondrial fission ↓
● F-actin, Angiogenesis ↑
● ICAM-1, VCAM-1 ↓
Zou et al., 202223Empagliflozin (10 mg/kg/day for 7 days)




Empagliflozin (10 µM for 12 h)
Mice were divided into:
(1) Sham operation group
(2) I/R group
(3) I/R & empagliflozin group
Sub-studies of 6 or 10 animals per group
Human coronary artery endothelial cells (HCAECs) subjected to IRI
● eNOS ↑
● Mitochondrial fission ↓
● Cytoplasmic ROS ↓
● Mitochondrial ROS ↓
● VE-cadherin ↑
● ICAM-1 ↓
AuthorDrug/DosageExperimental modelEffects of SGLT2 inhibitors on the endothelium
Adingupu et al., 20191Empagliflozin (1,5 mg/kg/day for 10 weeks)55 mice were divided into:
(1) ob/ob−/− obese, diabetic mice treated with
empagliflozin (n = 22)
(2) Untreated ob/ob−/− obese, diabetic mice (n = 21)
(3) Age-matched non-obese littermates as control
(n = 12)
● CFVR ↑
● NO ↑
● L-Arginine/ADMA ratio ↑
Bruckert et al., 202234Empagliflozin (30 mg/kg/day for 5 weeks)48 rats randomly divided into:
(1) Empagliflozin group
(2) Ang II group (hypertensive dose, 0.4 mg/kg/day)
(3) Empagliflozin & Ang II group
(4) Control group
Sub-studies of 3–12 animals per group
● eNOS ↑
● ROS ↓
● ICAM-1, VCAM-1, MCP-1, MMP-2, MMP-9 ↓
Cai et al., 202220Empagliflozin (10 mg/kg/day) for 7 daysCardiac microvascular endothelial cells (CMECs) of mice subjected to IRI
The mice were divided into:
(1) Sham operation group
(2) Sham operation & empagliflozin group
(3) I/R injury group
(4) I/R injury & empagliflozin group
Sub-studies of six animals per group
● eNOS ↑
● ROS ↓
● Mitochondrial fission ↓
● Mitochondrial apoptosis ↓
Campeau et al., 202241Empagliflozin (50 μM for 24 h)Human abdominal aortic endothelial cells (HAAECs) subjected to sustained GCX disruption● Inflammation caused by GCX disruption ↓
● Leukocyte (NB4 cells)—endothelium adhesion ↓
Cappetta et al., 202036Dapagliflozin (0,1 mg/kg/day for 6 weeks)67 rats were divided into:
(1) High-salt diet & dapagliflozin group (n = 25)
(2) High-salt diet & vehicle group (n = 30)
(3) Low-salt diet group (n = 12)
Sub-studies of 2–13 animals per group
● NHE1 activity ↓
● NF-kB, galectin-3 ↓
● VCAM-1, E-selectin, MCP-1, IL-6 ↓
Cooper et al., 201940Empagliflozin (50 μM for 24 h)Human abdominal aortic endothelial cells (HAAECs) exposed to TNF-α and wall shear stress● Glycocalyx integrity ↑
● Leukocyte (NB4 cells)—endothelium adhesion ↓
El-Daly et al., 201829Empagliflozin (25 nM up to 10 μM for 24 h)
Dapagliflozin (25 nM up to 10 μM for 24 h)
Mice aortic rings incubated in high glucose medium● NO-dependent relaxation through activation of PAR2 ↑
● EGFR-kinase/Src/Rho-kinase and PKC ↓
● ↓ ROS
Ganbaatar et al., 202038Empagliflozin (20 mg/kg/day for 8 or 12 weeks)Mice were randomly divided into:
(1) Control group
(2) Diabetic mice on vehicle
(3) Diabetic mice on empagliflozin
Sub-studies of 6–14 animals per group
● NOX-2, p22phox, MCP-1, ICAM-1, VCAM-1 ↓
● Vasoconstrictive eicosanoids (PGE2/TXB2) ↓
● Macrophage accumulation ↓
Gaspari et al., 201739Dapagliflozin (1 mg/kg/day for 4 weeks)






Dapagliflozin (1 nM to 1 mM for 24 h)
Aortic sections of Apo E−/− mice, which were divided into:
(1) Normal fat diet & vehicle
(2) High fat diet & vehicle
(3) Normal fat diet & dapagliflozin
(4) High fat diet & dapagliflozin
Sub-studies of 3–9 animals per group
HUVECs stimulated by TNF-α or hyperglycemia
● ICAM-1, VCAM ↓
● NF-kB ↓
He et al., 202230Dapagliflozin (1.0 mg/kg/day for 16 weeks)




Dapagliflozin (1 μΜ for 24 h)
Mice divided into:
(1) Normal diet group
(2) High-fat diet group
(3) High-fat diet & dapagliflozin group
Sub-studies of three animals per group
HUVECs stimulated with palmitic acid (PA)
● Angiogenesis ↑
● Cell viability ↑
● Mitochondrial homeostasis (SIRT1/PGC-1a pathway) ↑
Juni et al., 201925Empagliflozin (1 μM for 6 h)Human cardiac microvascular endothelial cells (CMECs) co-cultured with Cardiomyocytes (CMs) and exposed to TNF-α● Mitochondrial ROS ↓
● Cytoplasmic ROS ↓
● NO ↑
● Preserved CM contraction and relaxation
Juni et al., 202026Empagliflozin (1 μM for 6 h)Human cardiac microvascular endothelial cells (CMECs) co-cultured with Cardiomyocytes (CMs) and exposed to uraemic serum● Mitochondrial ROS ↓
● Cytoplasmic ROS ↓
● NO ↑
● Preserved CM contraction and relaxation
● Mitochondrial fragmentation ↓
Lee et al., 201819Dapagliflozin (60 mg/kg diet; 0.006%, for 8 weeks)47 Mice divided into:
(1) Control mice with standard diet (n = 11)
(2) Control mice with standard diet & dapagliflozin
(n = 12)
(3) Diabetic mice with standard diet (n = 12)
(4) Diabetic mice with standard diet & dapagliflozin
(n = 12)
Sub-studies of 3–12 animals per group
● Endothelium-dependent and -independent relaxation ↑
● PWV ↓
● IL-1β, IL-6, IL-10, IL-17, MCP-1, CCL-5 ↓
Li et al., 202133Empagliflozin (1 μΜ for 24 h)
Dapagliflozin (1 μΜ for 24 h)
Human coronary artery endothelial cells (HCAECs) subjected to cyclic stretch● NHE-1 and NOX-mediated ROS ↓
● Cell permeability ↓
● VE-cadherin ↑
Ma et al., 202224Dapagliflozin (40 mg/kg/day for 7 days)




Dapagliflozin (10 μΜ for 24 h)
Mice divided into:
(1) Sham operation group
(2) IRI group
(3) IRI & dapagliflozin group
Sub-studies of 6 or 10 animals per group
Human coronary artery endothelial cells (HCAECs) subjected to H/R injury
● eNOS ↑
● ICAM-1, IL-6, MCP1, TNF-α ↓
● VEGF ↑
● Mitochondrial fission ↓
● Cell survival ↑
● F-actin, Angiogenesis ↑
Mone et al., 202231Empagliflozin (1 μM for 24 h)Human umbilical vein endothelial cells (HUVECs) stimulated with high glucose levels
Human brain microvascular endothelial cells (hBMECs) stimulated with high glucose levels or H2O2
● Mitochondrial Ca2+ overload ↓
● Mitochondrial ROS ↓
● Endothelial leakage ↓ (claudin-5 and occluding mRNA ↑)
● Cell viability ↑
Nakao et al., 202112Empagliflozin (0,03% w/w)Murine model of left ventricular pressure overload generated by transverse aortic constriction (TAC)
Mice were divided into:
(1) TAC & empagliflozin group
(2) TAC—control group
(3) Sham operation & empagliflozin group
(4) Sham operation—control group
Sub-studies of 3–31 animals per group
● NO ↑
● eNOS ↑
Nikolaou et al., 2019 A35Empagliflozin (10 mg/kg/day for 6 weeks)IRI murine model
130 mice were divided into cohorts, with each cohort consisting of 2–4 groups with 5–8 animals each
● VEGF ↑
● Superoxide dismutase 2 ↑
● Cell survival ↑
Nikolaou et al., 2019 B35Empagliflozin (500 nM for 24 h)Human cardiac microvascular endothelial cells (CMECs) subjected to H/R injury● ROS ↓
● Cell survival ↑
Oelze et al., 201418Empagliflozin (10 and 30 mg/kg/day for 7 weeks)48 mice divided into:
(1) Untreated controls
(2) Diabetic mice & placebo
(3) Diabetic mice & low dose empagliflozin
(4) Diabetic mice & high dose empagliflozin
Three sub-studies were performed with 3,4, and 5 animals per group
● NO-dependent vasodilation ↑
● eNOSSer1177 phosphorylation ↑
● ROS, NOX-1, NOX-2, HO-1 ↓
● CCL-2, CD68, IL-6, IFN-γ, TNF-α, ICAM-1 ↓
Park et al., 202016Empagliflozin (30 mg/kg/day for 6 weeks)40 mice were equally divided into:
(1) Lean control
(2) Lean control & empagliflozin
(3) Obese, diabetic, and hypertensive mice with HF
(4) Obese, diabetic, and hypertensive mice with HF &
empagliflozin
Sub-studies of 3–10 animals per group
● NO-mediated relaxation ↑
● COX-derived prostanoids ↓
● eNOS ↑
● senescence markers (p53,p21,p16), tissue factor, VCAM-1 ↓
Soares et al., 20229Empagliflozin (14 mg/kg/day for 6 weeks)Aged mice with confirmed vascular dysfunction were divided into:
(1) Empagliflozin group (n = 29)
(2) Control group (n = 29)
Sub-studies of 5–10 animals per group
● eNOS ↑
● ROS, MDA ↓
● F-actin, P-cofilin, arterial stiffness ↓
Steven et al., 201717Empagliflozin (10 and 30 mg/kg/day for 6 weeks)





Empagliflozin (1 or 10 μM for 6 days)
35 diabetic ZDF rats and 16 lean controls divided into:
(1) Lean controls
(2) Diabetic rats
(3) Diabetic rats on low dose empagliflozin
(4) Diabetic rats on high dose empagliflozin
Sub-studies of 4–16 animals per group
Human umbilical vein endothelial cells (HUVECs) exposed to Ach after cultured in high glucose medium
● NO-dependent vasodilation ↑
● eNOSThr495 phosphorylation ↓
● ROS ↓
● ECs viability ↑
● IFN-γ, COX2, NOS-2, P-selectin, ICAM-1 ↓
Uthman et al., 201922Empagliflozin (1 μM for 4–24 h)

Dapagliflozin (1 μM for 4–24 h)
Human coronary artery endothelial cells (HCAECs) exposed to TNF-α
Human umbilical vein endothelial cells (HUVECs) exposed to TNF-α
● NO ↑
● ROS ↓
Zhou et al., 201821Empagliflozin (10 mg/kg/day for 20 weeks)Streptozotocin (STZ)-induced diabetic mice were divided into:
(1) Control group (n = 6)
(2) Diabetic group (n = 6)
(3) Diabetic & empagliflozin group (n = 6)
Their cardiac microvascular endothelial cells (CMECs) were also used
● Endothelium-dependent relaxation ↑
● eNOSSer1177 phosphorylation ↑
● Cytoplasmic ROS ↓
● Mitochondrial ROS ↓
● Mitochondrial fission ↓
● F-actin, Angiogenesis ↑
● ICAM-1, VCAM-1 ↓
Zou et al., 202223Empagliflozin (10 mg/kg/day for 7 days)




Empagliflozin (10 µM for 12 h)
Mice were divided into:
(1) Sham operation group
(2) I/R group
(3) I/R & empagliflozin group
Sub-studies of 6 or 10 animals per group
Human coronary artery endothelial cells (HCAECs) subjected to IRI
● eNOS ↑
● Mitochondrial fission ↓
● Cytoplasmic ROS ↓
● Mitochondrial ROS ↓
● VE-cadherin ↑
● ICAM-1 ↓

Abbreviations: ADMA ratio, asymmetric dimethylarginine ratio; Apo E, apolipoprotein E; CCL-5, chemokine (C-C motif) ligand-5; CD68, cluster of differentiation 68; CFVR, coronary flow velocity reserve; cGMP, cyclic guanosine monophosphate; CMECs, cardiac microvascular endothelial cells; CMs, cardiomyocytes; COX, cyclooxygenase; COX-2 = cyclooxygenase-2; EGFR, epidermal growth factor receptor; eNOS, endothelial nitric oxide synthase; GCX, glycocalyx; HAAECs, human abdominal aortic endothelial cells; hBMECs, human brain microvascular endothelial cells; HCAECs, human coronary artery endothelial cells; HO-1, heme oxygenase-1; H/R injury, hypoxia/reoxygenation injury; HUVECs, human umbilical vein endothelial cells; ICAM-1, intercellular adhesion molecule-1; IFN-γ, interferon-γ; IL-1β, interleukin-1β; IL-6, interleukin-6; IL-10, interleukin-10; IL-17, interleukin-17; IRI, ischemia/reperfusion injury; MCP-1, monocyte chemotactic protein-1; MDA, malondialdehyde; MMP-2, matrix metalloproteinase-2; MMP-9, matrix metalloproteinase-9; NF-kB, nuclear factor-kB; NHE1, Na+/H+ exchanger 1; NO, nitric oxide; NOS-2, inducible NO synthase-2; NOX-1, NADPH oxidase-1; NOX-2, NADPH oxidase-2; Ob, obese; p22phox, 22 kilodalton phagocyte oxidase; PAR2, proteinase-activated receptor 2; PGC-1a, peroxisome proliferator-activated receptor-gamma coactivator -1a; PGE2, prostaglandin E2; PKC, protein kinase C; PKG, protein kinase G; PWV, pulse wave velocity; Rho, Ras homologous; ROS, reactive oxygen species; SIRT1, silent information regulator 1; Src, sarcoma virus tyrosine kinase; TNF-α ,tumor necrosis factor-α; TXB2, thromboxane B2; VCAM-1, vascular cell adhesion molecule-1; VE-cadherin, vascular endothelial-cadherin; VEGF, vascular endothelial growth factor; VSMCs, vascular smooth muscle cells; ZSF1, Zucker diabetic fatty/spontaneously hypertensive heart failure F1 hybrid; ZDF rats, Zucker Diabetic fatty rats.

The role of NO

Nitric oxide (NO) improves endothelial function through several intracellular pathways, ultimately leading to vasorelaxation, endothelial regeneration, inhibition of leukocyte chemotaxis, and platelet adhesion.1 In specific, arginine is oxidized into citrulline and NO, via the activation of the endothelial nitric oxide synthase (eNOS). Protein kinase B, otherwise known as AKT, upregulates eNOS activity,12 whereas asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of NOs, blocking the formation of NO from arginine directly.1

In vivo studies

Adingupu et al.1 studied the effect of empagliflozin on coronary microcirculation and NO pathway, assessing the coronary flow velocity reserve (CFVR) and the L-arginine/ADMA ratio. After 10 weeks of treating prediabetic ob/ob−/− mice with empagliflozin, the untreated group had lower CFVR compared with lean controls and the treated group. The L-arginine/ADMA ratio was also found to be higher in the treated mice, indicating increased NO bioavailability. The authors speculated that the increase in L-arginine could be attributed to increased kidney synthesis.

