Empagliflozin rescues diabetic myocardial microvascular injury via AMPK-mediated inhibition of mitochondrial fission

Hao Zhou, Shuyi Wang, Pingjun Zhu, Shunying Hu, Yundai Chen, Jun Ren, Hao Zhou, Shuyi Wang, Pingjun Zhu, Shunying Hu, Yundai Chen, Jun Ren

Abstract

Impaired cardiac microvascular function contributes to diabetic cardiovascular complications although effective therapy remains elusive. Empagliflozin, a sodium-glucose cotransporter 2 (SGLT2) inhibitor recently approved for treatment of type 2 diabetes, promotes glycosuria excretion and offers cardioprotective actions beyond its glucose-lowering effects. This study was designed to evaluate the effect of empagliflozin on cardiac microvascular injury in diabetes and the underlying mechanism involved with a focus on mitochondria. Our data revealed that empagliflozin improved diabetic myocardial structure and function, preserved cardiac microvascular barrier function and integrity, sustained eNOS phosphorylation and endothelium-dependent relaxation, as well as improved microvessel density and perfusion. Further study suggested that empagliflozin exerted its effects through inhibition of mitochondrial fission in an adenosine monophosphate (AMP)-activated protein kinase (AMPK)-dependent manner. Empagliflozin restored AMP-to-ATP ratio to trigger AMPK activation, suppressed Drp1S616 phosphorylation, and increased Drp1S637 phosphorylation, ultimately leading to inhibition of mitochondrial fission. The empagliflozin-induced inhibition of mitochondrial fission preserved cardiac microvascular endothelial cell (CMEC) barrier function through suppressed mitochondrial reactive oxygen species (mtROS) production and subsequently oxidative stress to impede CMEC senescence. Empagliflozin-induced fission loss also favored angiogenesis by promoting CMEC migration through amelioration of F-actin depolymerization. Taken together, these results indicated the therapeutic promises of empagliflozin in the treatment of pathological microvascular changes in diabetes.

Keywords: AMPK; CMECs; Empagliflozin; Microvascular; Mitochondrial fission.

Copyright © 2017 The Authors. Published by Elsevier B.V. All rights reserved.

Figures

Fig. 1
Fig. 1
Effect of empagliflozin treatment on cardiac function and morphology in diabetes: Experimental diabetes was induced with STZ and mice were treated with empagliflozin (10 mg/kg/d) for 20 weeks (n = 6/group). A. Representative M-mode echocardiographic images collected using the parasternal long-axis view in each group; B–E. Quantitative analysis of cardiac function performed using echocardiography; F–G. Cardiac fibrosis following empagliflozin treatment assessed using Masson's staining; H. Representative electron microscopy images of cardiomyocytes in each group. Diabetes led to myocyte dissolution, muscular fiber twists and Z line disappearance, the effects of which were reversed by empagliflozin. Diabetes caused mitochondrial swelling and crista fragmentation (yellow arrow), which were in contrast with the clearly visible and regular mitochondria seen in controls. Empagliflozin recovered diabetes-distorted mitochondrial structural integrity. Mean ± SD, *P < 0.05 vs. Control (Cont) group; #P < 0.05 vs. Diabetic group.
Fig. 2
Fig. 2
Empagliflozin improves myocardial microcirculation perfusion by increasing microvessel density and reducing vascular remodeling. A. Representative MCE images were taken using a constant infusion of microbubbles at 20 mL/min. The signal intensity was determined by capturing a 10-s high-energy sequence at a frame rate of 30 Hz;.B. A quantitative analysis of perfusion was performed using Research-Arena software (Tomtech, Germany). A: plateau intensity, β: flow velocity. Myocardial blood flow (A × β) profiles in the basal septum for different groups (n = 6/group); C–D. Frozen cardiac sections were incubated with CD31 antibody to assess microvascular numbers by performing fluorescence microscopy; E. Microvascular image detection via ink staining; F–G. Immunohistochemistry of p-eNOS (Ser1177) expression; H–I. Endothelial-dependent and endothelial-independent relaxation responses were assessed by applying Ach (10−9–10−5 M) or SNP (10−10–10−6 M). J. Histological analysis of vascular fibrosis with Masson's staining; K–L. TEM was performed to observe changes in the vascular basement membrane. Diabetes induced hyperplasia of the vascular basement, which was reversed by empagliflozin. Mean ± SD, *P < 0.05 vs. Control (Cont) group; #P < 0.05 vs. Diabetic group.
