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

Matthias Oelze, Swenja Kröller-Schön, Philipp Welschof, Thomas Jansen, Michael Hausding, Yuliya Mikhed, Paul Stamm, Michael Mader, Elena Zinßius, Saule Agdauletova, Anna Gottschlich, Sebastian Steven, Eberhard Schulz, Serge P Bottari, Eric Mayoux, Thomas Münzel, Andreas Daiber, Matthias Oelze, Swenja Kröller-Schön, Philipp Welschof, Thomas Jansen, Michael Hausding, Yuliya Mikhed, Paul Stamm, Michael Mader, Elena Zinßius, Saule Agdauletova, Anna Gottschlich, Sebastian Steven, Eberhard Schulz, Serge P Bottari, Eric Mayoux, Thomas Münzel, Andreas Daiber

Abstract

Objective: In diabetes, vascular dysfunction is characterized by impaired endothelial function due to increased oxidative stress. Empagliflozin, as a selective sodium-glucose co-transporter 2 inhibitor (SGLT2i), offers a novel approach for the treatment of type 2 diabetes by enhancing urinary glucose excretion. The aim of the present study was to test whether treatment with empagliflozin improves endothelial dysfunction in type I diabetic rats via reduction of glucotoxicity and associated vascular oxidative stress.

Methods: Type I diabetes in Wistar rats was induced by an intravenous injection of streptozotocin (60 mg/kg). One week after injection empagliflozin (10 and 30 mg/kg/d) was administered via drinking water for 7 weeks. Vascular function was assessed by isometric tension recording, oxidative stress parameters by chemiluminescence and fluorescence techniques, protein expression by Western blot, mRNA expression by RT-PCR, and islet function by insulin ELISA in serum and immunohistochemical staining of pancreatic tissue. Advanced glycation end products (AGE) signaling was assessed by dot blot analysis and mRNA expression of the AGE-receptor (RAGE).

Results: Treatment with empagliflozin reduced blood glucose levels, normalized endothelial function (aortic rings) and reduced oxidative stress in aortic vessels (dihydroethidium staining) and in blood (phorbol ester/zymosan A-stimulated chemiluminescence) of diabetic rats. Additionally, the pro-inflammatory phenotype and glucotoxicity (AGE/RAGE signaling) in diabetic animals was reversed by SGLT2i therapy.

Conclusions: Empagliflozin improves hyperglycemia and prevents the development of endothelial dysfunction, reduces oxidative stress and improves the metabolic situation in type 1 diabetic rats. These preclinical observations illustrate the therapeutic potential of this new class of antidiabetic drugs.

Conflict of interest statement

Competing Interests: T.M. and A.D. received research grants from Boehringer Ingelheim Pharma GmbH & Co. KG. E.M. is an employee of Boehringer Ingelheim Pharma GmbH & Co. KG. The remaining authors declare that they have no competing interests in connection with this manuscript. This does not alter the authors' adherence to PLOS ONE policies on sharing data and materials.