Interestingly, Santos-Gallego et al.13 researched on an HF pig model and found that empagliflozin activates eNOS, increases NO availability, raises cGMP, enhances protein kinase G (PKG) and phosphorylates titin, hence, contributing to improvement in diastolic dysfunction and to benefits on HFpEF both on HF hospitalizations7 and on quality of life.14 The results of their research come in line with a paradigm proposed for HFpEF,15 where NO impairment causes diastolic dysfunction via the eNOS/NO/cGMP/PKG/hypophosphorylation of titin pathway.

Another research team,16 using a murine model of metabolic syndrome with HFpEF, showed that empagliflozin normalizes the NO-mediated endothelium-dependent relaxation and prevents the endothelium-dependent contractile response to acetylcholine (ACh), probably through averting the formation of cyclooxygenase-derived contractile prostanoids. Also, empagliflozin seems to maintain NO bioavailability through decreasing the levels of the senescence markers p53, p21, and p16.16

Furthermore, Steven et al.17 and Oelze et al.18 report a partial prevention of endothelial dysfunction, defined as impaired ACh-dependent relaxation, in diabetic rats after empagliflozin treatment. Empagliflozin reduced the upregulated cGMP-dependent kinase (cGK-I), which is known to critically modulate NO-dependent vasodilation, and increased the phosphorylation of VASP, a substrate protein of cGK-I. It also increased dihydrofolate reductase, an enzyme that converts BH2 to BH4.

Similarly, dapagliflozin has also been found to restore NO bioavailability. A study upon diabetic mice19 showed that treatment with dapagliflozin significantly improved the endothelium-dependent dilatation. This effect was observed using several doses of ACh with the total area under the curve being more than doubled.

Regarding eNOS signalling, some studies provide evidence supporting that SGLT2 inhibitors restore NO bioavailability by interfering with eNOS. In murine models9,16,20,21empagliflozin restored the downregulated cellular levels of eNOS, as well as the upregulated ET-1 expression.20 A study conducted upon a murine model of left ventricular pressure overload12 specifically showed that empagliflozin activates the AKT/eNOS/NO pathway, which suppresses endothelial apoptosis, maintains capillarization and improves systolic dysfunction. Characteristically, empagliflozin increased citrulline levels in cardiac tissue and reduced levels of arginine, indicating enhanced metabolism from arginine to citrulline and NO. Their results also suggest that empagliflozin inhibits catecholamine-induced downregulation of the AKT/eNOS/NO pathway by increasing hydroxybutyrate, which could suppress catecholamine-induced increase of reactive oxygen species (ROS). Steven et al.17 also reports that the inhibitory phosphorylation of eNOS at Thr495 was diminished by treatment with empagliflozin in diabetic mice; however, the attenuation of endothelial dysfunction by empagliflozin is potentially a result of improved NO/cGMP signalling and prevention of oxidative damage in this pathway.

Nevertheless, not all studies support the alteration of eNOS signalling by SGLT2 inhibitors. Uthman et al.22 showed that empagliflozin and dapagliflozin restore NO bioavailability after TNF-α stimulation in human coronary artery endothelial cells (HCAECs) in a way that does not involve eNOS phosphorylation, expression or signaling. They found no involvement of eNOSser1177 or eNOSThr495 phosphorylation, as well as no changes in caveolin-1 (Cav-1) and Cav-1/eNOS correlation. Cav-1 is known to have an inhibitory effect on eNOS, contributes to the compartmentalization of the eNOS in caveolae and plays an important role in endothelial signaling pathways in diseases involving the vasculature.

In vitro studies

Some in vitro studies have also investigated the effect of SGLT2 inhibitors on the NO-dependent endothelial function and have displayed promising results. In HCAECs following ischemia/reperfusion (I/R) injury, empagliflozin23and dapagliflozin24 restored the downregulated cellular levels of eNOS, in addition to the upregulated ET-1 expression,24 a finding consistent with the aforementioned in vivo studies.9,16,20,21

Furthermore, regarding myocardial contractility, two studies conducted by Juni et al.,25,26 demonstrated that human cardiac microvascular endothelial cells (CMECs) improve CM contraction and relaxation, with empagliflozin not only facilitating the endothelium to positively modulate CM function, but also maintaining it when CMECs were exposed to pro-inflammatory cytokines or uremic serum from patients with CKD. This beneficial effect of empagliflozin was found to be mediated by the endothelial-derived NO, whose bioavailability was increased due to reduced mitochondrial ROS generation. Interestingly, they also observed that the CMEC-derived NO was a more predominant contributor to CM contractile performance, rather than the endogenous NO. The cardiac microvascular endothelial enhancement of CM function that empagliflozin managed to restore implies a novel mechanism by which empagliflozin could restore CMEC function in a variety of heart failure types with MVD, including HFpEF.

The role of mitochondrial function

Mitochondria are involved in endothelial mobilization, senescence, growth, and proliferation as well as in censoring environmental stress and cellular adaptations. Their damage, therefore, contributes to endothelial dysfunction and cardiac microvascular I/R injury.20,23 They are also a primary source of ROS, thus their dysfunction further contributes to oxidative stress.9 Moreover, the superoxides derived from mitochondria reduce NO production, while also promoting apoptosis by activating caspases.23 SGLT2 inhibitors, therefore, could improve endothelial function through interfering with several of the above mechanisms.

In vivo studies

A variety of studies provide evidence supporting that SGLT2 inhibitors exert their cardioprotective effects through interfering with mitochondrial homeostasis. More specifically, data indicate that SGLT2 inhibitors prevent mitochondrial fission,20,21 a process with the potential to damage mitochondrial DNA during mitosis and induce endothelial apoptosis, which in turn causes microvessel swelling and vascular cell collapse.20 Several pathways through which SGLT2 inhibitors prevent mitochondrial fission have been mentioned, such as reduction of Drp1 phosphorylation.21 The phosphorylation of Drp1 enables its shift onto mitochondria and is promoted by upstream signalling molecules, including Rho-associated coiled-coil containing protein kinase (ROCK1) and adenosine monophosphate-activated protein kinase (AMPK). Empagliflozin inhibits mitochondrial fission in an AMPK-dependent manner.21

Moreover, Cai et al.20 found that empagliflozin suppresses mitochondrial fission by activating mitophagy on the CMECs of a murine I/R injury model, by specifically activating the FUN14 domain-containing 1 (FUNDC1)-dependent mitophagy through the AMPKα1/Unc-51-like autophagy activating kinase 1 (ULK1) pathway. Notably, mitophagy is a protective mechanism that repairs poorly structured mitochondria through normalization of mitochondrial fission and fusion, thus alleviating mitochondrial damage and preserving microvascular structure and function.20

Aside from mitochondrial fission and fusion, empagliflozin has also been found to stabilize the mitochondrial membrane potential (MMP) and reduce the mitochondrial permeability transition pore opening rate in a murine model of I/R injury, ultimately reducing caspase-9-mediated mitochondrial apoptosis.20 Moreover, SGLT2 inhibitors improve the energetics of the muscle and ATP generation by the mitochondria, as demonstrated with empagliflozin in pigs27 and with dapagliflozin in humans.28

In vitro studies

In line with the in vivo studies, the beneficial effects of SGLT2 inhibitors on mitochondrial function have also been shown in vitro. Zou et al.23 demonstrated that empagliflozin inhibits mitochondrial fission through dephosphorylation of the mitochondrial fission 1 protein (Fis1) in I/R injured HCAECs. The SGLT2 inhibitor specifically protected the microvasculature by inhibiting the DNA-PKcs/Fis1/mitochondrial fission pathway, due to its anti-oxidative effects, resulting to a repressed mitochondrial fission and an improved mitochondrial metabolism. Dapagliflozin similarly restores the balance between mitochondrial fission and fusion. In HCAECs exposed to hypoxia-reoxygenation,24dapagliflozin normalized mitochondrial morphology, by inhibiting the mitochondrial fission factors Drp1 and Mff, while upregulating the mitochondrial fusion regulators Opa1 and Mfn2.

SGLT2 inhibitors also have the ability to improve mitochondrial function in ways unaffiliated with their fission and fusion. In a murine model of hyperglycemia,29empagliflozin and dapagliflozin managed to increase the basal oxygen consumption, promoting a more efficient mitochondrial metabolism, and decrease the proton leak, indicative of better mitochondrial integrity. Additionally, He et al.30 established that dapagliflozin could increase a previously reduced MMP and prevent mitochondrial structural injury caused by palmitic acid (PA) in human umbilical vein endothelial cells (HUVECs) via the activation of the SIRT1/PGC-1a signaling pathway. Finally, in a recent study regarding HUVECs stimulated with high glucose levels, empagliflozin attenuated the mitochondrial Ca2+ overload and subsequently reduced the enhanced mitochondrial ROS production.31

The role of anti-oxidative effect

Moderate concentration of ROS play an important role in maintaining the proliferation and survival of endothelial cells, as well as in regulating vascular function and endothelial integrity.32,33 Excessive levels, though, lead to deleterious vascular effects, such as increased endothelial permeability.32,33 Mounting evidence reveals that empagliflozin displays anti-oxidative properties, thus protecting the endothelium, by inhibiting both intracellular and mitochondrial ROS (mtROS) production.20,21,23 This anti-oxidative effect has been linked to elevated levels of anti-oxidative molecules such as glutathione (GSH), superoxide dismutase (SOD), and glutathione peroxidase (GPX).20

In vivo studies

Empagliflozin treatment has been reported to reduce plasma levels of malondialdehyde (MDA), a marker of lipid peroxidation, as well as to up- or downregulate key proteins and pathways involved in ROS synthesis.9 Steven et al.17 and Oelze et al.18 also report a reduction in ROS formation after empagliflozin treatment in diabetic mice, as empagliflozin normalized the decreased level of the redox-sensitive enzyme mitochondrial aldehyde dehydrogenase (ALDH-2) in heart mitochondria, whereas it reduced the expression of 3-nitrotyrosine (3NT) or 4-hydroxynonenal (HNE)-positive proteins, as well as other parameters indicative of the oxidative stress extent.

Regarding the role of angiotensin II and SGLT2 inhibition, Bruckert et al.34 evaluated the effect of empagliflozin on a murine model of angiotensin II-induced hypertension, reporting that empagliflozin preserves the diastolic function of the heart. This effect is mediated by averting endothelial cell activation in both the macro and coronary microcirculation and preventing the activation of the deleterious Ang II/NADPH oxidases/SGLT1- and 2 pro-oxidant pathways in endothelial cells. Empagliflozin specifically prevented the stimulatory effect of Ang II on the mRNA up-regulation of several deleterious NADPH oxidase subunits, such as p47phox, p22phox, and NOX1, as well as the downregulation of the vasoprotective NOX4.

Moreover, a study conducted by Nikolaou et al.35 in non-diabetic mice showed that chronic (6 weeks) administration of empagliflozin reduces myocardial infarct size and attenuates biomarkers of cardiac oxidative stress after I/R injury. It specifically lessened MDA and protein carbonyls (PCs) in the myocardium, while it upregulated the mRNA levels of superoxide dismutase 2 (Sod2), a regulator of oxidative stress. Regarding the underlying mechanism, chronic empagliflozin administration activated the STAT-3 pathway and improved the formation of Y(705)STAT-3 dimers, which regulate many anti-oxidant and anti-apoptotic genes. Notably, acute administration of empagliflozin did not activate STAT-3, while empagliflozin was also found to inhibit ROS production in the absence of STAT-3 activation.

Finally, a recent study by Santos-Gallego et al.58 showed that empagliflozin reduces microvascular obstruction, decreases MI size, ameliorates ischemia/reperfusion injury, and reduces oxidative stress in a porcine model of percutaneous I/R. Therefore, this study further confirms the positive impact of empagliflozin in the reduction of oxidative stress, which could possibly be a result of the slight ketonemia induced by SGLT2 inhibitors and the alteration of the metabolic substrate for the ischemic myocardium.

In vitro studies

As aforementioned,25,26empagliflozin reduces ROS production in human CMECs exposed to pro-inflammatory cytokines and uremic acid, contributing to an endothelial NO-mediated improvement of contraction and relaxation of cardiomyocytes. Moreover, in a human model of TNF-α-stimulated HCAECs,22empagliflozin and dapagliflozin similarly reduced the increased ROS levels. Mone et al.,31 also studying human brain microvascular endothelial cells (hBMECs) treated with empagliflozin and then incubated with increasing concentrations of H2O2, showed a significant improvement of cell viability after 5 h, in the group pretreated with empagliflozin.

Recently, Li et al.33 revealed that SGLT2 inhibitors prevent ROS production in a HCAEC model of enhanced cyclic stretch, thus displaying their ability to alleviate oxidative stress specifically caused by mechanical forces. The researchers used Na+/H + Exchanger 1 (NHE1) and NADPH oxidase inhibitors, which blocked ROS production induced by stretch. Notably, no further reduction in ROS production was noticed when empagliflozin was co-administered, suggesting that the anti-oxidative properties of empagliflozin are mediated by NHE1 and NOXs. NHE1 inhibition is also known to block the production of inflammatory molecules and protect the endothelium by lowering the accumulation of Na+ and Ca2+.36 However, the role of NHE1 inhibition as a cardioprotective effect of SGLT2 inhibitors is still under investigation, as some contrasting data demonstrate an unrelated to NHE1 effect of empagliflozin.26

The role of inflammation

Inflammatory cytokines are one of the main stimuli of ventricular remodelling, contributing to the diastolic left ventricular dysfunction in HFpEF.37 SGLT2 inhibitors may exert their beneficial effects through suppressing microvascular inflammation.

In vivo studies

SGLT2 inhibitors have been well documented to attenuate endothelial dysfunction by suppressing inflammation. Ganbaatar et al.38 demonstrated that 8-week empagliflozin treatment in diabetic apolipoprotein E-deficient mice significantly ameliorated diabetes-induced endothelial dysfunction, as determined by the vascular response to ACh, and reduced vasoconstrictive eicosanoids such as PGE2 and TXB2, as well as inflammatory and adhesion molecules in aortic endothelial cells and in perivascular adipose tissue. Steven et al.17 also reported a reduction in the expression of inflammatory molecules such as interferon-γ (IFN-γ), cyclooxygenase-2 (COX2), and inducible NO synthase (NOS2) after SGLT2 inhibition.

Dapagliflozin, in a similar manner, is also reported to reduce several circulating inflammatory markers. In diabetic mice, dapagliflozin decreased the levels of MCP-1, CCL5, IL-1β, IL-6, and IL-17,19 while in euglycemic mice on a high-salt diet, it attenuated the upregulated NF-kB levels,36 in addition to myocardial galectin-3 (an NF-κB activator), MCP-1, IL-6, and E-selectin. This anti-inflammatory effect of dapagliflozin has also been reported in mice after cardiac I/R injury.24

Moreover, SGLT2 inhibitors may prevent the expression of the adhesive proteins ICAM-1 and VCAM-1, as shown by several studies upon murine models.16–18,20,21,23,24,34,36,38,39 They also reduce the expression of E-selectin36 and P-selectin.17 These observations bear significant clinical relevance, as the upregulation of adhesion molecule expression indicates endothelial dysfunction and predisposes to atherosclerosis development.39

In vitro studies

Consistent with the results of the aforementioned in vivo studies in regard to circulating inflammatory markers, Gaspari et al.39 reports a significant suppression of TNF-α-mediated NF-kB expression in HUVECs stimulated with TNF-α.