Fig. 3
Fig. 3
Empagliflozin maintains microvessel integrity and barrier function. A–B. The co-immunofluorescence of VE-cadherin and CD31 indicates the continuity of gap junctional proteins; C–D. Representative fluorescence images for intercellular adhesion molecule (ICAM)-1; E–F. Immunohistochemical staining of vascular cell adhesion molecule (VCAM)-1; G. HE staining to assess red blood cell adhesion to microvessel walls in different groups; H–I. A TUNEL assay was performed to assess CMEC apoptosis. Mean ± SD, *P < 0.05 vs. Control (Cont) group; #P < 0.05 vs. Diabetic group.
Fig. 4
Fig. 4
Empagliflozin diminishes diabetic-induced mitochondrial fragmentation and regulated the balance of proteins responsible for mitochondrial fission and fusion. CMECs were obtained from the control group, diabetic group and empagliflozin group. After isolation, empagliflozin-treated CMECs were treated with FCCP to trigger mitochondrial fission. A. EM was performed to observe mitochondrial fission in vivo. Diabetes induced mitochondrial disintegration into numerous round fragments of varying sizes accompanied by the disappearance of crista (white arrow), unlike the characteristic reticulo-tubular mitochondrial morphology seen in the control group, and these changes were reversed by empagliflozin; B. CD31 immunocytochemistry of CMECs and Dil-acetylated low-density lipoprotein intake assay. (green, CD31; red, Dil-LDL); C. CMECs were isolated and cultured in vitro, and MitoTracker Deep Red was used to label the mitochondria, whose morphology was analyzed using fluorescence microscopy; D. Co-localization of Drp1 and mitochondria. The boxed area under each micrograph represents the amplification of the white square. More Drp1 was located on fragmented mitochondria, while empagliflozin reduced Drp1 migration onto mitochondria; E. Changes in proteins related to mitochondrial fission and fusion. Empagliflozin reduced fission-associated factors and increased fusion-involved protein levels. Mean ± SD, *P < 0.05 vs. Control (Cont) group; #P < 0.05 vs. Diabetic group, &P < 0.05 vs. Diabetic + EMPA group.
Fig. 5
Fig. 5
Empagliflozin protects CMECs against cellular senescence and barrier dysfunction by suppressing fission-mediated mtROS overproduction. A. Representative images of β-galactosidase staining, which was the marker for aged cells. B. Quantification of β-galactosidase-positive endothelial cells. C. The immunofluorescence of intracellular ROS and mtROS as assessed by DCF-DA and MitoSOX™ Red Mitochondrial Superoxide Indicator. Mitoquinone (MitoQ, 2 μM) was applied for 24 h to reduce the cellular oxidative damage. D–E. Cell cycle distribution was detected by performing flow cytometric quantification. Loss of fission due to empagliflozin and mdivi1 contributed to the cell cycle transition from G0/G1 to S phase, suggesting the anti-senescence effects of empagliflozin occur via fission inhibition. F. A TER assay was performed to detect CMEC barrier function during mitochondrial fission. TER increases when endothelial cells adhere and spread out and decreases when endothelial cells retract or lose adhesion, reflecting endothelial barrier integrity. G. FITC-dextran clearance was measured to assess changes in endothelial permeability. FITC-dextran was applied on top of the inserts and allowed to permeate through cell monolayers. The increased endothelial permeability resulted in the retention of more FITC-dextran. Thus, FITC content remaining in the plate indicates the extent of CMEC permeability. H–J. The change of expression of p-eNOS (Ser1177), ICAM1 and VCAM1. Mean ± SD, *P < 0.05 vs. Control (Cont) group; #P < 0.05 vs. Diabetic group, &P < 0.05 vs. Diabetic + EMPA group, %P < 0.05 vs. Diabetic + EMPA + FCCP group.
Fig. 6
Fig. 6
Suppression of fission by empagliflozin contributes to CMEC migration and neovascularization by preserving F-actin homeostasis. A–B. The endothelial migration response to SDF-1 was analyzed by performing a transwell assay. C. Co-immunofluorescence of F-actin and mitochondria to establish the role of mitochondrial fission in F-actin homeostasis. Fragmented mitochondria were accompanied by F-actin fiber disorder or dissolution, which was reversed by empagliflozin or mdivi1. D–F. Changes in the expression of F-actin and its metabolite G-actin as assessed by western blotting. G–H. A wound-healing assay was carried out to detect cell motility with F-actin depolymerization. I–J. The influence of mitochondrial fission on the capacity of CMECs to undergo angiogenesis via F-actin was measured by performing a Matrigel assay in vitro. Mean ± SD, *P < 0.05 vs. Control (Cont) group; #P < 0.05 vs. Diabetic group, &P < 0.05 vs. Diabetic + EMPA group.