Figures

Figure 1. Glucagon and insulin measurements in…
Figure 1. Glucagon and insulin measurements in controls and diabetic rats 8 weeks after STZ injection and 7 weeks of SGLT2i (low and high dose) treatment.
Representative immunohistochemical stainings of pancreatic tissue for glucagon (A), insulin (B) and ELISA (C) or RIA (D) based determination of insulin serum levels. The data are the means ±SEM of 3–4 (A,B), 3–5 (C) or 9–11 (D) animals/group. *, p<0.05 vs. control and #, p<0.05 vs. STZ-injected group.
Figure 2. Effects of SGLT2i treatment on…
Figure 2. Effects of SGLT2i treatment on vascular parameters in diabetic rats.
Microscopic determination of wall thickness (grey) and collagen content (black) by sirius red staining of paraffinated aortic sections (A). Representative microscope images are shown along with the densitometric quantification. Effects of SGLT2i therapy on endothelium-dependent and independent vascular relaxation by the vasodilators acetylcholine (ACh, B) and nitroglycerin (GTN, C), respectively. Data are the means±SEM from 6–7 (A) and 9–12 (B,C) animals/group. Each single value for an animal corresponds to the means of 4 individual aortic rings from this animal. *, p<0.05 vs. control and #, p<0.05 vs. STZ-injected group. For the vascular function data the significance levels were determined by two-way-ANOVA and significances for the entire curves are indicated when at least one compared concentration condition showed significant differences. Vasodilator potency (EC50, pD2) and efficacy (max. relaxation) were also calculated and subjected to statistical analysis using one-way-ANOVA. The data and results are presented in Table 1.
Figure 3. Effects of SGLT2i treatment on…
Figure 3. Effects of SGLT2i treatment on oxidative stress parameters in diabetic rats.
Leukocyte-derived ROS (oxidative burst) in whole blood at 30 min upon zymosan A (A) stimulation along with the effects of the NADPH oxidase isoform 2 (Nox2) inhibitor VAS2870 and the intracellular calcium chelator BAPTA-AM (B) and upon PDBu stimulation (C). (D) Quantification of cardiac NADPH oxidase activity in membrane preparations by lucigenin (5 µM)-derived chemiluminescence. Dihydroethidium (DHE, 1 µM)-fluorescence microtopography was used to assess the effects of SGLT2i treatment on vascular (E) and endothelial (F) ROS production with and without incubation with the eNOS inhibitor L-NAME. Representative microscope images are shown along with the densitometric quantification. Red fluorescence indicates ROS formation whereas green fluorescence represents basal laminae autofluorescence. Data are the means±SEM from 6–7 (A,B), 5 (C), 7–8 (D), 9 (E) or 5–6 (F) animals/group. *, p<0.05 vs. control and #, p<0.05 vs. STZ-injected and §, p<0.05 vs. low dose SGLT2i treated and $, p<0.05 vs. w/o L-NAME.
Figure 4. Effects of SGLT2i treatment on…
Figure 4. Effects of SGLT2i treatment on aortic protein expression of the NO/cGMP signaling cascade as well as oxidative stress and inflammatory pathways in diabetic rats.
Expression of endothelial nitric oxide synthase (eNOS, A), serine1177 phosphorylated eNOS (B), dihydrofolate reductase (DHFR, C), ratio of cGK-I and serine239 phosphorylated VASP (D) were assessed by Western blotting analysis and specific antibodies. Expression of NADPH oxidases Nox1 (E) and Nox2 (F), heme oxygenase-1 (HO-1) (G) and monocyte-chemoattractant-protein-1 (MCP-1 or CCL-2, H) were assessed by Western blotting analysis and specific antibodies. Representative blots for all proteins are shown in supplemental Figure S6 in File S1. The data are expressed as % of control and are the means ± SEM from 8–9 (A), 5–6 (B), 7 (C), 4 (D), 6–7 (E), 7–9 (F), 7–9 (G) and 4–6 (H) animals/group. *, p<0.05 vs. control and #, p<0.05 vs. STZ-injected and $, p<0.05 vs. low dose SGLT2i treated.
Figure 5. Effects of SGLT2i treatment on…
Figure 5. Effects of SGLT2i treatment on AGE/RAGE signaling in diabetic rats.
Quantification of AGE-positive proteins by dot blot analysis (A) and RAGE expression was assessed by Western blotting analysis with specific antibodies (B) and quantitative RT-PCR analysis (C). Representative blots are shown at the bottom of the densitometric quantifications. Serum methylglyoxal levels were assessed by HPLC-based quantification (D). Representative chromatograms are shown in supplemental Figure S7 in File S1. The data are expressed as % of control and are the means ±SEM from 7 (A), 6–7 (B) and 8–11 (C,D) animals/group. *, p<0.05 vs. control and #, p<0.05 vs. STZ–injected.
Figure 6. Effects of SGLT2i treatment on…
Figure 6. Effects of SGLT2i treatment on aortic mRNA expression of pro-inflammatory genes in diabetic rats.
Expression of the monocyte chemoattractant protein-1 (CCL-2, A), the monocyte/macrophage-specific protein CD68 (B), the cytokine interleukin-6 (IL-6, C), the immune-signaling proteins interferon-γ (IFN-γ, D) and tumor necrosis factor-α (TNF-α, E) and the intercellular adhesion molecule-1 (ICAM-1, F) was assessed by quantitative RT-PCR. The data are expressed as % of control and are the means ±SEM from 7–9 (AC), 9–10 (DE) and 6–7 animals/group. *, p<0.05 vs. control and #, p<0.05 vs. STZ-injected.
Figure 7. Summary of beneficial effects of…
Figure 7. Summary of beneficial effects of SGLT2i treatment on diabetes induced vascular dysfunction and other adverse effects of hyperglycemia in rats.