Interestingly, SGLT2 inhibitors further exert their anti-inflammatory properties by protecting the endothelial integrity. VE-cadherins, claudin-5, and occludin are junctional proteins expressed on endothelial cells that play a vital role in endothelial integrity and vascular permeability. Studies on HCAECs after I/R injury or stretch-induced endothelial barrier dysfunction33 showed that empagliflozin restores VE-cadherin expression by raising the sarcoma virus tyrosine kinase (Src) and the focal adhesion kinase (Fak), which regulate VE-cadherin activity.20,23Dapagliflozin has also been found to revert stretch-induced VE-cadherin degradation.33 In hBMECs stimulated with high glucose concentrations, empagliflozin restored the downregulated mRNA levels of claudin-5 and occludin.31

However, regarding adhesion molecule expression, the in vitro studies do not verify the results of the aforementioned in vivo models. Expression of ICAM-1 and VCAM-1 remained unchanged in human CMECs,25 HCAECs,22 and HUVECs,22 all exposed to TNF-α, after treatment with an SGLT2 inhibitor. Furthermore, Cooper et al.40 also demonstrated that empagliflozin does not significantly alter VCAM‑1 and ICAM‑1 expression on human abdominal aortic endothelial cells (HAAECs) under static and flow conditions. Collectively though, data suggest that SGLT2 inhibitors demonstrate a protective role towards the endothelial integrity and barrier function; an important effect, as it lessens the likelihood of microthrombus formation.20,21,23

Another reported feature of empagliflozin is its ability to restore the integrity of the endothelial glycocalyx (GCX). The endothelial GCX is a dynamic structure that consists of proteoglycans, glycosaminoglycans, and glycolipids forming a functional layer lining the cell surface, which protects the microvascular environment. Loss of its integrity, caused for example by exposure to high levels of glucose or ROS, increases the atherogenic potential. Indeed, deterioration of endothelial GCX has been associated with endothelial dysfunction and lipid accumulation in the subendothelial space.40 Cooper et al.40 demonstrated that empagliflozin restores the level of heparan sulfate (HS) intensity, the most abundant component of the GCX, under static and flow culture conditions in heparinase III-treated HAAECs. Campeau et al.41 also showed the impact of GCX disruption on the induction of endoplasmic reticulum (ER) stress and the ability of empagliflozin to resolve the associated inflammatory response of endothelial cells through the transcriptional downregulation of key unfolded protein response genes. Their results indicated that empagliflozin attenuates EC inflammation independently of HS-mediated functions. Last but not least, of interest is the ERTU-SODIUM study, which is currently investigating the effects of SGLT2 inhibitors on glycocalyx, proteoglycans, and glycosaminoglycans in vivo in HFrEF patients.42

The role of endothelial cell viability and angiogenesis

A variety of studies have investigated the impact of SGLT2 inhibitors on angiogenesis and EC's viability both in vitro and in vivo. It is well known that diabetes alters the dynamics of angiogenesis; therefore, treatment with SGLT2 inhibitors, on top of glycemic control, could attenuate the hyperglycemia-induced effects on the vasculature.32

In vivo studies

Zhou et al.21 demonstrated upon a diabetic murine model that empagliflozin increases CMEC survival and delays vascular fibrosis, thus alleviating vascular degeneration, enriching microvascular density, and increasing cardiac perfusion. More specifically, empagliflozin preserved the angiogenic capacity of isolated CMECs through stabilization of F-actin, promotion of a well-arranged F-actin cytoskeleton and increased F-actin levels. Data are still conflicting; however, as Soares et al.9 report that empagliflozin decreased F-actin and P-cofilin content in the mesenteric arteries of a murine model. The reduction in F-actin and P-cofilin was associated with reduced arterial stiffness, which was also ameliorated by empagliflozin.

In vitro studies

In agreement with Zhou et al.,21 Ma et al.24 similarly demonstrated the ability of dapagliflozin to sustain cytoskeletal integrity and preserve F-actin expression in vitro. Using HCAECs subjected to hypoxia-reoxygenation (H/R), they managed to demonstrate that dapagliflozin prevents H/R-mediated Ca2+/calmodulin-dependent protein kinase II (CaMKII) phosphorylation, which in turn leads to F-actin polymerization. This CaMKII inactivation is promoted by repression in calcium overload, which was found to be restricted by preventing XO-induced SERCA2 oxidation and inactivation. Interestingly, the inhibition of this pathway further alleviates oxidative stress, as the activation of XO in addition to cytoplasmic and mitochondrial calcium overload produce excess ROS, leading to reduced bioavailability of NO, microvascular contraction, and apoptosis.

Moreover, Nikolaou et al.35 found that empagliflozin increases human microvascular endothelial cells viability in normoxia and hypoxia, with the protective effects being STAT-3 dependent under hypoxic conditions. They also demonstrated that empagliflozin upregulates VEGF levels, thus potentiating the survival and proliferation of endothelial cells. Indeed, STAT-3 and VEGF pathways have already been reported to have a mutual relationship. In support of these findings, dapagliflozin also restored VEGF expression in HCAECs after exposure to hypoxia-reoxygenation and counteracted the extent of apoptosis.24 However, the mechanism by which SGLT2 inhibitors increase endothelial cell proliferation prior to the ischemia is still unclear and could also rely on other cells such as smooth muscle cells and fibroblasts.35

Last but not least, dapagliflozin reversed the apoptosis in addition to the inhibition of angiogenesis caused by PA in HUVECs.30 PA was used to mimic a high-fat environment. The expression of apoptosis-related proteins, including BAX, Bcl-2, and cleaved caspase 3, was also reduced by dapagliflozin.

Clinical trials: data on the effect of SGLT2 inhibitors in human studies

Even though the complex and interconnected pathophysiological mechanisms concerning the effect of SGLT2 inhibitors on the endothelium are not yet fully understood, evidence indicates their key role in the improvement of the endothelial function among a broad spectrum of patients. In this part of the review, we present trials assessing SGLT2 inhibition in endothelial cell function and microcirculation (Table 2). An attempt has been made to differentiate between the trials assessing endothelial function and microcirculation according to the used indexes. It should be noted, however, that endothelial and microvascular function are two concepts that overlap and share common underlying mechanisms, as microcirculation is regulated by both endothelial-dependent and independent mechanisms. Flow-mediated dilation (FMD) and reactive hyperaemia index (RHI) have been classified as endothelial function indexes, as the vasodilatory response to shear stress that they assess is thought to be NO-mediated. On the contrary, myocardial flow reserve (MFR) is assessed as a measure of microcirculatory function, mainly reflecting the myocyte tone of vessels, which is regulated in an endothelium-independent manner. Lastly, CFVR, is also considered a measure of microcirculatory function, as the stressors used to estimate it cause vasodilation through non-endothelial dependent mechanisms.

Table 2

Clinical studies regarding the effect of SGLT2 inhibitors in endothelial function and microcirculation

