1. Academic Validation
  2. Empagliflozin and Dapagliflozin Reduce ROS Generation and Restore NO Bioavailability in Tumor Necrosis Factor α-Stimulated Human Coronary Arterial Endothelial Cells

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(5):865-886. doi: 10.33594/000000178.
Laween Uthman 1 Anna Homayr 1 2 Rio P Juni 3 Eva L Spin 1 Raphaela Kerindongo 1 Marleen Boomsma 1 Markus W Hollmann 1 Benedikt Preckel 1 Pieter Koolwijk 3 Victor W M van Hinsbergh 3 Coert J Zuurbier 1 Martin Albrecht 2 Nina C Weber 4
Affiliations

Affiliations

  • 1 Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam UMC, location Academic Medical Centre (AMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands.
  • 2 Department of Anesthesiology and Intensive Care Medicine, UKSH, Campus Kiel, Kiel, Germany.
  • 3 Department of Physiology, Amsterdam UMC, location VU medical center (VUMC), VU University, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands.
  • 4 Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam UMC, location Academic Medical Centre (AMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands, [email protected].
Abstract

Background/aims: Heart failure is characterized by chronic low-grade vascular inflammation, which in itself can lead to endothelial dysfunction. Clinical trials showed reductions in heart failure-related hospitalizations of type 2 diabetic patients using sodium glucose co-transporter 2 inhibitors (SGLT2i's). Whether and how SGLT2i's directly affect the endothelium under inflammatory conditions is not completely understood. The aim of the study was to investigate whether the SGLT2i Empagliflozin (EMPA) and Dapagliflozin (DAPA) reduce tumor necrosis factor α (TNFα) induced endothelial inflammation in vitro.

Methods: Human coronary arterial endothelial cells (HCAECs) and human umbilical vein endothelial cells (HUVECs) were (pre-)incubated with 1 µM EMPA or DAPA and subsequently exposed to 10 ng/ml TNFα. ROS and NO were measured using live cell imaging. Target proteins were either determined by infrared western blotting or fluorescence activated cell sorting (FACS). The connection between Cav-1 and eNOS was determined by co-immunoprecipitation.

Results: Nitric oxide (NO) bioavailability was reduced by TNFα and both EMPA and DAPA restored NO levels in TNFα-stimulated HCAECs. Intracellular ROS was increased by TNFα, and this increase was completely abolished by EMPA and DAPA in HCAECs by means of live cell imaging. eNOS signaling was significantly disturbed after 24 h when cells were exposed to TNFα for 24h, yet the presence of both SGLT2is did not prevent this disruption. TNFα-induced enhanced permeability at t=24h was unaffected in HUVECs by EMPA. Similarly, adhesion molecule expression (VCAM-1 and ICAM-1) was elevated after 4h TNFα (1.5-5.5 fold increase of VCAM-1 and 4-12 fold increase of ICAM-1) but were unaffected by EMPA and DAPA in both cell types. Although we detected expression of SGLT2 protein levels, the fact that we could not silence this expression by means of siRNA and the mRNA levels of SGLT2 were not detectable in HCAECs, suggests aspecificity or our SGLT2 antibody and absence of SGLT2 in our cells.

Conclusion: These data suggest that EMPA and DAPA rather restore NO bioavailability by inhibiting ROS generation than by affecting eNOS expression or signaling, barrier function and adhesion molecules expression in TNFα-induced endothelial cells. Furthermore, the observed effects cannot be ascribed to the inhibition of SGLT2 in endothelial cells.

Keywords

SGLT2 inhibitors; Inflammation; Endothelial cell; Empagliflozin; TNFα.

Figures
Products