References

    1. Nathan DM, Cleary PA, Backlund JY, Genuth SM, Lachin JM, et al. (2005) Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes. N Engl J Med 353: 2643–2653.
    1. Jay D, Hitomi H, Griendling KK (2006) Oxidative stress and diabetic cardiovascular complications. Free Radic Biol Med 40: 183–192.
    1. Hink U, Li H, Mollnau H, Oelze M, Matheis E, et al. (2001) Mechanisms Underlying Endothelial Dysfunction in Diabetes Mellitus. Circ Res 88: E14–E22.
    1. Wenzel P, Daiber A, Oelze M, Brandt M, Closs E, et al. (2008) Mechanisms underlying recoupling of eNOS by HMG-CoA reductase inhibition in a rat model of streptozotocin-induced diabetes mellitus. Atherosclerosis 198: 65–76.
    1. Wenzel P, Schulz E, Oelze M, Muller J, Schuhmacher S, et al. (2008) AT1-receptor blockade by telmisartan upregulates GTP-cyclohydrolase I and protects eNOS in diabetic rats. Free Radic Biol Med 45: 619–626.
    1. Schuhmacher S, Oelze M, Bollmann F, Kleinert H, Otto C, et al. (2011) Vascular Dysfunction in Experimental Diabetes Is Improved by Pentaerithrityl Tetranitrate but not Isosorbide-5-Mononitrate Therapy. Diabetes 60: 2608–2616.
    1. Lin MI, Fulton D, Babbitt R, Fleming I, Busse R, et al. (2003) Phosphorylation of threonine 497 in endothelial nitric-oxide synthase coordinates the coupling of L-arginine metabolism to efficient nitric oxide production. J Biol Chem 278: 44719–44726.
    1. Loot AE, Schreiber JG, Fisslthaler B, Fleming I (2009) Angiotensin II impairs endothelial function via tyrosine phosphorylation of the endothelial nitric oxide synthase. J Exp Med 206: 2889–2896.
    1. Forbes JM, Cooper ME, Thallas V, Burns WC, Thomas MC, et al. (2002) Reduction of the accumulation of advanced glycation end products by ACE inhibition in experimental diabetic nephropathy. Diabetes 51: 3274–3282.
    1. Yamamoto Y, Yamagishi S, Yonekura H, Doi T, Tsuji H, et al. (2000) Roles of the AGE-RAGE system in vascular injury in diabetes. Ann N Y Acad Sci 902: 163–170 discussion 170–162.
    1. Coughlan MT, Thorburn DR, Penfold SA, Laskowski A, Harcourt BE, et al. (2009) RAGE-induced cytosolic ROS promote mitochondrial superoxide generation in diabetes. J Am Soc Nephrol 20: 742–752.
    1. Wautier MP, Chappey O, Corda S, Stern DM, Schmidt AM, et al. (2001) Activation of NADPH oxidase by AGE links oxidant stress to altered gene expression via RAGE. Am J Physiol Endocrinol Metab 280: E685–694.
    1. Nishikawa T, Edelstein D, Du XL, Yamagishi S, Matsumura T, et al. (2000) Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage. Nature 404: 787–790.
    1. Schulz E, Wenzel P, Munzel T, Daiber A (2014) Mitochondrial redox signaling: Interaction of mitochondrial reactive oxygen species with other sources of oxidative stress. Antioxid Redox Signal 20: 308–324.
    1. Kroller-Schon S, Steven S, Kossmann S, Scholz A, Daub S, et al. (2014) Molecular mechanisms of the crosstalk between mitochondria and NADPH oxidase through reactive oxygen species-studies in white blood cells and in animal models. Antioxid Redox Signal 20: 247–266.
    1. Wenzel P, Knorr M, Kossmann S, Stratmann J, Hausding M, et al. (2011) Lysozyme M-positive monocytes mediate angiotensin II-induced arterial hypertension and vascular dysfunction. Circulation 124: 1370–1381.
    1. Rieg T, Masuda T, Gerasimova M, Mayoux E, Platt K, et al. (2014) Increase in SGLT1-mediated transport explains renal glucose reabsorption during genetic and pharmacological SGLT2 inhibition in euglycemia. Am J Physiol Renal Physiol 306: F188–193.
    1. Evans JL, Goldfine ID (2013) Aging and insulin resistance: just say iNOS. Diabetes 62: 346–348.