ReferenceDrug/DosagePatientsMeasured indexOutcome
Irace et al., 201943Empagliflozin (10 mg/day, up-titrated at 25 mg/day in 13 patients)
Sitagliptin (100 mg/day)
Liraglutide (0.6 mg/day, up-titrated at 1.2 mg/day or even at 1.8 mg/day)
(1) 20 T2DM patients treated with empagliflozin
(2) 15 T2DM patients treated with incretin
(3) 25 nondiabetic healthy subjects as controls
FMD• Significant difference of peak wall shear stress and FMD in the empagliflozin group compared to control (P < 0.05 and P = 0.04, respectively)
• ↑ Brachial artery ultrasound-based FMD after 1 month of empagliflozin treatment (4.8 ± 4.5% vs. 7.7 ± 5.3%, P = 0.03)
• ↑ Brachial artery ultrasound-based FMD after 3 months of empagliflozin treatment (4.8 ± 4.5% vs. 8.5 ± 5.6%, P = 0.03)
• ↑ Peak brachial artery wall shear stress after 1 month of empagliflozin treatment (61±20 vs. 65 ± 23 dynes/cm2, P = 0.04)
• ↑ Peak brachial artery wall shear stress after 3 months of empagliflozin treatment (61±20 vs. 68 ± 25 dynes/cm2, P = 0.04)
• No significant changes in the incretin-based therapy group
Lunder et al., 201811Empagliflozin (25 mg/day)
Metformin (2000 mg/day)
40 T1DM patients randomized into four groups:
(1) Empagliflozin,
(2) Metformin,
(3) Empagliflozin/metformin and
(4) placebo
FMD
RHI
• No comparison was made between the treatment groups and the control.
• Within each treatment group, brachial artery ultrasound-based FMD and RHI were significantly improved after 12 weeks of treatment.
Shigiyama et al., 201747Dapagliflozin (5 mg/day) & Metformin (750 mg/day)
Metformin (1500 mg/day)
80 T2DM patients were randomized to receive either dapagliflozin on top of metformin or metformin alone for 16 weeks.FMD• FMD tended to improve only in the dapagliflozin group, but ΔFMD was comparable between the two groups.
• ↑ FMD in dapagliflozin group compared with metformin group in a subgroup analysis of patients with HbA1c ≥ 7.0% (ΔFMD = 1.05 ± 2.59 and −0.94 ± 2.39, respectively, P = 0.041).
• ↓ 8-OHdG in the dapagliflozin group compared with the metformin group (P < 0.001).
Solini et al., 201748Dapagliflozin (10 mg/day)
Hydrochlorothiazide (12.5 mg/day)
(1) 16 T2DM patients treated with dapagliflozin for two days
(2) 10 T2DM patients treated with hydrochlorothiazide for two days as controls
FMD• ↑ FMD after 2 days of treatment with dapagliflozin (2.8 ± 2.2 to 4.0 ± 2.1%, P < 0.05).
• No significant change in FMD within the control group.
Solini et al., 201949Dapagliflozin (10 mg/day)
Hydrochlorothiazide (12.5 mg/day)
40 T2DM patients with hypertension were equally randomized to either dapagliflozin or hydrochlorothiazide for 4 weeks.FMD• No significant change in FMD in any group.
• The miRNAs linked to endothelial dysfunction (miR-200b and miR-27b) were equally increased in both groups [in the dapagliflozin group: 0.379 (0.580) to 2.802 (4.179) and 0.072 (0.050) to 0.172 (0.209), respectively].
Sposito et al., 202145Dapagliflozin (10 mg/day)
Glibenclamide (5 mg/day)
98 T2DM patients with atherosclerotic disease were randomized 1:1 to 12 weeks of therapy with dapagliflozin or glibenclamide in addition to metformin.FMD• ↑ 3.3% median increase in rest FMD at 1 min in the dapagliflozin group (P = 0.0001).
• ↓ 1.2% median decrease in rest FMD at 1 min in the glibenclamide group (P = 0.0001).
• FMD after I/R slightly increased (+2.1%) in the dapagliflozin group and decreased (−0.4%) in the glibenclamide group, but the changes did not reach statistical significance.
• Dapagliflozin—in contrast to glibenclamide—improved the paradoxical arterial constriction observed during FMD.
• Significant difference between the two groups regarding the end diastolic velocity, peak systolic velocity, and resistive indices measured at 1 and 5 min.
• ↑ 10% in the 1-min nitrite level in the dapagliflozin group vs. ↓ 4% in the glibenclamide group.
Zainordin et al., 201946Dapagliflozin (10 mg/day)81 T2DM patients with established ischemic heart disease were randomized using stratified randomization to 12 weeks of therapy with dapagliflozin or placebo in addition to diet, exercise, insulin, and/or metformin.FMD• No significant difference in FMD after 12 weeks, neither between the dapagliflozin and the placebo group nor within each group.
• ↑ NMD in the dapagliflozin group compared to placebo after 12 weeks of treatment (19.64 ± 9.71% vs. 15.11 ± 8.44%, P = 0.038).
• ↓ ICAM-1 within the dapagliflozin group after 12 weeks of treatment (−83.9 ± 205.9 ng/mL, P < 0.02).
Ilyas et al., 202151Dapagliflozin (10 mg/day)19 T2DM patients with HFrEF received dapagliflozin or placebo for 2 weeks and, after a 2-week washout period, they were crossed over for 2 more weeks of therapy.RHI• ↓ RHI after 2 weeks on dapagliflozin
Sugiyama et al., 201850Dapagliflozin (5 mg/day)54 patients with uncontrolled T2DM were nonrandomly allocated to either dapagliflozin or non-SGLT2 inhibitor medications for 6 months.RHI• Absolute and percentage changes in the LnRHI significantly greater in the dapagliflozin group when compared to control (P = 0.028).
• ↑ LnRHI after 6 months in the dapagliflozin group (0.450 ± 0.135 to 0.662 ± 0.230, P < 0.01). The LnRHI change in the non-SGLT2 inhibitor group was insignificant.
• The proportion of patients with an increase in LnRHI > 15% was 74.1% in the dapagliflozin group, but only 40.7% in the non-SGLT2 inhibitor group (P = 0.01).
• 4.156 (95% CI: 1.312–13.169) odds ratio in the univariate logistic regression analysis for the improvement of microvascular endothelial function among the dapagliflozin group (P = 0.015).
Tanaka et al., 201944Empagliflozin (10 mg/day)117 patients with T2DM and cardiovascular disease were equally randomized to either empagliflozin or placebo for 24 weeks.RHI• Absolute changes in RHI were −0.006 (SD 0.478) in the empagliflozin group and −0.025 (0.454) in the placebo group.
Suhrs et al., 202254Empagliflozin (25 mg/day)26 T2DM participants were randomized (1:1) to either sequence 1: Placebo for 12 weeks followed by a wash out period of 2 weeks and empagliflozin 25 mg for 12 weeks; or sequence 2: empagliflozin 25 mg for 12 weeks followed by a wash-out period of 2 weeks and placebo for 12 weeks.CFVR• No significant effect on CFVR, neither after empagliflozin treatment nor placebo
• Significant difference in hyperaemia CFV when the empagliflozin change was compared to the placebo change (−0.04 ± 0.15 vs. 0.05 ± 0.13, P = 0.044); however, the difference is small and can be regarded as a change finding.
Jürgens et al., 202152Empagliflozin (25 mg/day)90 T2DM patients were randomized to either empagliflozin or placebo for 13 weeks.MFR• No significant change in MFR
• No significant change in RPP adjusted MFR
• No significant difference between groups in rest flow (mL/g per min)
• No significant difference between groups in stress flow (mL/g per min)
• No significant change in reversible extent, TPD (%)
Lauritsen et al., 202153Empagliflozin (25 mg/day)13 T2DM patients were randomized (1:1) to receive either empagliflozin or placebo.MFR
MBF
• Empagliflozin reduced resting MBF compared to placebo (0.74 ± 0.10 vs 0.85 ± 0.10 mL/g/min, P < 0.01). This decrease in MBF remained significant even after being adjusted for RPP (1.01 vs. 1.06 mL/g/min, P = 0.04)
• No significant effect on stress MBF or MFR
Leccisotti et al., 202255Dapagliflozin (10 mg/day)16 patients with T2DM and stable CAD were equally randomized to receive either dapagliflozin or placebo for 4 weeks.MFR• ↑ MFR in the dapagliflozin group (2.56 ± 0.26 vs. 3.59 ± 0.35, P = 0.006), in contrast to placebo (2.34 ± 0.21 vs. 2.38 ± 0.24, P = 0.81). P for interaction = 0.001.
• ↑ MFR corrected for resting RPP in the dapagliflozin group (2.22 ± 0.25 vs. 3.23 ± 0.4, P = 0.008), in contrast to placebo. P for interaction = 0.019.
• ↓ Resting MBF in the dapagliflozin group, even after correction for resting RPP (1.15 ± 0.09 vs. 0.92 ± 0.10 mL/min/g, P = 0.005), compared with the placebo group (1.20 ± 0.10 vs. 1.18 ± 0.17 mL/min/g, P = 0.53). P for interaction = 0.045.
• Trend towards an increase in stress MBF in the dapagliflozin group (2.32 ± 0.15 vs. 2.64 ± 0.20 mL/min/g, P = 0.054), compared with placebo (2.47 ± 0.15 vs. 2.36 ± 0.16 mL/min/g, P = 0.55).
ReferenceDrug/DosagePatientsMeasured indexOutcome
Irace et al., 201943Empagliflozin (10 mg/day, up-titrated at 25 mg/day in 13 patients)
Sitagliptin (100 mg/day)
Liraglutide (0.6 mg/day, up-titrated at 1.2 mg/day or even at 1.8 mg/day)
(1) 20 T2DM patients treated with empagliflozin
(2) 15 T2DM patients treated with incretin
(3) 25 nondiabetic healthy subjects as controls
FMD• Significant difference of peak wall shear stress and FMD in the empagliflozin group compared to control (P < 0.05 and P = 0.04, respectively)
• ↑ Brachial artery ultrasound-based FMD after 1 month of empagliflozin treatment (4.8 ± 4.5% vs. 7.7 ± 5.3%, P = 0.03)
• ↑ Brachial artery ultrasound-based FMD after 3 months of empagliflozin treatment (4.8 ± 4.5% vs. 8.5 ± 5.6%, P = 0.03)
• ↑ Peak brachial artery wall shear stress after 1 month of empagliflozin treatment (61±20 vs. 65 ± 23 dynes/cm2, P = 0.04)
• ↑ Peak brachial artery wall shear stress after 3 months of empagliflozin treatment (61±20 vs. 68 ± 25 dynes/cm2, P = 0.04)
• No significant changes in the incretin-based therapy group
Lunder et al., 201811Empagliflozin (25 mg/day)
Metformin (2000 mg/day)
40 T1DM patients randomized into four groups:
(1) Empagliflozin,
(2) Metformin,
(3) Empagliflozin/metformin and
(4) placebo
FMD
RHI
• No comparison was made between the treatment groups and the control.
• Within each treatment group, brachial artery ultrasound-based FMD and RHI were significantly improved after 12 weeks of treatment.
Shigiyama et al., 201747Dapagliflozin (5 mg/day) & Metformin (750 mg/day)
Metformin (1500 mg/day)
80 T2DM patients were randomized to receive either dapagliflozin on top of metformin or metformin alone for 16 weeks.FMD• FMD tended to improve only in the dapagliflozin group, but ΔFMD was comparable between the two groups.
• ↑ FMD in dapagliflozin group compared with metformin group in a subgroup analysis of patients with HbA1c ≥ 7.0% (ΔFMD = 1.05 ± 2.59 and −0.94 ± 2.39, respectively, P = 0.041).
• ↓ 8-OHdG in the dapagliflozin group compared with the metformin group (P < 0.001).
Solini et al., 201748Dapagliflozin (10 mg/day)
Hydrochlorothiazide (12.5 mg/day)
(1) 16 T2DM patients treated with dapagliflozin for two days
(2) 10 T2DM patients treated with hydrochlorothiazide for two days as controls
FMD• ↑ FMD after 2 days of treatment with dapagliflozin (2.8 ± 2.2 to 4.0 ± 2.1%, P < 0.05).
• No significant change in FMD within the control group.
Solini et al., 201949Dapagliflozin (10 mg/day)
Hydrochlorothiazide (12.5 mg/day)
40 T2DM patients with hypertension were equally randomized to either dapagliflozin or hydrochlorothiazide for 4 weeks.FMD• No significant change in FMD in any group.
• The miRNAs linked to endothelial dysfunction (miR-200b and miR-27b) were equally increased in both groups [in the dapagliflozin group: 0.379 (0.580) to 2.802 (4.179) and 0.072 (0.050) to 0.172 (0.209), respectively].
Sposito et al., 202145Dapagliflozin (10 mg/day)
Glibenclamide (5 mg/day)
98 T2DM patients with atherosclerotic disease were randomized 1:1 to 12 weeks of therapy with dapagliflozin or glibenclamide in addition to metformin.FMD• ↑ 3.3% median increase in rest FMD at 1 min in the dapagliflozin group (P = 0.0001).
• ↓ 1.2% median decrease in rest FMD at 1 min in the glibenclamide group (P = 0.0001).
• FMD after I/R slightly increased (+2.1%) in the dapagliflozin group and decreased (−0.4%) in the glibenclamide group, but the changes did not reach statistical significance.
• Dapagliflozin—in contrast to glibenclamide—improved the paradoxical arterial constriction observed during FMD.
• Significant difference between the two groups regarding the end diastolic velocity, peak systolic velocity, and resistive indices measured at 1 and 5 min.
• ↑ 10% in the 1-min nitrite level in the dapagliflozin group vs. ↓ 4% in the glibenclamide group.
Zainordin et al., 201946Dapagliflozin (10 mg/day)81 T2DM patients with established ischemic heart disease were randomized using stratified randomization to 12 weeks of therapy with dapagliflozin or placebo in addition to diet, exercise, insulin, and/or metformin.FMD• No significant difference in FMD after 12 weeks, neither between the dapagliflozin and the placebo group nor within each group.
• ↑ NMD in the dapagliflozin group compared to placebo after 12 weeks of treatment (19.64 ± 9.71% vs. 15.11 ± 8.44%, P = 0.038).
• ↓ ICAM-1 within the dapagliflozin group after 12 weeks of treatment (−83.9 ± 205.9 ng/mL, P < 0.02).
Ilyas et al., 202151Dapagliflozin (10 mg/day)19 T2DM patients with HFrEF received dapagliflozin or placebo for 2 weeks and, after a 2-week washout period, they were crossed over for 2 more weeks of therapy.RHI• ↓ RHI after 2 weeks on dapagliflozin
Sugiyama et al., 201850Dapagliflozin (5 mg/day)54 patients with uncontrolled T2DM were nonrandomly allocated to either dapagliflozin or non-SGLT2 inhibitor medications for 6 months.RHI• Absolute and percentage changes in the LnRHI significantly greater in the dapagliflozin group when compared to control (P = 0.028).
• ↑ LnRHI after 6 months in the dapagliflozin group (0.450 ± 0.135 to 0.662 ± 0.230, P < 0.01). The LnRHI change in the non-SGLT2 inhibitor group was insignificant.
• The proportion of patients with an increase in LnRHI > 15% was 74.1% in the dapagliflozin group, but only 40.7% in the non-SGLT2 inhibitor group (P = 0.01).
• 4.156 (95% CI: 1.312–13.169) odds ratio in the univariate logistic regression analysis for the improvement of microvascular endothelial function among the dapagliflozin group (P = 0.015).
Tanaka et al., 201944Empagliflozin (10 mg/day)117 patients with T2DM and cardiovascular disease were equally randomized to either empagliflozin or placebo for 24 weeks.RHI• Absolute changes in RHI were −0.006 (SD 0.478) in the empagliflozin group and −0.025 (0.454) in the placebo group.
Suhrs et al., 202254Empagliflozin (25 mg/day)26 T2DM participants were randomized (1:1) to either sequence 1: Placebo for 12 weeks followed by a wash out period of 2 weeks and empagliflozin 25 mg for 12 weeks; or sequence 2: empagliflozin 25 mg for 12 weeks followed by a wash-out period of 2 weeks and placebo for 12 weeks.CFVR• No significant effect on CFVR, neither after empagliflozin treatment nor placebo
• Significant difference in hyperaemia CFV when the empagliflozin change was compared to the placebo change (−0.04 ± 0.15 vs. 0.05 ± 0.13, P = 0.044); however, the difference is small and can be regarded as a change finding.
Jürgens et al., 202152Empagliflozin (25 mg/day)90 T2DM patients were randomized to either empagliflozin or placebo for 13 weeks.MFR• No significant change in MFR
• No significant change in RPP adjusted MFR
• No significant difference between groups in rest flow (mL/g per min)
• No significant difference between groups in stress flow (mL/g per min)
• No significant change in reversible extent, TPD (%)
Lauritsen et al., 202153Empagliflozin (25 mg/day)13 T2DM patients were randomized (1:1) to receive either empagliflozin or placebo.MFR
MBF
• Empagliflozin reduced resting MBF compared to placebo (0.74 ± 0.10 vs 0.85 ± 0.10 mL/g/min, P < 0.01). This decrease in MBF remained significant even after being adjusted for RPP (1.01 vs. 1.06 mL/g/min, P = 0.04)
• No significant effect on stress MBF or MFR
Leccisotti et al., 202255Dapagliflozin (10 mg/day)16 patients with T2DM and stable CAD were equally randomized to receive either dapagliflozin or placebo for 4 weeks.MFR• ↑ MFR in the dapagliflozin group (2.56 ± 0.26 vs. 3.59 ± 0.35, P = 0.006), in contrast to placebo (2.34 ± 0.21 vs. 2.38 ± 0.24, P = 0.81). P for interaction = 0.001.
• ↑ MFR corrected for resting RPP in the dapagliflozin group (2.22 ± 0.25 vs. 3.23 ± 0.4, P = 0.008), in contrast to placebo. P for interaction = 0.019.
• ↓ Resting MBF in the dapagliflozin group, even after correction for resting RPP (1.15 ± 0.09 vs. 0.92 ± 0.10 mL/min/g, P = 0.005), compared with the placebo group (1.20 ± 0.10 vs. 1.18 ± 0.17 mL/min/g, P = 0.53). P for interaction = 0.045.
• Trend towards an increase in stress MBF in the dapagliflozin group (2.32 ± 0.15 vs. 2.64 ± 0.20 mL/min/g, P = 0.054), compared with placebo (2.47 ± 0.15 vs. 2.36 ± 0.16 mL/min/g, P = 0.55).

Abbreviations: 8-OHdG, 8-hydroxy-2ʹ-deoxyguanosin; CAD, coronary artery disease; CFV, coronary flow velocity; CFVR, coronary flow velocity reserve; CI, confidence interval; FMD, flow-mediated dilatation; HFrEF, heart failure with reduced ejection fraction; ICAM-1, intercellular adhesion molecule-1; LnRHI, natural logarithmic transformation of reactive-hyperemia index; MBF, myocardial blood flow; MFR, myocardial flow reserve; NMD, nitroglycerin-mediated dilation; RHI, reactive hyperaemia index; RPP, rate–pressure product; SGLT2 inhibitor, sodium-glucose cotransporter 2 inhibitor; T1DM, type 1 diabetes mellitus; T2DM, type 2 diabetes mellitus; TPD, total perfusion deficit.

Table 2

Clinical studies regarding the effect of SGLT2 inhibitors in endothelial function and microcirculation