Fig. 7
Fig. 7
AMPK signaling is activated by empagliflozin via increases in the AMP/ATP ratio, contributing to Drp1 modifications and subsequently mitochondrial fission. A–D. Empagliflozin increased p-AMPK levels. Activated AMPK was involved in regulating Drp1 phosphorylation. E. Empagliflozin increased the AMP/ATP ratio, which was responsible for AMPK activation. Mean ± SD, *P < 0.05 vs. Control (Cont) group; #P < 0.05 vs. Diabetic group, &P < 0.05 vs. Diabetic+EMPA group. AI: AICAR, cC: compound C.
Fig. 8
Fig. 8
Scheme depicting proposed mechanisms involved in empagliflozin-offered protection against microvasculature damage in diabetes. Empagliflozin activates AMPK pathways through regulation of the AMP/ATP ratio. Activated AMPK pathways regulates Drp1 posttranscriptional phosphorylation modifications at Ser616 and Ser637, leading to the inability of Drp1 to translocate onto mitochondria and mitochondrial fission impairment. The loss of mitochondrial fission retards cellular senescence and preserves endothelial barrier/permeability by suppressing superfluous ROS. In consequence, endothelial migration and vascularization are improved by balanced F-actin degradation. Moreover, empagliflozin reduces CMEC apoptosis, increases cardiac microvessel density, promotes eNOS phosphorylation and alleviates vascular collagen deposition, leading to improved endothelial function and preserved vascular remodeling, ultimately lower levels of inflammatory cell penetration and better vascular relaxation. Through these aforementioned mechanisms, empagliflozin eventually facilitates diabetic myocardial perfusion and protects the heart against hyperglycemic injury.

References

    1. Zhou H., Hu S., Jin Q., Shi C., Zhang Y., Zhu P., Ma Q., Tian F., Chen Y. Mff-dependent mitochondrial fission contributes to the pathogenesis of cardiac microvasculature ischemia/reperfusion injury via induction of mROS-mediated cardiolipin oxidation and HK2/VDAC1 disassociation-involved mPTP opening. J. Am. Heart Assoc. 2017;6(3)
    1. Sawada N., Jiang A., Takizawa F., Safdar A., Manika A., Tesmenitsky Y., Kang K.T., Bischoff J., Kalwa H., Sartoretto J.L., Kamei Y., Benjamin L.E., Watada H., Ogawa Y., Higashikuni Y., Kessinger C.W., Jaffer F.A., Michel T., Sata M., Croce K., Tanaka R., Arany Z. Endothelial PGC-1alpha mediates vascular dysfunction in diabetes. Cell Metab. 2014;19(2):246–258.
    1. Hamilton S.J., Chew G.T., Watts G.F. Therapeutic regulation of endothelial dysfunction in type 2 diabetes mellitus. Diabetes Vasc. Dis. Res. 2007;4(2):89–102.
    1. Eelen G., De Zeeuw P., Simons M., Carmeliet P. Endothelial cell metabolism in normal and diseased vasculature. Circ. Res. 2015;116(7):1231–1244.
    1. Steven S., Oelze M., Hanf A., Kroller-Schon S., Kashani F., Roohani S., Welschof P., Kopp M., Godtel-Armbrust U., Xia N., Li H., Schulz E., Lackner K.J., Wojnowski L., Bottari S.P., Wenzel P., Mayoux E., Munzel T., Daiber A. The SGLT2 inhibitor empagliflozin improves the primary diabetic complications in ZDF rats. Redox Biol. 2017;13:370–385.
    1. Zinman B., Lachin J.M., Inzucchi S.E. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N. Engl. J. Med. 2016;374(11):1094.
    1. Zinman B., Wanner C., Lachin J.M., Fitchett D., Bluhmki E., Hantel S., Mattheus M., Devins T., Johansen O.E., Woerle H.J., Broedl U.C., Inzucchi S.E., O E.-R. Investigators, empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N. Engl. J. Med. 2015;373(22):2117–2128.
    1. Cefalu W.T. Paradoxical insights into whole body metabolic adaptations following SGLT2 inhibition. J. Clin. Invest. 2014;124(2):485–487.