    1. Sarashina A, Koiwai K, Seman LJ, Yamamura N, Taniguchi A, et al. (2013) Safety, tolerability, pharmacokinetics and pharmacodynamics of single doses of empagliflozin, a sodium glucose cotransporter 2 (SGLT2) inhibitor, in healthy Japanese subjects. Drug Metab Pharmacokinet 28: 213–219.
    1. Ferrannini E, Seman L, Seewaldt-Becker E, Hantel S, Pinnetti S, et al. (2013) A Phase IIb, randomized, placebo-controlled study of the SGLT2 inhibitor empagliflozin in patients with type 2 diabetes. Diabetes Obes Metab 15: 721–728.
    1. Hansen HH, Jelsing J, Hansen CF, Hansen G, Vrang N, et al. (2014) The sodium glucose cotransporter type 2 inhibitor empagliflozin preserves beta-cell mass and restores glucose homeostasis in the male zucker diabetic fatty rat. J Pharmacol Exp Ther 350: 657–664.
    1. Luippold G, Klein T, Mark M, Grempler R (2012) Empagliflozin, a novel potent and selective SGLT-2 inhibitor, improves glycaemic control alone and in combination with insulin in streptozotocin-induced diabetic rats, a model of type 1 diabetes mellitus. Diabetes Obes Metab 14: 601–607.
    1. Grempler R, Thomas L, Eckhardt M, Himmelsbach F, Sauer A, et al. (2012) Empagliflozin, a novel selective sodium glucose cotransporter-2 (SGLT-2) inhibitor: characterisation and comparison with other SGLT-2 inhibitors. Diabetes Obes Metab 14: 83–90.
    1. Vickers SP, Cheetham SC, Headland KR, Dickinson K, Grempler R, et al. (2014) Combination of the sodium-glucose cotransporter-2 inhibitor empagliflozin with orlistat or sibutramine further improves the body-weight reduction and glucose homeostasis of obese rats fed a cafeteria diet. Diabetes Metab Syndr Obes 7: 265–275.
    1. Oelze M, Knorr M, Schuhmacher S, Heeren T, Otto C, et al. (2011) Vascular dysfunction in streptozotocin-induced experimental diabetes strictly depends on insulin deficiency. J Vasc Res 48: 275–284.
    1. Oelze M, Kroller-Schon S, Steven S, Lubos E, Doppler C, et al... (2013) Glutathione Peroxidase-1 Deficiency Potentiates Dysregulatory Modifications of Endothelial Nitric Oxide Synthase and Vascular Dysfunction in Aging. Hypertension.
    1. Daiber A, Oelze M, Coldewey M, Bachschmid M, Wenzel P, et al. (2004) Oxidative stress and mitochondrial aldehyde dehydrogenase activity: a comparison of pentaerythritol tetranitrate with other organic nitrates. Mol Pharmacol 66: 1372–1382.
    1. Kroller-Schon S, Knorr M, Hausding M, Oelze M, Schuff A, et al. (2012) Glucose-independent improvement of vascular dysfunction in experimental sepsis by dipeptidyl-peptidase 4 inhibition. Cardiovasc Res 96: 140–149.
    1. Oelze M, Daiber A, Brandes RP, Hortmann M, Wenzel P, et al. (2006) Nebivolol inhibits superoxide formation by NADPH oxidase and endothelial dysfunction in angiotensin II-treated rats. Hypertension 48: 677–684.
    1. Shiryaeva YK, Krylin VV, Titov VN (2012) Chromatographic determination of methyl glyoxal in blood plasma as the test for glycotoxicity and accumulation of glycation end-products. Bull Exp Biol Med 153: 114–117.
    1. Wenzel P, Hink U, Oelze M, Seeling A, Isse T, et al. (2007) Number of nitrate groups determines reactivity and potency of organic nitrates: a proof of concept study in ALDH-2-/- mice. Brit J Pharmacol 150: 526–533.
    1. Hausding M, Jurk K, Daub S, Kroller-Schon S, Stein J, et al. (2013) CD40L contributes to angiotensin II-induced pro-thrombotic state, vascular inflammation, oxidative stress and endothelial dysfunction. Basic Res Cardiol 108: 386.
    1. Pricci F, Leto G, Amadio L, Iacobini C, Cordone S, et al. (2003) Oxidative stress in diabetes-induced endothelial dysfunction involvement of nitric oxide and protein kinase C. Free Radic Biol Med. 35: 683–694.