ReferenceDrug/DosagePatientsMeasured indexOutcome
Irace et al., 201943Empagliflozin (10 mg/day, up-titrated at 25 mg/day in 13 patients)
Sitagliptin (100 mg/day)
Liraglutide (0.6 mg/day, up-titrated at 1.2 mg/day or even at 1.8 mg/day)
(1) 20 T2DM patients treated with empagliflozin
(2) 15 T2DM patients treated with incretin
(3) 25 nondiabetic healthy subjects as controls
FMD• Significant difference of peak wall shear stress and FMD in the empagliflozin group compared to control (P < 0.05 and P = 0.04, respectively)
• ↑ Brachial artery ultrasound-based FMD after 1 month of empagliflozin treatment (4.8 ± 4.5% vs. 7.7 ± 5.3%, P = 0.03)
• ↑ Brachial artery ultrasound-based FMD after 3 months of empagliflozin treatment (4.8 ± 4.5% vs. 8.5 ± 5.6%, P = 0.03)
• ↑ Peak brachial artery wall shear stress after 1 month of empagliflozin treatment (61±20 vs. 65 ± 23 dynes/cm2, P = 0.04)
• ↑ Peak brachial artery wall shear stress after 3 months of empagliflozin treatment (61±20 vs. 68 ± 25 dynes/cm2, P = 0.04)
• No significant changes in the incretin-based therapy group
Lunder et al., 201811Empagliflozin (25 mg/day)
Metformin (2000 mg/day)
40 T1DM patients randomized into four groups:
(1) Empagliflozin,
(2) Metformin,
(3) Empagliflozin/metformin and
(4) placebo
FMD
RHI
• No comparison was made between the treatment groups and the control.
• Within each treatment group, brachial artery ultrasound-based FMD and RHI were significantly improved after 12 weeks of treatment.
Shigiyama et al., 201747Dapagliflozin (5 mg/day) & Metformin (750 mg/day)
Metformin (1500 mg/day)
80 T2DM patients were randomized to receive either dapagliflozin on top of metformin or metformin alone for 16 weeks.FMD• FMD tended to improve only in the dapagliflozin group, but ΔFMD was comparable between the two groups.
• ↑ FMD in dapagliflozin group compared with metformin group in a subgroup analysis of patients with HbA1c ≥ 7.0% (ΔFMD = 1.05 ± 2.59 and −0.94 ± 2.39, respectively, P = 0.041).
• ↓ 8-OHdG in the dapagliflozin group compared with the metformin group (P < 0.001).
Solini et al., 201748Dapagliflozin (10 mg/day)
Hydrochlorothiazide (12.5 mg/day)
(1) 16 T2DM patients treated with dapagliflozin for two days
(2) 10 T2DM patients treated with hydrochlorothiazide for two days as controls
FMD• ↑ FMD after 2 days of treatment with dapagliflozin (2.8 ± 2.2 to 4.0 ± 2.1%, P < 0.05).
• No significant change in FMD within the control group.
Solini et al., 201949Dapagliflozin (10 mg/day)
Hydrochlorothiazide (12.5 mg/day)
40 T2DM patients with hypertension were equally randomized to either dapagliflozin or hydrochlorothiazide for 4 weeks.FMD• No significant change in FMD in any group.
• The miRNAs linked to endothelial dysfunction (miR-200b and miR-27b) were equally increased in both groups [in the dapagliflozin group: 0.379 (0.580) to 2.802 (4.179) and 0.072 (0.050) to 0.172 (0.209), respectively].
Sposito et al., 202145Dapagliflozin (10 mg/day)
Glibenclamide (5 mg/day)
98 T2DM patients with atherosclerotic disease were randomized 1:1 to 12 weeks of therapy with dapagliflozin or glibenclamide in addition to metformin.FMD• ↑ 3.3% median increase in rest FMD at 1 min in the dapagliflozin group (P = 0.0001).
• ↓ 1.2% median decrease in rest FMD at 1 min in the glibenclamide group (P = 0.0001).
• FMD after I/R slightly increased (+2.1%) in the dapagliflozin group and decreased (−0.4%) in the glibenclamide group, but the changes did not reach statistical significance.
• Dapagliflozin—in contrast to glibenclamide—improved the paradoxical arterial constriction observed during FMD.
• Significant difference between the two groups regarding the end diastolic velocity, peak systolic velocity, and resistive indices measured at 1 and 5 min.
• ↑ 10% in the 1-min nitrite level in the dapagliflozin group vs. ↓ 4% in the glibenclamide group.
Zainordin et al., 201946Dapagliflozin (10 mg/day)81 T2DM patients with established ischemic heart disease were randomized using stratified randomization to 12 weeks of therapy with dapagliflozin or placebo in addition to diet, exercise, insulin, and/or metformin.FMD• No significant difference in FMD after 12 weeks, neither between the dapagliflozin and the placebo group nor within each group.
• ↑ NMD in the dapagliflozin group compared to placebo after 12 weeks of treatment (19.64 ± 9.71% vs. 15.11 ± 8.44%, P = 0.038).
• ↓ ICAM-1 within the dapagliflozin group after 12 weeks of treatment (−83.9 ± 205.9 ng/mL, P < 0.02).
Ilyas et al., 202151Dapagliflozin (10 mg/day)19 T2DM patients with HFrEF received dapagliflozin or placebo for 2 weeks and, after a 2-week washout period, they were crossed over for 2 more weeks of therapy.RHI• ↓ RHI after 2 weeks on dapagliflozin
Sugiyama et al., 201850Dapagliflozin (5 mg/day)54 patients with uncontrolled T2DM were nonrandomly allocated to either dapagliflozin or non-SGLT2 inhibitor medications for 6 months.RHI• Absolute and percentage changes in the LnRHI significantly greater in the dapagliflozin group when compared to control (P = 0.028).
• ↑ LnRHI after 6 months in the dapagliflozin group (0.450 ± 0.135 to 0.662 ± 0.230, P < 0.01). The LnRHI change in the non-SGLT2 inhibitor group was insignificant.
• The proportion of patients with an increase in LnRHI > 15% was 74.1% in the dapagliflozin group, but only 40.7% in the non-SGLT2 inhibitor group (P = 0.01).
• 4.156 (95% CI: 1.312–13.169) odds ratio in the univariate logistic regression analysis for the improvement of microvascular endothelial function among the dapagliflozin group (P = 0.015).
Tanaka et al., 201944Empagliflozin (10 mg/day)117 patients with T2DM and cardiovascular disease were equally randomized to either empagliflozin or placebo for 24 weeks.RHI• Absolute changes in RHI were −0.006 (SD 0.478) in the empagliflozin group and −0.025 (0.454) in the placebo group.
Suhrs et al., 202254Empagliflozin (25 mg/day)26 T2DM participants were randomized (1:1) to either sequence 1: Placebo for 12 weeks followed by a wash out period of 2 weeks and empagliflozin 25 mg for 12 weeks; or sequence 2: empagliflozin 25 mg for 12 weeks followed by a wash-out period of 2 weeks and placebo for 12 weeks.CFVR• No significant effect on CFVR, neither after empagliflozin treatment nor placebo
• Significant difference in hyperaemia CFV when the empagliflozin change was compared to the placebo change (−0.04 ± 0.15 vs. 0.05 ± 0.13, P = 0.044); however, the difference is small and can be regarded as a change finding.
Jürgens et al., 202152Empagliflozin (25 mg/day)90 T2DM patients were randomized to either empagliflozin or placebo for 13 weeks.MFR• No significant change in MFR
• No significant change in RPP adjusted MFR
• No significant difference between groups in rest flow (mL/g per min)
• No significant difference between groups in stress flow (mL/g per min)
• No significant change in reversible extent, TPD (%)
Lauritsen et al., 202153Empagliflozin (25 mg/day)13 T2DM patients were randomized (1:1) to receive either empagliflozin or placebo.MFR
MBF
• Empagliflozin reduced resting MBF compared to placebo (0.74 ± 0.10 vs 0.85 ± 0.10 mL/g/min, P < 0.01). This decrease in MBF remained significant even after being adjusted for RPP (1.01 vs. 1.06 mL/g/min, P = 0.04)
• No significant effect on stress MBF or MFR
Leccisotti et al., 202255Dapagliflozin (10 mg/day)16 patients with T2DM and stable CAD were equally randomized to receive either dapagliflozin or placebo for 4 weeks.MFR• ↑ MFR in the dapagliflozin group (2.56 ± 0.26 vs. 3.59 ± 0.35, P = 0.006), in contrast to placebo (2.34 ± 0.21 vs. 2.38 ± 0.24, P = 0.81). P for interaction = 0.001.
• ↑ MFR corrected for resting RPP in the dapagliflozin group (2.22 ± 0.25 vs. 3.23 ± 0.4, P = 0.008), in contrast to placebo. P for interaction = 0.019.
• ↓ Resting MBF in the dapagliflozin group, even after correction for resting RPP (1.15 ± 0.09 vs. 0.92 ± 0.10 mL/min/g, P = 0.005), compared with the placebo group (1.20 ± 0.10 vs. 1.18 ± 0.17 mL/min/g, P = 0.53). P for interaction = 0.045.
• Trend towards an increase in stress MBF in the dapagliflozin group (2.32 ± 0.15 vs. 2.64 ± 0.20 mL/min/g, P = 0.054), compared with placebo (2.47 ± 0.15 vs. 2.36 ± 0.16 mL/min/g, P = 0.55).
ReferenceDrug/DosagePatientsMeasured indexOutcome
Irace et al., 201943Empagliflozin (10 mg/day, up-titrated at 25 mg/day in 13 patients)
Sitagliptin (100 mg/day)
Liraglutide (0.6 mg/day, up-titrated at 1.2 mg/day or even at 1.8 mg/day)
(1) 20 T2DM patients treated with empagliflozin
(2) 15 T2DM patients treated with incretin
(3) 25 nondiabetic healthy subjects as controls
FMD• Significant difference of peak wall shear stress and FMD in the empagliflozin group compared to control (P < 0.05 and P = 0.04, respectively)
• ↑ Brachial artery ultrasound-based FMD after 1 month of empagliflozin treatment (4.8 ± 4.5% vs. 7.7 ± 5.3%, P = 0.03)
• ↑ Brachial artery ultrasound-based FMD after 3 months of empagliflozin treatment (4.8 ± 4.5% vs. 8.5 ± 5.6%, P = 0.03)
• ↑ Peak brachial artery wall shear stress after 1 month of empagliflozin treatment (61±20 vs. 65 ± 23 dynes/cm2, P = 0.04)
• ↑ Peak brachial artery wall shear stress after 3 months of empagliflozin treatment (61±20 vs. 68 ± 25 dynes/cm2, P = 0.04)
• No significant changes in the incretin-based therapy group
Lunder et al., 201811Empagliflozin (25 mg/day)
Metformin (2000 mg/day)
40 T1DM patients randomized into four groups:
(1) Empagliflozin,
(2) Metformin,
(3) Empagliflozin/metformin and
(4) placebo
FMD
RHI
• No comparison was made between the treatment groups and the control.
• Within each treatment group, brachial artery ultrasound-based FMD and RHI were significantly improved after 12 weeks of treatment.
Shigiyama et al., 201747Dapagliflozin (5 mg/day) & Metformin (750 mg/day)
Metformin (1500 mg/day)
80 T2DM patients were randomized to receive either dapagliflozin on top of metformin or metformin alone for 16 weeks.FMD• FMD tended to improve only in the dapagliflozin group, but ΔFMD was comparable between the two groups.
• ↑ FMD in dapagliflozin group compared with metformin group in a subgroup analysis of patients with HbA1c ≥ 7.0% (ΔFMD = 1.05 ± 2.59 and −0.94 ± 2.39, respectively, P = 0.041).
• ↓ 8-OHdG in the dapagliflozin group compared with the metformin group (P < 0.001).
Solini et al., 201748Dapagliflozin (10 mg/day)
Hydrochlorothiazide (12.5 mg/day)
(1) 16 T2DM patients treated with dapagliflozin for two days
(2) 10 T2DM patients treated with hydrochlorothiazide for two days as controls
FMD• ↑ FMD after 2 days of treatment with dapagliflozin (2.8 ± 2.2 to 4.0 ± 2.1%, P < 0.05).
• No significant change in FMD within the control group.
Solini et al., 201949Dapagliflozin (10 mg/day)
Hydrochlorothiazide (12.5 mg/day)
40 T2DM patients with hypertension were equally randomized to either dapagliflozin or hydrochlorothiazide for 4 weeks.FMD• No significant change in FMD in any group.
• The miRNAs linked to endothelial dysfunction (miR-200b and miR-27b) were equally increased in both groups [in the dapagliflozin group: 0.379 (0.580) to 2.802 (4.179) and 0.072 (0.050) to 0.172 (0.209), respectively].
Sposito et al., 202145Dapagliflozin (10 mg/day)
Glibenclamide (5 mg/day)
98 T2DM patients with atherosclerotic disease were randomized 1:1 to 12 weeks of therapy with dapagliflozin or glibenclamide in addition to metformin.FMD• ↑ 3.3% median increase in rest FMD at 1 min in the dapagliflozin group (P = 0.0001).
• ↓ 1.2% median decrease in rest FMD at 1 min in the glibenclamide group (P = 0.0001).
• FMD after I/R slightly increased (+2.1%) in the dapagliflozin group and decreased (−0.4%) in the glibenclamide group, but the changes did not reach statistical significance.
• Dapagliflozin—in contrast to glibenclamide—improved the paradoxical arterial constriction observed during FMD.
• Significant difference between the two groups regarding the end diastolic velocity, peak systolic velocity, and resistive indices measured at 1 and 5 min.
• ↑ 10% in the 1-min nitrite level in the dapagliflozin group vs. ↓ 4% in the glibenclamide group.
Zainordin et al., 201946Dapagliflozin (10 mg/day)81 T2DM patients with established ischemic heart disease were randomized using stratified randomization to 12 weeks of therapy with dapagliflozin or placebo in addition to diet, exercise, insulin, and/or metformin.FMD• No significant difference in FMD after 12 weeks, neither between the dapagliflozin and the placebo group nor within each group.
• ↑ NMD in the dapagliflozin group compared to placebo after 12 weeks of treatment (19.64 ± 9.71% vs. 15.11 ± 8.44%, P = 0.038).
• ↓ ICAM-1 within the dapagliflozin group after 12 weeks of treatment (−83.9 ± 205.9 ng/mL, P < 0.02).
Ilyas et al., 202151Dapagliflozin (10 mg/day)19 T2DM patients with HFrEF received dapagliflozin or placebo for 2 weeks and, after a 2-week washout period, they were crossed over for 2 more weeks of therapy.RHI• ↓ RHI after 2 weeks on dapagliflozin
Sugiyama et al., 201850Dapagliflozin (5 mg/day)54 patients with uncontrolled T2DM were nonrandomly allocated to either dapagliflozin or non-SGLT2 inhibitor medications for 6 months.RHI• Absolute and percentage changes in the LnRHI significantly greater in the dapagliflozin group when compared to control (P = 0.028).
• ↑ LnRHI after 6 months in the dapagliflozin group (0.450 ± 0.135 to 0.662 ± 0.230, P < 0.01). The LnRHI change in the non-SGLT2 inhibitor group was insignificant.
• The proportion of patients with an increase in LnRHI > 15% was 74.1% in the dapagliflozin group, but only 40.7% in the non-SGLT2 inhibitor group (P = 0.01).
• 4.156 (95% CI: 1.312–13.169) odds ratio in the univariate logistic regression analysis for the improvement of microvascular endothelial function among the dapagliflozin group (P = 0.015).
Tanaka et al., 201944Empagliflozin (10 mg/day)117 patients with T2DM and cardiovascular disease were equally randomized to either empagliflozin or placebo for 24 weeks.RHI• Absolute changes in RHI were −0.006 (SD 0.478) in the empagliflozin group and −0.025 (0.454) in the placebo group.
Suhrs et al., 202254Empagliflozin (25 mg/day)26 T2DM participants were randomized (1:1) to either sequence 1: Placebo for 12 weeks followed by a wash out period of 2 weeks and empagliflozin 25 mg for 12 weeks; or sequence 2: empagliflozin 25 mg for 12 weeks followed by a wash-out period of 2 weeks and placebo for 12 weeks.CFVR• No significant effect on CFVR, neither after empagliflozin treatment nor placebo
• Significant difference in hyperaemia CFV when the empagliflozin change was compared to the placebo change (−0.04 ± 0.15 vs. 0.05 ± 0.13, P = 0.044); however, the difference is small and can be regarded as a change finding.
Jürgens et al., 202152Empagliflozin (25 mg/day)90 T2DM patients were randomized to either empagliflozin or placebo for 13 weeks.MFR• No significant change in MFR
• No significant change in RPP adjusted MFR
• No significant difference between groups in rest flow (mL/g per min)
• No significant difference between groups in stress flow (mL/g per min)
• No significant change in reversible extent, TPD (%)
Lauritsen et al., 202153Empagliflozin (25 mg/day)13 T2DM patients were randomized (1:1) to receive either empagliflozin or placebo.MFR
MBF
• Empagliflozin reduced resting MBF compared to placebo (0.74 ± 0.10 vs 0.85 ± 0.10 mL/g/min, P < 0.01). This decrease in MBF remained significant even after being adjusted for RPP (1.01 vs. 1.06 mL/g/min, P = 0.04)
• No significant effect on stress MBF or MFR
Leccisotti et al., 202255Dapagliflozin (10 mg/day)16 patients with T2DM and stable CAD were equally randomized to receive either dapagliflozin or placebo for 4 weeks.MFR• ↑ MFR in the dapagliflozin group (2.56 ± 0.26 vs. 3.59 ± 0.35, P = 0.006), in contrast to placebo (2.34 ± 0.21 vs. 2.38 ± 0.24, P = 0.81). P for interaction = 0.001.
• ↑ MFR corrected for resting RPP in the dapagliflozin group (2.22 ± 0.25 vs. 3.23 ± 0.4, P = 0.008), in contrast to placebo. P for interaction = 0.019.
• ↓ Resting MBF in the dapagliflozin group, even after correction for resting RPP (1.15 ± 0.09 vs. 0.92 ± 0.10 mL/min/g, P = 0.005), compared with the placebo group (1.20 ± 0.10 vs. 1.18 ± 0.17 mL/min/g, P = 0.53). P for interaction = 0.045.
• Trend towards an increase in stress MBF in the dapagliflozin group (2.32 ± 0.15 vs. 2.64 ± 0.20 mL/min/g, P = 0.054), compared with placebo (2.47 ± 0.15 vs. 2.36 ± 0.16 mL/min/g, P = 0.55).

Abbreviations: 8-OHdG, 8-hydroxy-2ʹ-deoxyguanosin; CAD, coronary artery disease; CFV, coronary flow velocity; CFVR, coronary flow velocity reserve; CI, confidence interval; FMD, flow-mediated dilatation; HFrEF, heart failure with reduced ejection fraction; ICAM-1, intercellular adhesion molecule-1; LnRHI, natural logarithmic transformation of reactive-hyperemia index; MBF, myocardial blood flow; MFR, myocardial flow reserve; NMD, nitroglycerin-mediated dilation; RHI, reactive hyperaemia index; RPP, rate–pressure product; SGLT2 inhibitor, sodium-glucose cotransporter 2 inhibitor; T1DM, type 1 diabetes mellitus; T2DM, type 2 diabetes mellitus; TPD, total perfusion deficit.