    1. Dixon J.B. Obesity in 2015: advances in managing obesity. Nat. Rev. Endocrinol. 2016;12(2):65–66.
    1. Quintero M., Colombo S.L., Godfrey A., Moncada S. Mitochondria as signaling organelles in the vascular endothelium. Proc. Natl. Acad. Sci. USA. 2006;103(14):5379–5384.
    1. Fuhrmann D.C., Brune B. Mitochondrial composition and function under the control of hypoxia. Redox Biol. 2017;12:208–215.
    1. Wang W., Wang Y., Long J., Wang J., Haudek S.B., Overbeek P., Chang B.H., Schumacker P.T., Danesh F.R. Mitochondrial fission triggered by hyperglycemia is mediated by ROCK1 activation in podocytes and endothelial cells. Cell Metab. 2012;15(2):186–200.
    1. Makino A., Scott B.T., Dillmann W.H. Mitochondrial fragmentation and superoxide anion production in coronary endothelial cells from a mouse model of type 1 diabetes. Diabetologia. 2010;53(8):1783–1794.
    1. Zhou H., Zhu P., Guo J., Hu N., Wang S., Li D., Hu S., Ren J., Cao F., Chen Y. Ripk3 induces mitochondrial apoptosis via inhibition of FUNDC1 mitophagy in cardiac IR injury. Redox Biol. 2017;13:498–507.
    1. Zhou H., Du W., Li Y., Shi C., Hu N., Ma S., Wang W., Ren J. Effects of melatonin on fatty liver disease: the role of NR4A1/DNA-PKcs/p53 pathway, mitochondrial fission, and mitophagy. J. Pineal Res. 2018;64(1)
    1. Ding M., Ning J., Feng N., Li Z., Liu Z., Wang Y., Wang Y., Li X., Huo C., Jia X., Xu R., Fu F., Wang X., Pei J. Dynamin-related protein 1-mediated mitochondrial fission contributes to post-traumatic cardiac dysfunction in rats and the protective effect of melatonin. J. Pineal Res. 2018;64(1)
    1. Yang X., Wang H., Ni H.M., Xiong A., Wang Z., Sesaki H., Ding W.X., Yang L. Inhibition of Drp1 protects against senecionine-induced mitochondria-mediated apoptosis in primary hepatocytes and in mice. Redox Biol. 2017;12:264–273.
    1. Park S.J., Lee H., Jo D.S., Jo Y.K., Shin J.H., Kim H.B., Seo H.M., Rubinsztein D.C., Koh J.Y., Lee E.K., Cho D.H. Heterogeneous nuclear ribonucleoprotein A1 post-transcriptionally regulates Drp1 expression in neuroblastoma cells. Biochim. Biophys. Acta. 2015;1849(12):1423–1431.
    1. Kashatus D.F. Restraining the divider: a Drp1-phospholipid interaction inhibits Drp1 activity and shifts the balance from mitochondrial fission to fusion. Mol. Cell. 2016;63(6):913–915.
    1. Wang Q., Zhang M., Torres G., Wu S., Ouyang C., Xie Z., Zou M.H. Metformin suppresses diabetes-accelerated atherosclerosis via the inhibition of Drp1-mediated mitochondrial fission. Diabetes. 2017;66(1):193–205.
    1. Zhou H., Zhang Y., Hu S., Shi C., Zhu P., Ma Q., Jin Q., Cao F., Tian F., Chen Y. Melatonin protects cardiac microvasculature against ischemia/reperfusion injury via suppression of mitochondrial fission-VDAC1-HK2-mPTP-mitophagy axis. J. Pineal Res. 2017;63(1)
    1. Zhang Y., Zhou H., Wu W., Shi C., Hu S., Yin T., Ma Q., Han T., Zhang Y., Tian F., Chen Y. Liraglutide protects cardiac microvascular endothelial cells against hypoxia/reoxygenation injury through the suppression of the SR-Ca(2+)-XO-ROS axis via activation of the GLP-1R/PI3K/Akt/survivin pathways. Free Radic. Biol. Med. 2016;95:278–292.
    1. Lee H.J., Jung Y.H., Choi G.E., Ko S.H., Lee S.J., Lee S.H., Han H.J. BNIP3 induction by hypoxia stimulates FASN-dependent free fatty acid production enhancing therapeutic potential of umbilical cord blood-derived human mesenchymal stem cells. Redox Biol. 2017;13:426–443.