    1. Yu Y, Lyons TJ (2005) A lethal tetrad in diabetes: hyperglycemia, dyslipidemia, oxidative stress, and endothelial dysfunction. Am J Med Sci 330: 227–232.
    1. Ferrannini E, Muscelli E, Frascerra S, Baldi S, Mari A, et al. (2014) Metabolic response to sodium-glucose cotransporter 2 inhibition in type 2 diabetic patients. J Clin Invest 124: 499–508.
    1. Perkins BA, Cherney DZ, Partridge H, Soleymanlou N, Tschirhart H, et al. (2014) Sodium-Glucose Cotransporter 2 Inhibition and Glycemic Control in Type 1 Diabetes: Results of an 8-Week Open-Label Proof-of-Concept Trial. Diabetes Care 37: 1480–1483.
    1. Cherney DZ, Perkins BA, Soleymanlou N, Har R, Fagan N, et al. (2014) The effect of empagliflozin on arterial stiffness and heart rate variability in subjects with uncomplicated type 1 diabetes mellitus. Cardiovasc Diabetol 13: 28.
    1. Bierhaus A, Fleming T, Stoyanov S, Leffler A, Babes A, et al. (2012) Methylglyoxal modification of Nav1.8 facilitates nociceptive neuron firing and causes hyperalgesia in diabetic neuropathy. Nat Med 18: 926–933.
    1. Sun M, Yokoyama M, Ishiwata T, Asano G (1998) Deposition of advanced glycation end products (AGE) and expression of the receptor for AGE in cardiovascular tissue of the diabetic rat. Int J Exp Pathol 79: 207–222.
    1. Wautier JL, Wautier MP, Schmidt AM, Anderson GM, Hori O, et al. (1994) Advanced glycation end products (AGEs) on the surface of diabetic erythrocytes bind to the vessel wall via a specific receptor inducing oxidant stress in the vasculature: a link between surface-associated AGEs and diabetic complications. Proc Natl Acad Sci U S A 91: 7742–7746.
    1. Jeong KH, Lee TW, Ihm CG, Lee SH, Moon JY, et al. (2009) Effects of sildenafil on oxidative and inflammatory injuries of the kidney in streptozotocin-induced diabetic rats. Am J Nephrol 29: 274–282.
    1. Kumar S, Prasad S, Sitasawad SL (2013) Multiple antioxidants improve cardiac complications and inhibit cardiac cell death in streptozotocin-induced diabetic rats. PLoS One 8: e67009.
    1. Chen Z, Zhang J, Stamler JS (2002) Identification of the enzymatic mechanism of nitroglycerin bioactivation. Proc Natl Acad Sci U S A 99: 8306–8311.
    1. Sydow K, Daiber A, Oelze M, Chen Z, August M, et al. (2004) Central role of mitochondrial aldehyde dehydrogenase and reactive oxygen species in nitroglycerin tolerance and cross-tolerance. J Clin Invest 113: 482–489.
    1. Oelze M, Knorr M, Schell R, Kamuf J, Pautz A, et al. (2011) Regulation of human mitochondrial aldehyde dehydrogenase (ALDH-2) activity by electrophiles in vitro. J Biol Chem 286: 8893–8900.
    1. Wenzel P, Hink U, Oelze M, Schuppan S, Schaeuble K, et al. (2007) Role of reduced lipoic acid in the redox regulation of mitochondrial aldehyde dehydrogenase (ALDH-2) activity. Implications for mitochondrial oxidative stress and nitrate tolerance. J Biol Chem 282: 792–799.
    1. Chen CH, Budas GR, Churchill EN, Disatnik MH, Hurley TD, et al. (2008) Activation of aldehyde dehydrogenase-2 reduces ischemic damage to the heart. Science 321: 1493–1495.
    1. Wang J, Wang H, Hao P, Xue L, Wei S, et al. (2011) Inhibition of aldehyde dehydrogenase 2 by oxidative stress is associated with cardiac dysfunction in diabetic rats. Mol Med 17: 172–179.
    1. Osar Z, Samanci T, Demirel GY, Damci T, Ilkova H (2004) Nicotinamide effects oxidative burst activity of neutrophils in patients with poorly controlled type 2 diabetes mellitus. Exp Diabesity Res 5: 155–162.
    1. Chang FY, Shaio MF (1995) Respiratory burst activity of monocytes from patients with non-insulin-dependent diabetes mellitus. Diabetes Res Clin Pract 29: 121–127.

Source: PubMed

3
Abonner