Trials regarding endothelial function

A variety of studies provide insight into the effect of SGLT2 inhibitors on endothelial function. Lunder et al.11 studied 40 insulin-dependent male patients that were randomized into four groups: the empagliflozin, the metformin, the empagliflozin/metformin, and the placebo group. Endothelial function was assessed using brachial artery ultrasound-based FMD and RHI, measured at inclusion and after 12 weeks of treatment. Brachial artery FMD and RHI were significantly improved after treatment in all treatment groups and most prominently in the empagliflozin/metformin. In the placebo group, neither FMD nor RHI was found significantly changed. However, no comparison was made between the treatment groups and the control, thus the results of this study should be interpreted cautiously. The same research team also found a significant improvement in arterial stiffness after empagliflozin treatment compared to placebo, therefore, providing another mechanism through which SGLT2 inhibitors exert their cardioprotective properties.

Similarly, Irace et al.43 investigated the effect of empagliflozin on endothelial function in patients with type 2 DM. The patients were treated either with empagliflozin (n = 20) or incretin (n = 15). Endothelial function was assessed by measuring brachial artery ultrasound-based FMD and shear stress at baseline and after 1 and 3 months. Patients treated with empagliflozin had a significant increase in both peak wall shear stress (61±20 vs. 68 ± 25 dynes/cm2 after 3 months, P = 0.04) and FMD (4.8 ± 4.5% vs. 8.5 ± 5.6% after 3 months, P = 0.03), with no difference being observed in the incretin-based therapy group. The difference of peak wall shear stress and FMD was also significant when the empagliflozin group was compared to control (P < 0.05 and P = 0.04, respectively). Significant effects of empagliflozin were reported as early as after 1 month of treatment. The cohort was further divided into three FMD categories according to the time of maximal dilation such as early, late, and no dilators. Empagliflozin significantly increased the early dilators and decreased the late and no dilators, while no significant change was observed with the incretin-based therapy. This observation has important clinical relevance, as delayed dilation has been linked to underlying cardiovascular disease and cardiovascular risk factors.43

Contrasting with the two previously described studies, the EMBLEM trial,44 including a total of 117 adults with type 2 DM and cardiovascular disease, who were equally randomized to receive either empagliflozin or placebo for 24 weeks, showed no effect of empagliflozin treatment in endothelial function, as measured by RHI. It should be noted that the participants had relatively well-controlled concomitant diseases, such as blood pressure, BMI, and HbA1c.

Regarding dapagliflozin, a few studies have also investigated its effect on endothelial dysfunction. Sposito et al.45 studied 98 diabetic patients with atherosclerotic disease, who were randomized 1:1 to 12 weeks of therapy with dapagliflozin or glibenclamide, in addition to metformin. Endothelial function was primarily assessed by measuring 1-min brachial artery ultrasound-based-FMD both at rest and after 15 min of ischemia followed by 15 min of reperfusion time (I/R). The post-treatment change in rest FMD at 1 min was significantly different between the two groups, with a median 3.3% increase being displayed in the dapagliflozin group, while the result remained statistically significant after adjustment for the office systolic and diastolic blood pressure change and pulse pressure (PP) change. Interestingly, dapagliflozin also improved the paradoxical arterial constriction that was observed in 30% of the patients after cuff deflation during the FMD protocol, which could be attributed to improved endothelial function. The authors additionally reported a significant difference between the two groups regarding the 1-min nitrite level, as well as the end diastolic velocity, peak systolic velocity, and resistive indices measured at 1 and 5 min. All aforementioned effects of dapagliflozin indicate an improvement in macro- and microvascular endothelial function in patients with type 2 diabetes mellitus and atherosclerotic burden.

In a similar manner, Zainordin et al.46 investigated the effects of dapagliflozin on endothelial dysfunction in type 2 diabetics with established ischemic heart disease, but no significant difference in FMD was found neither between the dapagliflozin and the placebo group nor within each group. However, the placebo group showed a worsening of ΔFMD, whereas the dapagliflozin group did not display any change. The researchers also measured the nitroglycerin-mediated dilation (NMD) and found that after 12 weeks of therapy, it was significantly higher in the dapagliflozin group compared to placebo. The endothelial function was additionally evaluated by surrogate markers, one of which (ICAM-1) was significantly reduced after treatment with dapagliflozin (−83.9 ± 205.9 ng/mL, P < 0.02), in contrast to the placebo group, where it remained unchanged.

Shigiyama et al.47 examined a total of 80 patients with type 2 DM who were randomized to receive either metformin or metformin on top of dapagliflozin for 16 weeks. FMD displayed a tendency towards improvement in the dapagliflozin group, but ΔFMD was comparable between the two groups. However, in the subgroup analysis of patients with HbA1c≥7.0%, FMD showed a significant improvement in the dapagliflozin group compared with the metformin group (ΔFMD = 1.05 ± 2.59 and −0.94 ± 2.39, respectively, P = 0.041). Considering the comparable glycemic control in the two groups, the authors speculate that dapagliflozin's action is mediated through mechanisms other than its glucose lowering effect.

Moreover, Solini et al.,48 in a pilot study, showed a significant increase in FMD independent of blood pressure when 16 patients with type 2 DM were treated with dapagliflozin for 2 days (2.8 ± 2.2 to 4.0 ± 2.1%, P < 0.05), compared to 10 patients treated with hydrochlorothiazide, whose FMD did not present any significant change. In a subsequent study,49 40 patients with type 2 DM and hypertension were randomized to receive for 4 weeks either dapagliflozin or hydrochlorothiazide, but neither group presented any significant change in FMD, while miRNAs linked to endothelial dysfunction (miR-200b and miR-27b) were found equally increased in both groups.

Sugiyama et al.50 studied 54 patients with uncontrolled type 2 DM, who were non-randomly allocated to either dapagliflozin or non-SGLT2 inhibitor medication for 6 months in order to assess the peripheral microvascular endothelial function, using reactive hyperemia-peripheral arterial tonometry (RT-PAT) and calculating the natural logarithmic transformed value of the RH-PAT index (LnRHI). Treatment with dapagliflozin resulted in a significant intra-group improvement of the endothelial function (LnRHI: from 0.450 ± 0.135 to 0.662 ± 0.230, P < 0.01). The absolute and percentage changes in the LnRHI were also significantly greater when the dapagliflozin group was compared to control (P = 0.028). The beneficial role of dapagliflozin is further highlighted by the fact that 74.1% of the patients in this group presented with an increase in LnRHI exceeding 15%, as opposed to the control group, where only 40.7% had a similar increase (P = 0.01).

In contrast to the results of the aforementioned studies, a double-blind, crossover, placebo-controlled trial51 demonstrated a worsening of microvascular endothelial function in 19 type 2 diabetic HFrEF patients after a 2-week treatment with dapagliflozin. The patients were specifically found with a reduced peripheral capillary-derived RHI [1.40 (1.23, 1.84) vs. 1.29 (1.21, 1.56), P < 0.05]. However, the results should be interpreted cautiously, as the number of participants was relatively small.

Trials on microcirculation

Several studies have investigated the effects of SGLT2 inhibitors on coronary microcirculation. The SIMPLE trial52 evaluated 90 patients with type 2 DM, who were randomly assigned to either empagliflozin or placebo for 13 weeks. MFR was quantified noninvasively by Rubidium-82 positron emission tomography/computed tomography (PET/CT) at inclusion and at week 13. There was no significant change in MFR between baseline and week 13 in either the empagliflozin or placebo groups. The result remained insignificant when adjusted for the rate–pressure product to account for baseline workload. The researchers also evaluated myocardial blood flow (MBF) during rest and stress, as well as reversible cardiac ischemia; however, no difference was observed between the groups. Some possible explanations behind the failure of the study to display a statistically significant result, as speculated by the authors, are the high prevalence of concomitant treatment that could have blunted the impact of empagliflozin on MFR, in addition to the relatively high baseline MFR of this cohort.

Lauritsen et al.53 also studied the effect of empagliflozin on MFR and MBF in patients with type 2 DM. A total of 13 patients were randomized to either empagliflozin treatment or placebo and, after 4 weeks, 15O-H2O PET/CT was used to evaluate MFR and MBF. Interestingly, compared to placebo, empagliflozin significantly reduced resting MBF by 13% (empagliflozin: 0.74 ± 0.10 mL/g/min; placebo: 0.85 ± 0.10 mL/g/min; P < 0.01), a result that remained significant even after adjustment for cardiac workload. On the other hand, empagliflozin treatment did not significantly affect stress MBF or MFR, but it should be noted that the latter displayed a tendency towards improvement (P = 0.09).

Moreover, Suhrs et al.54 analysed the effect of empagliflozin on CFVR in type 2 diabetics. A total of 26 patients were randomized 1:1 to each treatment sequence, treated with empagliflozin and placebo for 12 weeks with a 2-week wash-out period. CFVR was measured with transthoracic doppler echocardiography. No significant effect was observed on CFVR, neither after empagliflozin treatment nor placebo, thus not supporting the hypothesis that the beneficial effect of SGLT2 inhibitors is due to improvement of the non-endothelial dependent coronary microvascular function. When empagliflozin treatment was compared with placebo, a significant difference (P = 0.044) was shown in hyperemia CFV, but it was small and regarded as a change finding.

Finally, the DAPAHEART trial55 demonstrated for the first time in humans a significant increase in MFR after treatment with a SGLT2 inhibitor. A total of 16 patients with type 2 DM and stable coronary artery disease were equally randomized to receive for 4 weeks either dapagliflozin or placebo. MFR was assessed using 13N-ammonia PET/CT. Patients in the dapagliflozin group showed a significant improvement in MFR (2.56 ± 0.26 vs. 3.59 ± 0.35, P = 0.006) compared with the placebo group. The result remained significant even after correction for resting rate–pressure product (2.22 ± 0.25 vs. 3.23 ± 0.4, P = 0.008). The dapagliflozin arm also displayed a significantly lower resting MBF and a trend towards an increased stress MBF, compared with the placebo group.

Just as importantly, SGLT2 inhibitors have also been found to improve arterial stiffness and peripheral vascular resistance. A double-blind, placebo-controlled randomized clinical trial56 investigated the effects of empagliflozin in nondiabetic patients with HFrEF and found that pulse wave velocity (PWV) was significantly reduced in the treatment group compared to controls (−0.58 cm/s vs. 0.60 cm/s, P < 0.01). Similarly, in a post hoc analysis57 of data from phase III trials including T2DM patients, empagliflozin was shown to significantly (P < 0.001) reduce PP, mean arterial pressure, and double product (DP or RPP). There was also a trend towards reduction in the ambulatory arterial stiffness index with empagliflozin in one of the cohorts (P = 0.059).

Clinical perspectives and future directions

Currently SGLT2 inhibitors constitute the only drug class that has shown positive results in HFpEF and is suggested by heart failure guidelines8,59 for treating patients with HF regardless of their EF. Endothelial and microvascular dysfunction have a key role in the pathogenesis of HF, with myocardial and endothelial inflammation, oxidative stress and impaired vasodilation being some of the microcirculation-related pathogenetic mechanisms. Notably, specifically in HFpEF, MVD is highly prevalent, reaching up to 70–80% of patients.60,61 In addition, the association of common comorbidities among HFpEF patients (hypertension, metabolic syndrome, and dyslipidemia) with the development of endothelial dysfunction are suggestive of the key role MVD has in the pathogenesis of HFpEF.15 Intriguingly, other agents acting on the endothelium and microcirculation, such as ACE and PDE-5 inhibitors, have failed until now to show any clinical benefit in HFpEF.

As thoroughly described thus far, plenty preclinical and clinical data demonstrate the endothelial and microcirculatory function improvement induced by empagliflozin and dapagliflozin. Yet still, these studies have limitations. In this area of research with small animal studies, it is likely that there is a publication bias with some neutral results not being published. Furthermore, the in vivo studies have almost exclusively used mice and rats as their models, thus deterring us from extrapolating the results to humans. The in vitro studies using human cells overcome this limitation; however, these studies are limited. Moreover, the cells used are of high diversity in origin and developmental stage, in addition to being cultured mainly under static conditions. Lastly, the evaluation of methodological quality in animal studies—as opposed to human clinical trials—is challenging due to lack of standardized assessment scales.

Regarding clinical trials, most of them verify the beneficial effects displayed in the preclinical models. However, an important aspect that should be clarified is that some of the effects of SGLT2 inhibitors on the endothelium and microcirculation could also be attributed to the accompanying weight loss. On top of that, a few trials have failed to show significant beneficial SGLT2 effects and they should not be disregarded. The conflicting results might be explained by the differences seen in patient population, treatment protocols, indexes assessed, and no thorough pre-protocol assessment of the microcirculation status. This highlights the need for more intensified research on this particular topic. Additionally, there are still no trials assessing endothelial or microvascular functional indexes after treatment with SGLT2 inhibitors specifically in patients with HFpEF. Therefore, the focus from now onwards should be redirected on examining the impact of SGLT2 inhibition on the microcirculation of HFpEF patients. Finally, MVD characterizes several other cardiovascular diseases with limited therapeutic options, such as Myocardial Infarction with No Obstructed Coronary Arteries, Refractory Angina, and Takotsubo Cardiomyopathy. Given the pleiotropic effects of SGLT2 inhibitors on microcirculation and endothelial function, it is possible that this drug class could be of therapeutic benefit in these patients. Thus, future trials should assess whether SGLT2 inhibition could provide clinical benefit in other pathologies with microcirculation disorders, besides HFpEF.

Conclusion

SGLT2 inhibitors have well-documented cardioprotective effects and constitute a new therapeutic option for heart failure patients regardless of the presence of diabetes. Interestingly, preclinical studies establish several molecular mechanisms whereby empagliflozin and dapagliflozin improve endothelial and microcirculatory function, while the clinical trials verify these positive effects. However, there is still much debate about the exact underlying mechanisms for the impact of SGLT2 on the cardiovascular system, as conflicting data still exist. Therefore, larger scale studies are needed to verify the existing evidence and shed light on the complex responsible pathways for the rather unique beneficial actions of SGLT2 in humans.

Acknowledgement

None.

Funding

None.

Conflict of interest: None.

Data availability statement

The data for this review are available under request from the authors.

References

1.

Adingupu
DD
,
Göpel
SO
,
Grönros
J
,
Behrendt
M
,
Sotak
M
,
Miliotis
T
,
Dahlqvist
U
,
Gan
L-M
,
Jönsson-Rylander
A-C
.
SGLT2 inhibition with empagliflozin improves coronary microvascular function and cardiac contractility in prediabetic ob/ob/ mice
.
Cardiovasc Diabetol
2019
;
18
:
16
.

2.

Zinman
B
,
Wanner
C
,
Lachin
JM
,
Fitchett
D
,
Bluhmki
E
,
Hantel
S
,
Mattheus
M
,
Devins
T
,
Johansen
OE
,
Woerle
HJ
,
Broedl
UC
,
Inzucchi
SE
.
EMPA-REG OUTCOME investigators. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes
.
N Engl J Med
2015
;
373
:
2117
2128
.

3.

Neal
B
,
Perkovic
V
,
Mahaffey
KW
,
De Zeeuw
D
,
Fulcher
G
,
Erondu
N
,
Shaw
W
,
Law
G
,
Desai
M
,
Matthews
DR
.
CANVAS Program Collaborative Group. Canagliflozin and cardiovascular and renal events in type 2 diabetes
.
N Engl J Med
2017
;
377
:
644
657
.