    1. Das N., Mandala A., Naaz S., Giri S., Jain M., Bandyopadhyay D., Reiter R.J., Roy S.S. Melatonin protects against lipid-induced mitochondrial dysfunction in hepatocytes and inhibits stellate cell activation during hepatic fibrosis in mice. J. Pineal Res. 2017;62(4)
    1. Zhou H., Li D., Shi C., Xin T., Yang J., Zhou Y., Hu S., Tian F., Wang J., Chen Y. Effects of Exendin-4 on bone marrow mesenchymal stem cell proliferation, migration and apoptosis in vitro. Sci. Rep. 2015;5:12898.
    1. Zhou H., Yang J., Xin T., Zhang T., Hu S., Zhou S., Chen G., Chen Y. Exendin-4 enhances the migration of adipose-derived stem cells to neonatal rat ventricular cardiomyocyte-derived conditioned medium via the phosphoinositide 3-kinase/Akt-stromal cell-derived factor-1alpha/CXC chemokine receptor 4 pathway. Mol. Med. Rep. 2015;11(6):4063–4072.
    1. Yang X., Xu Y., Wang T., Shu D., Guo P., Miskimins K., Qian S.Y. Inhibition of cancer migration and invasion by knocking down delta-5-desaturase in COX-2 overexpressed cancer cells. Redox Biol. 2017;11:653–662.
    1. Gao Y., Xiao X., Zhang C., Yu W., Guo W., Zhang Z., Li Z., Feng X., Hao J., Zhang K., Xiao B., Chen M., Huang W., Xiong S., Wu X., Deng W. Melatonin synergizes the chemotherapeutic effect of 5-fluorouracil in colon cancer by suppressing PI3K/AKT and NF-kappaB/iNOS signaling pathways. J. Pineal Res. 2017;62(2)
    1. Malinda K.M. In vivo matrigel migration and angiogenesis assay. Methods Mol. Biol. 2009;467:287–294.
    1. Sigala F., Efentakis P., Karageorgiadi D., Filis K., Zampas P., Iliodromitis E.K., Zografos G., Papapetropoulos A., Andreadou I. Reciprocal regulation of eNOS, H2S and CO-synthesizing enzymes in human atheroma: correlation with plaque stability and effects of simvastatin. Redox Biol. 2017;12:70–81.
    1. Yu S., Wang X., Geng P., Tang X., Xiang L., Lu X., Li J., Ruan Z., Chen J., Xie G., Wang Z., Ou J., Peng Y., Luo X., Zhang X., Dong Y., Pang X., Miao H., Chen H., Liang H. Melatonin regulates PARP1 to control the senescence-associated secretory phenotype (SASP) in human fetal lung fibroblast cells. J. Pineal Res. 2017;63(1)
    1. Zhou H., Li D., Zhu P., Hu S., Hu N., Ma S., Zhang Y., Han T., Ren J., Cao F., Chen Y. Melatonin suppresses platelet activation and function against cardiac ischemia/reperfusion injury via PPARgamma/FUNDC1/mitophagy pathways. J. Pineal Res. 2017;63(4)
    1. Jin Q., Li R., Hu N., Xin T., Zhu P., Hu S., Ma S., Zhu H., Ren J., Zhou H. DUSP1 alleviates cardiac ischemia/reperfusion injury by suppressing the Mff-required mitochondrial fission and Bnip3-related mitophagy via the JNK pathways. Redox Biol. 2018;14:576–587.
    1. Shi C., Cai Y., Li Y., Li Y., Hu N., Ma S., Hu S., Zhu P., Wang W., Zhou H. Yap promotes hepatocellular carcinoma metastasis and mobilization via governing cofilin/F-actin/lamellipodium axis by regulation of JNK/Bnip3/SERCA/CaMKII pathways. Redox Biol. 2018;14:59–71.
    1. Kaul S. Is the mortality benefit with empagliflozin in type 2 diabetes mellitus too good to be true? Circulation. 2016;134(2):94–96.
    1. Rosenson R.S., Fioretto P., Dodson P.M. Does microvascular disease predict macrovascular events in type 2 diabetes? Atherosclerosis. 2011;218(1):13–18.
    1. Katakam P.V., Wappler E.A., Katz P.S., Rutkai I., Institoris A., Domoki F., Gaspar T., Grovenburg S.M., Snipes J.A., Busija D.W. Depolarization of mitochondria in endothelial cells promotes cerebral artery vasodilation by activation of nitric oxide synthase. Arterioscler. Thromb. Vasc. Biol. 2013;33(4):752–759.