4.

Wiviott
SD
,
Raz
I
,
Bonaca
MP
,
Mosenzon
O
,
Kato
ET
,
Cahn
A
,
Silverman
MG
,
Zelniker
TA
,
Kuder
JF
,
Murphy
SA
,
Bhatt
DL
,
Leiter
LA
,
Mcguire
DK
,
Wilding
JPH
,
Ruff
CT
,
Gause-Nilsson
IAM
,
Fredriksson
M
,
Johansson
PA
,
Langkilde
A-M
,
Sabatine
MS
.
Dapagliflozin and cardiovascular outcomes in type 2 diabetes
.
N Engl J Med
2019
;
380
:
347
357
.

5.

Mcmurray
JJV
,
Solomon
SD
,
Inzucchi
SE
,
Køber
L
,
Kosiborod
MN
,
Martinez
FA
,
Ponikowski
P
,
Sabatine
MS
,
Anand
IS
,
Bělohlávek
J
,
Böhm
M
,
Chiang
C-E
,
Chopra
VK
,
De Boer
RA
,
Desai
AS
,
Diez
M
,
Drozdz
J
,
Dukát
A
,
Ge
J
,
Howlett
JG
,
Katova
T
,
Kitakaze
M
,
Ljungman
CEA
,
Merkely
B
,
Nicolau
JC
,
O'meara
E
,
Petrie
MC
,
Vinh
PN
,
Schou
M
,
Tereshchenko
S
,
Verma
S
,
Held
C
,
Demets
DL
,
Docherty
KF
,
Jhund
PS
,
Bengtsson
O
,
Sjöstrand
M
,
Langkilde
A-M
.
Dapagliflozin in patients with heart failure and reduced ejection fraction
.
N Engl J Med
2019
;
381
:
1995
2008
.

6.

Packer
M
,
Anker
SD
,
Butler
J
,
Filippatos
G
,
Pocock
SJ
,
Carson
P
,
Januzzi
J
,
Verma
S
,
Tsutsui
H
,
Brueckmann
M
,
Jamal
W
,
Kimura
K
,
Schnee
J
,
Zeller
C
,
Cotton
D
,
Bocchi
E
,
Böhm
M
,
Choi
D-J
,
Chopra
V
,
Chuquiure
E
,
Giannetti
N
,
Janssens
S
,
Zhang
J
,
Gonzalez Juanatey
JR
,
Kaul
S
,
Brunner-La Rocca
H-P
,
Merkely
B
,
Nicholls
SJ
,
Perrone
S
,
Pina
I
,
Ponikowski
P
,
Sattar
N
,
Senni
M
,
Seronde
M-F
,
Spinar
J
,
Squire
I
,
Taddei
S
,
Wanner
C
,
Zannad
F
.
Cardiovascular and renal outcomes with Empagliflozin in heart failure
.
N Engl J Med
2020
;
383
:
1413
1424
.

7.

Anker
SD
,
Butler
J
,
Filippatos
G
,
Ferreira
JP
,
Bocchi
E
,
Böhm
M
,
Brunner–La Rocca
H-P
,
Choi
D-J
,
Chopra
V
,
Chuquiure-Valenzuela
E
,
Giannetti
N
,
Gomez-Mesa
JE
,
Janssens
S
,
Januzzi
JL
,
Gonzalez-Juanatey
JR
,
Merkely
B
,
Nicholls
SJ
,
Perrone
SV
,
Piña
IL
,
Ponikowski
P
,
Senni
M
,
Sim
D
,
Spinar
J
,
Squire
I
,
Taddei
S
,
Tsutsui
H
,
Verma
S
,
Vinereanu
D
,
Zhang
J
,
Carson
P
,
Lam
CSP
,
Marx
N
,
Zeller
C
,
Sattar
N
,
Jamal
W
,
Schnaidt
S
,
Schnee
JM
,
Brueckmann
M
,
Pocock
SJ
,
Zannad
F
,
Packer
M
Empagliflozin in heart failure with a preserved ejection fraction
.
N Engl J Med
2021
;
385
:
1451
1461
.

8.

Santulli
G
,
Wang
X
,
Mone
P
Updated ACC/AHA/HFSA 2022 guidelines on heart failure: what's new? From epidemiology to clinical management
.
Eur Heart J Cardiovasc Pharmacother
2022
;
8
:
e23
e24
. doi:

9.

Soares
RN
,
Ramirez-Perez
FI
,
Cabral-Amador
FJ
,
Morales-Quinones
M
,
Foote
CA
,
Ghiarone
T
,
Sharma
N
,
Power
G
,
Smith
JA
,
Rector
RS
,
Martinez-Lemus
LA
,
Padilla
J
,
Manrique-Acevedo
C
SGLT2 inhibition attenuates arterial dysfunction and decreases vascular F-actin content and expression of proteins associated with oxidative stress in aged mice
.
Geroscience
2022
;
44
:
1657
1675
.

10.

Vancheri
F
,
Longo
G
,
Vancheri
S
,
Henein
M
.
Coronary microvascular dysfunction
.
J Clin Med
2020
;
9
:
2880
.

11.

Lunder
M
,
Janić
M
,
Japelj
M
,
Juretič
A
,
Janež
A
,
Šabovič
M
.
Empagliflozin on top of metformin treatment improves arterial function in patients with type 1 diabetes mellitus
.
Cardiovasc Diabetol
2018
;
17
:
153
.

12.

Nakao
M
,
Shimizu
I
,
Katsuumi
G
,
Yoshida
Y
,
Suda
M
,
Hayashi
Y
,
Ikegami
R
,
Hsiao
YT
,
Okuda
S
,
Soga
T
,
Minamino
T
.
Empagliflozin maintains capillarization and improves cardiac function in a murine model of left ventricular pressure overload
.
Sci Rep
2021
;
11
:
18384
.

13.

Santos-Gallego
CG
,
Requena-Ibanez
JA
,
San Antonio
R
,
Garcia-Ropero
A
,
Ishikawa
K
,
Watanabe
S
,
Picatoste
B
,
Vargas-Delgado
AP
,
Flores-Umanzor
EJ
,
Sanz
J
,
Fuster
V
,
Badimon
JJ
.
Empagliflozin ameliorates diastolic dysfunction and left ventricular fibrosis/stiffness in nondiabetic heart failure: a multimodality study
.
JACC Cardiovasc Imaging
2021
;
14
:
393
407
.

14.

Requena-Ibáñez
JA
,
Santos-Gallego
CG
,
Rodriguez-Cordero
A
,
Vargas-Delgado
AP
,
Badimón
JJ
.
Empagliflozin improves quality of life in nondiabetic HFrEF patients. Sub-analysis of the EMPATROPISM trial
.
Diabetes Metab Syndr
2022
;
16
:
102417
.

15.

Paulus
WJ
,
Tschöpe
C
.
A novel paradigm for heart failure with preserved ejection fraction: comorbidities drive myocardial dysfunction and remodeling through coronary microvascular endothelial inflammation
.
J Am Coll Cardiol
2013
;
62
:
263
271
.

16.

Park
S-H
,
Farooq
MA
,
Gaertner
S
,
Bruckert
C
,
Qureshi
AW
,
Lee
H-H
,
Benrahla
D
,
Pollet
B
,
Stephan
D
,
Ohlmann
P
,
Lessinger
J-M
,
Mayoux
E
,
Auger
C
,
Morel
O
,
Schini-Kerth
VB
.
Empagliflozin improved systolic blood pressure, endothelial dysfunction and heart remodeling in the metabolic syndrome ZSF1 rat
.
Cardiovasc Diabetol
2020
;
19
:
19
.

17.

Steven
S
,
Oelze
M
,
Hanf
A
,
Kröller-Schön
S
,
Kashani
F
,
Roohani
S
,
Welschof
P
,
Kopp
M
,
Gödtel-Armbrust
U
,
Xia
N
,
Li
H
,
Schulz
E
,
Lackner
KJ
,
Wojnowski
L
,
Bottari
SP
,
Wenzel
P
,
Mayoux
E
,
Münzel
T
,
Daiber
A
.
The SGLT2 inhibitor empagliflozin improves the primary diabetic complications in ZDF rats
.
Redox Biol
2017
;
13
:
370
385
.

18.

Oelze
M
,
Kröller-Schön
S
,
Welschof
P
,
Jansen
T
,
Hausding
M
,
Mikhed
Y
,
Stamm
P
,
Mader
M
,
Zinßius
E
,
Agdauletova
S
,
Gottschlich
A
,
Steven
S
,
Schulz
E
,
Bottari
SP
,
Mayoux
E
,
Münzel
T
,
Daiber
A
.
The sodium-glucose co-transporter 2 inhibitor empagliflozin improves diabetes-induced vascular dysfunction in the streptozotocin diabetes rat model by interfering with oxidative stress and glucotoxicity
.
PLoS One
2014
;
9
:
e112394
.

19.

Lee
DM
,
Battson
ML
,
Jarrell
DK
,
Hou
S
,
Ecton
KE
,
Weir
TL
,
Gentile
CL
.
SGLT2 inhibition via dapagliflozin improves generalized vascular dysfunction and alters the gut microbiota in type 2 diabetic mice
.
Cardiovasc Diabetol
2018
;
17
:
62
.

20.

Cai
C
,
Guo
Z
,
Chang
X
,
Li
Z
,
Wu
F
,
He
J
,
Cao
T
,
Wang
K
,
Shi
N
,
Zhou
H
,
Toan
S
,
Muid
D
,
Tan
Y
.
Empagliflozin attenuates cardiac microvascular ischemia/reperfusion through activating the AMPKα1/ULK1/FUNDC1/mitophagy pathway
.
Redox Biol
2022
;
52
:
102288
.

21.

Zhou
H
,
Wang
S
,
Zhu
P
,
Hu
S
,
Chen
Y
,
Ren
J
.
Empagliflozin rescues diabetic myocardial microvascular injury via AMPK-mediated inhibition of mitochondrial fission
.
Redox Biol
2018
;
15
:
335
346
.

22.

Uthman
L
,
Homayr
A
,
Juni
RP
,
Spin
EL
,
Kerindongo
R
,
Boomsma
M
,
Hollmann
MW
,
Preckel
B
,
Koolwijk
P
,
van Hinsbergh
VWM
,
Zuurbier
CJ
,
Albrecht
M
,
Weber
NC
.
Empagliflozin and Dapagliflozin reduce ROS generation and restore NO bioavailability in tumor necrosis factor α-stimulated Human coronary arterial endothelial cells
.
Cell Physiol Biochem
2019
;
53
:
865
886
.

23.

Zou
R
,
Shi
W
,
Qiu
J
,
Zhou
N
,
Du
N
,
Zhou
H
,
Chen
X
,
Ma
L
.
Empagliflozin attenuates cardiac microvascular ischemia/reperfusion injury through improving mitochondrial homeostasis
.
Cardiovasc Diabetol
2022
;
21
:
106
.

24.

Ma
L
,
Zou
R
,
Shi
W
,
Zhou
N
,
Chen
S
,
Zhou
H
,
Chen
X
,
Wu
Y
.
SGLT2 inhibitor dapagliflozin reduces endothelial dysfunction and microvascular damage during cardiac ischemia/reperfusion injury through normalizing the XO-SERCA2-CaMKII-coffilin pathways
.
Theranostics
2022
;
12
:
5034
5050
.

25.

Juni
RP
,
Kuster
DWD
,
Goebel
M
,
Helmes
M
,
Musters
RJP
,
Van Der Velden
J
,
Koolwijk
P
,
Paulus
WJ
,
Van Hinsbergh
VWM
.
Cardiac microvascular endothelial enhancement of cardiomyocyte function is impaired by inflammation and restored by Empagliflozin
.
JACC Basic Transl Sci
2019
;
4
:
575
591
.

26.

Juni
RP
,
Al-Shama
R
,
Kuster
DWD
,
Van Der Velden
J
,
Hamer
HM
,
Vervloet
MG
,
Eringa
EC
,
Koolwijk
P
,
Van Hinsbergh
VWM
.
Empagliflozin restores chronic kidney disease-induced impairment of endothelial regulation of cardiomyocyte relaxation and contraction
.
Kidney Int
2021
;
99
:
1088
1101
.

27.

Santos-Gallego
CG
,
Requena-Ibanez
JA
,
San Antonio
R
,
Ishikawa
K
,
Watanabe
S
,
Picatoste
B
,
Flores
E
,
Garcia-Ropero
A
,
Sanz
J
,
Hajjar
RJ
,
Fuster
V
,
Badimon
JJ
.
Empagliflozin ameliorates adverse left ventricular remodeling in nondiabetic heart failure by enhancing myocardial energetics
.
J Am Coll Cardiol
2019
;
73
:
1931
1944
.

28.

Santos-Gallego
CG
,
Mayr
M
,
Badimon
J
.
SGLT2 Inhibitors in heart failure: targeted metabolomics and energetic metabolism
.
Circulation
2022
;
146
:
819
821
.

29.

El-Daly
M
,
Pulakazhi Venu
VK
,
Saifeddine
M
,
Mihara
K
,
Kang
S
,
Fedak
PWM
,
Alston
LA
,
Hirota
SA
,
Ding
H
,
Triggle
CR
,
Hollenberg
MD
.
Hyperglycaemic impairment of PAR2-mediated vasodilation: prevention by inhibition of aortic endothelial sodium-glucose-co-transporter-2 and minimizing oxidative stress
.
Vascul Pharmacol
2018
;
109
:
56
71
.

30.

He
L
,
Li
Y
,
Zhang
D
,
Song
H
,
Xu
D
,
Song
Z
.
Dapagliflozin improves endothelial cell dysfunction by regulating mitochondrial production via the SIRT1/PGC-1α pathway in obese mice
.
Biochem Biophys Res Commun
2022
;
615
:
123
130
.

31.

Mone
P
,
Varzideh
F
,
Jankauskas
SS
,
Pansini
A
,
Lombardi
A
,
Frullone
S
,
Santulli
G
.
SGLT2 Inhibition via Empagliflozin improves endothelial function and reduces mitochondrial oxidative stress: insights from frail hypertensive and diabetic patients
.
Hypertension
2022
;
79
:
1633
1643
.

32.

Li
X
,
Preckel
B
,
Hermanides
J
,
Hollmann
MW
,
Zuurbier
CJ
,
Weber
NC
.
Amelioration of endothelial dysfunction by sodium glucose cotransporter 2 inhibitors: pieces of the puzzle explaining their cardiovascular protection
.
Br J Pharmacol
2022
;
179
:
4047
4062
.

33.

Li
X
,
Römer
G
,
Kerindongo
RP
,
Hermanides
J
,
Albrecht
M
,
Hollmann
MW
,
Zuurbier
CJ
,
Preckel
B
,
Weber
NC
.
Sodium glucose Co-transporter 2 inhibitors ameliorate endothelium barrier dysfunction induced by cyclic stretch through inhibition of reactive oxygen species
.
Int J Mol Sci
2021
;
22
:
6044
.

34.

Bruckert
C
,
Matsushita
K
,
Mroueh
A
,
Amissi
S
,
Auger
C
,
Houngue
U
,
Remila
L
,
Chaker
AB
,
Park
S-H
,
Algara-Suarez
P
,
Belcastro
E
,
Jesel
L
,
Ohlmann
P
,
Morel
O
,
Schini-Kerth
VB
.
Empagliflozin prevents angiotensin II-induced hypertension related micro and macrovascular endothelial cell activation and diastolic dysfunction in rats despite persistent hypertension: role of endothelial SGLT1 and 2
.
Vasc Pharmacol
2022
;
146
:
107095
.

35.

Nikolaou
PE
,
Efentakis
P
,
Abu Qourah
F
,
Femminò
S
,
Makridakis
M
,
Kanaki
Z
,
Varela
A
,
Tsoumani
M
,
Davos
CH
,
Dimitriou
CA
,
Tasouli
A
,
Dimitriadis
G
,
Kostomitsopoulos
N
,
Zuurbier
CJ
,
Vlahou
A
,
Klinakis
A
,
Brizzi
MF
,
Iliodromitis
EK
,
Andreadou
I
.
Chronic Empagliflozin treatment reduces myocardial infarct size in nondiabetic mice through STAT-3-mediated protection on microvascular endothelial cells and reduction of oxidative stress
.
Antioxid Redox Signal
2021
;
34
:
551
571
.

36.

Cappetta
D
,
De Angelis
A
,
Ciuffreda
LP
,
Coppini
R
,
Cozzolino
A
,
Miccichè
A
,
Dell'aversana
C
,
D'amario
D
,
Cianflone
E
,
Scavone
C
,
Santini
L
,
Palandri
C
,
Naviglio
S
,
Crea
F
,
Rota
M
,
Altucci
L
,
Rossi
F
,
Capuano
A
,
Urbanek
K
,
Berrino
L
.
Amelioration of diastolic dysfunction by dapagliflozin in a non-diabetic model involves coronary endothelium
.
Pharmacol Res
2020
;
157
:
104781
.

37.

Dyck
JRB
,
Sossalla
S
,
Hamdani
N
,
Coronel
R
,
Weber
NC
,
Light
PE
,
Zuurbier
CJ
.
Cardiac mechanisms of the beneficial effects of SGLT2 inhibitors in heart failure: evidence for potential off-target effects
.
J. Mol. Cell. Cardiol.
2022
;
167
:
17
31
.

38.

Ganbaatar
B
,
Fukuda
D
,
Shinohara
M
,
Yagi
S
,
Kusunose
K
,
Yamada
H
,
Soeki
T
,
Hirata
K-I
,
Sata
M
.
Empagliflozin ameliorates endothelial dysfunction and suppresses atherogenesis in diabetic apolipoprotein E-deficient mice
.
Eur J Pharmacol
2020
;
875
:
173040
.

39.

Gaspari
T
,
Spizzo
I
,
Liu
H
,
Hu
Y
,
Simpson
RW
,
Widdop
RE
,
Dear
AE
.
Dapagliflozin attenuates human vascular endothelial cell activation and induces vasorelaxation: a potential mechanism for inhibition of atherogenesis
.
Diab Vasc Dis Res
2018
;
15
:
64
73
.

40.

Cooper
S
,
Teoh
H
,
Campeau
MA
,
Verma
S
,
Leask
RL
.
Empagliflozin restores the integrity of the endothelial glycocalyx in vitro
.
Mol Cell Biochem
2019
;
459
:
121
130
.

41.

Campeau
M-A
,
Leask
RL
.
Empagliflozin mitigates endothelial inflammation and attenuates endoplasmic reticulum stress signaling caused by sustained glycocalyx disruption
.
Sci Rep
2022
;
12
:
12681
.

42.

Nijst
P
,
Verbrugge
FH
,
Grieten
L
,
Dupont
M
,
Steels
P
,
Tang
WHW
,
Mullens
W
.
The pathophysiological role of interstitial sodium in heart failure
.
J Am Coll Cardiol
2015
;
65
:
378
388
.

43.

Irace
C
,
Cutruzzolà
A
,
Parise
M
,
Fiorentino
R
,
Frazzetto
M
,
Gnasso
C
,
Casciaro
F
,
Gnasso
A
.
Effect of empagliflozin on brachial artery shear stress and endothelial function in subjects with type 2 diabetes: results from an exploratory study
.
Diab Vasc Dis Res
2020
;
17
:
1479164119883540
.

44.

Tanaka
A
,
Shimabukuro
M
,
Machii
N
,
Teragawa
H
,
Okada
Y
,
Shima
KR
,
Takamura
T
,
Taguchi
I
,
Hisauchi
I
,
Toyoda
S
,
Matsuzawa
Y
,
Tomiyama
H
,
Yamaoka-Tojo
M
,
Yoshida
H
,
Sato
Y
,
Ikehara
Y
,
Ueda
S
,
Higashi
Y
,
Node
K
,
Ako
J
,
Amano
H
,
Inoue
T
,
Jinnouchi
H
,
Kawaguchi
A
,
Kawano
Y
,
Kimura
K
,
Kurozumi
A
,
Kusumoto
T
,
Maruhashi
T
,
Misu
H
,
Nakamura
K
,
Narisawa
M
,
Nishi
J
,
Ota
T
,
Oyama
J-I
,
Sakuma
M
,
Shiina
K
,
Sugiyama
S
,
Suzuki
K
,
Takahashi
N
,
Takemoto
Y
,
Takeshita
Y
,
Tamaki
H
,
Tamura
A
,
Tanaka
K
,
Toita
T
,
Torimoto
K
,
Uehara
H
,
Uemura
F
,
Yamakawa
K
,
Yufu
K
.
Effect of empagliflozin on endothelial function in patients with type 2 diabetes and cardiovascular disease: results from the multicenter, randomized, placebo-controlled, double-blind EMBLEM trial
.
Diabetes Care
2019
;
42
:
e159
e161
.

45.

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
.

46.

Zainordin
NA
,
Hatta
SFWM
,
Mohamed Shah
FZ
,
Rahman
TA
,
Ismail
N
,
Ismail
Z
,
Abdul Ghani
R
.
Effects of Dapagliflozin on endothelial dysfunction in type 2 diabetes with established ischemic heart disease (EDIFIED)
.
J Endocr Soc
2019
;
4
:
bvz017
.

47.

Shigiyama
F
,
Kumashiro
N
,
Miyagi
M
,
Ikehara
K
,
Kanda
E
,
Uchino
H
,
Hirose
T
.
Effectiveness of dapagliflozin on vascular endothelial function and glycemic control in patients with early-stage type 2 diabetes mellitus: DEFENCE study
.
Cardiovasc Diabetol
2017
;
16
:
84
.

48.

Solini
A
,
Giannini
L
,
Seghieri
M
,
Vitolo
E
,
Taddei
S
,
Ghiadoni
L
,
Bruno
RM
.
Dapagliflozin acutely improves endothelial dysfunction, reduces aortic stiffness and renal resistive index in type 2 diabetic patients: a pilot study
.
Cardiovasc Diabetol
2017
;
16
:
138
.

49.

Solini
A
,
Seghieri
M
,
Giannini
L
,
Biancalana
E
,
Parolini
F
,
Rossi
C
,
Dardano
A
,
Taddei
S
,
Ghiadoni
L
,
Bruno
RM
.
The effects of Dapagliflozin on systemic and renal vascular function display an epigenetic signature
.
J Clin Endocrinol Metab
2019
;
104
:
4253
4263
.

50.

Sugiyama
S
,
Jinnouchi
H
,
Kurinami
N
,
Hieshima
K
,
Yoshida
A
,
Jinnouchi
K
,
Nishimura
H
,
Suzuki
T
,
Miyamoto
F
,
Kajiwara
K
,
Jinnouchi
T
.
The SGLT2 inhibitor Dapagliflozin significantly improves the peripheral microvascular endothelial function in patients with uncontrolled type 2 diabetes mellitus
.
Intern Med
2018
;
57
:
2147
2156
.

51.

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
.

52.

Jürgens
M
,
Schou
M
,
Hasbak
P
,
Kjær
A
,
Wolsk
E
,
Zerahn
B
,
Wiberg
M
,
Brandt‐Jacobsen
NH
,
Gæde
P
,
Rossing
P
,
Faber
J
,
Inzucchi
SE
,
Gustafsson
F
,
Kistorp
C
.
Effects of empagliflozin on myocardial flow reserve in patients with type 2 diabetes mellitus: the SIMPLE trial
.
J Am Heart Assoc
2021
;
10
:
e020418
.

53.

Lauritsen
KM
,
Nielsen
BRR
,
Tolbod
LP
,
Johannsen
M
,
Hansen
J
,
Hansen
TK
,
Wiggers
H
,
Møller
N
,
Gormsen
LC
,
Søndergaard
E
.
SGLT2 Inhibition does not affect myocardial fatty acid oxidation or uptake, but reduces myocardial glucose uptake and blood flow in individuals with type 2 Diabetes: a randomized double-blind, placebo-controlled crossover trial
.
Diabetes
2021
;
70
:
800
808
.

54.

Suhrs
HE
,
Nilsson
M
,
Bové
KB
,
Zander
M
,
Prescott
E
.
Effect of empagliflozin on coronary microvascular function in patients with type 2 diabetes mellitus-A randomized, placebo-controlled cross-over study
.
PLoS One
2022
;
17
:
e0263481
.

55.

Leccisotti
L
,
Cinti
F
,
Sorice
GP
,
D'amario
D
,
Lorusso
M
,
Guzzardi
MA
,
Mezza
T
,
Gugliandolo
S
,
Cocchi
C
,
Capece
U
,
Indovina
L
,
Ferraro
PM
,
Iozzo
P
,
Crea
F
,
Giordano
A
,
Giaccari
A
.
Dapagliflozin improves myocardial flow reserve in patients with type 2 diabetes: the DAPAHEART Trial: a preliminary report
.
Cardiovasc Diabetol
2022
;
21
:
173
.

56.

Requena-Ibáñez
JA
,
Santos-Gallego
CG
,
Rodriguez-Cordero
A
,
Vargas-Delgado
AP
,
Mancini
D
,
Sartori
S
,
Atallah-Lajam
F
,
Giannarelli
C
,
Macaluso
F
,
Lala
A
,
Sanz
J
,
Fuster
V
,
Badimon
JJ
.
Mechanistic insights of Empagliflozin in nondiabetic patients with HFrEF: from the EMPA-TROPISM study
.
JACC Heart Fail
2021
;
9
:
578
589
.

57.

Chilton
R
,
Tikkanen
I
,
Cannon
CP
,
Crowe
S
,
Woerle
HJ
,
Broedl
UC
,
Johansen
OE
.
Effects of empagliflozin on blood pressure and markers of arterial stiffness and vascular resistance in patients with type 2 diabetes
.
Diabetes Obes Metab
2015
;
17
:
1180
1193
.

58.

Santos-Gallego
CG
,
Requena-Ibáñez
JA
,
Picatoste
B
,
Fardman
B
,
Ishikawa
K
,
Mazurek
R
,
Pieper
M
,
Sartori
S
,
Rodriguez-Capitán
J
,
Fuster
V
,
Badimon
JJ
.
Cardioprotective effect of empagliflozin and circulating ketone bodies during acute myocardial infarction
.
Circ Cardiovasc Imaging
2023
;
16
:
e015298
.

59.

Mcdonagh
TA
,
Metra
M
,
Adamo
M
,
Gardner
RS
,
Baumbach
A
,
Böhm
M
,
Burri
H
,
Butler
J
,
Čelutkienė
J
,
Chioncel
O
,
Cleland
JGF
,
Coats
AJS
,
Crespo-Leiro
MG
,
Farmakis
D
,
Gilard
M
,
Heymans
S
,
Hoes
AW
,
Jaarsma
T
,
Jankowska
EA
,
Lainscak
M
,
Lam
CSP
,
Lyon
AR
,
Mcmurray
JJV
,
Mebazaa
A
,
Mindham
R
,
Muneretto
C
,
Francesco Piepoli
M
,
Price
S
,
Rosano
GMC
,
Ruschitzka
F
,
Kathrine Skibelund
A
,
De Boer
RA
,
Christian Schulze
P
,
Abdelhamid
M
,
Aboyans
V
,
Adamopoulos
S
,
Anker
SD
,
Arbelo
E
,
Asteggiano
R
,
Bauersachs
J
,
Bayes-Genis
A
,
Borger
MA
,
Budts
W
,
Cikes
M
,
Damman
K
,
Delgado
V
,
Dendale
P
,
Dilaveris
P
,
Drexel
H
,
Ezekowitz
J
,
Falk
V
,
Fauchier
L
,
Filippatos
G
,
Fraser
A
,
Frey
N
,
Gale
CP
,
Gustafsson
F
,
Harris
J
,
Iung
B
,
Janssens
S
,
Jessup
M
,
Konradi
A
,
Kotecha
D
,
Lambrinou
E
,
Lancellotti
P
,
Landmesser
U
,
Leclercq
C
,
Lewis
BS
,
Leyva
F
,
Linhart
A
,
Løchen
M-L
,
Lund
LH
,
Mancini
D
,
Masip
J
,
Milicic
D
,
Mueller
C
,
Nef
H
,
Nielsen
J-C
,
Neubeck
L
,
Noutsias
M
,
Petersen
SE
,
Sonia Petronio
A
,
Ponikowski
P
,
Prescott
E
,
Rakisheva
A
,
Richter
DJ
,
Schlyakhto
E
,
Seferovic
P
,
Senni
M
,
Sitges
M
,
Sousa-Uva
M
,
Tocchetti
CG
,
Touyz
RM
,
Tschoepe
C
,
Waltenberger
J
,
Adamo
M
,
Baumbach
A
,
Böhm
M
,
Burri
H
,
Čelutkienė
J
,
Chioncel
O
,
Cleland
JGF
,
Coats
AJS
,
Crespo-Leiro
MG
,
Farmakis
D
,
Gardner
RS
,
Gilard
M
,
Heymans
S
,
Hoes
AW
,
Jaarsma
T
,
Jankowska
EA
,
Lainscak
M
,
Lam
CSP
,
Lyon
AR
,
Mcmurray
JJV
,
Mebazaa
A
,
Mindham
R
,
Muneretto
C
,
Piepoli
MF
,
Price
S
,
Rosano
GMC
,
Ruschitzka
F
,
Skibelund
AK
.
2021 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure [published correction appears in Eur Heart J. 2021 Oct 14]
.
Eur Heart J
2021
;
42
:
3599
3726
.

60.

Rush
CJ
,
Berry
C
,
Oldroyd
KG
,
Rocchiccioli
JP
,
Lindsay
MM
,
Touyz
RM
,
Murphy
CL
,
Ford
TJ
,
Sidik
N
,
Mcentegart
MB
,
Lang
NN
,
Jhund
PS
,
Campbell
RT
,
Mcmurray
JJV
,
Petrie
MC
.
Prevalence of coronary artery disease and coronary microvascular dysfunction in patients with heart failure with preserved ejection fraction
.
JAMA Cardiol
2021
;
6
:
1130
1143
.

61.

Löffler
AI
,
Pan
JA
,
Balfour
PC
,
Shaw
PW
,
Yang
Y
,
Nasir
M
,
Auger
DA
,
Epstein
FH
,
Kramer
CM
,
Gan
L-M
,
Salerno
M
.
Frequency of coronary microvascular dysfunction and diffuse myocardial fibrosis (Measured by Cardiovascular Magnetic Resonance) in patients with heart failure and preserved left ventricular ejection fraction
.
Am J Cardiol
2019
;
124
:
1584
1589
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)