    1. Chilton R., Tikkanen I., Cannon C.P., Crowe S., Woerle H.J., Broedl U.C., Johansen O.E. 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(12):1180–1193.
    1. Gilbert R.E., Krum H. Heart failure in diabetes: effects of anti-hyperglycaemic drug therapy. Lancet. 2015;385(9982):2107–2117.
    1. Sarwar C.M., Papadimitriou L., Pitt B., Pina I., Zannad F., Anker S.D., Gheorghiade M., Butler J. Hyperkalemia in heart failure. J. Am. Coll. Cardiol. 2016;68(14):1575–1589.
    1. Wang D., Luo P., Wang Y., Li W., Wang C., Sun D., Zhang R., Su T., Ma X., Zeng C., Wang H., Ren J., Cao F. Glucagon-like peptide-1 protects against cardiac microvascular injury in diabetes via a cAMP/PKA/Rho-dependent mechanism. Diabetes. 2013;62(5):1697–1708.
    1. Xu S., Pi H., Zhang L., Zhang N., Li Y., Zhang H., Tang J., Li H., Feng M., Deng P., Guo P., Tian L., Xie J., He M., Lu Y., Zhong M., Zhang Y., Wang W., Reiter R.J., Yu Z., Zhou Z. Melatonin prevents abnormal mitochondrial dynamics resulting from the neurotoxicity of cadmium by blocking calcium-dependent translocation of Drp1 to the mitochondria. J. Pineal Res. 2016;60(3):291–302.
    1. Bereiter-Hahn J., Voth M., Mai S., Jendrach M. Structural implications of mitochondrial dynamics. Biotechnol. J. 2008;3(6):765–780.
    1. Hatch A.L., Ji W.K., Merrill R.A., Strack S., Higgs H.N. Actin filaments as dynamic reservoirs for Drp1 recruitment. Mol. Biol. Cell. 2016;27(20):3109–3121.
    1. Liu Y., Li H., Bubolz A.H., Zhang D.X., Gutterman D.D. Endothelial cytoskeletal elements are critical for flow-mediated dilation in human coronary arterioles. Med. Biol. Eng. Comput. 2008;46(5):469–478.
    1. Li S., Xu S., Roelofs B.A., Boyman L., Lederer W.J., Sesaki H., Karbowski M. Transient assembly of F-actin on the outer mitochondrial membrane contributes to mitochondrial fission. J. Cell Biol. 2015;208(1):109–123.
    1. Preau S., Delguste F., Yu Y., Remy-Jouet I., Richard V., Saulnier F., Boulanger E., Neviere R. Endotoxemia engages the RhoA kinase pathway to impair cardiac function by altering cytoskeleton, mitochondrial fission, and autophagy. Antioxid. Redox Signal. 2016;24(10):529–542.
    1. Prudent J., Mcbride H.M. Mitochondrial dynamics: ER actin tightens the Drp1 noose. Curr. Biol. 2016;26(5):R207–R209.
    1. Zhang C.S., Lin S.C. AMPK promotes autophagy by facilitating mitochondrial fission. Cell Metab. 2016;23(3):399–401.
    1. Toyama E.Q., Herzig S., Courchet J., Lewis T.L., Jr., Loson O.C., Hellberg K., Young N.P., Chen H., Polleux F., Chan D.C., Shaw R.J. Metabolism. AMP-activated protein kinase mediates mitochondrial fission in response to energy stress. Science. 2016;351(6270):275–281.
    1. Kang J.W., Hong J.M., Lee S.M. Melatonin enhances mitophagy and mitochondrial biogenesis in rats with carbon tetrachloride-induced liver fibrosis. J. Pineal Res. 2016;60(4):383–393.
    1. Li J., Wang Y., Wang Y., Wen X., Ma X.N., Chen W., Huang F., Kou J., Qi L.W., Liu B., Liu K. Pharmacological activation of AMPK prevents Drp1-mediated mitochondrial fission and alleviates endoplasmic reticulum stress-associated endothelial dysfunction. J. Mol. Cell. Cardiol. 2015;86:62–74.
    1. Rovira-Llopis S., Banuls C., Diaz-Morales N., Hernandez-Mijares A., Rocha M., Victor V.M. Mitochondrial dynamics in type 2 diabetes: pathophysiological implications. Redox Biol. 2017;11:637–645.
    1. Zhu H., Jin Q., Li Y., Ma Q., Wang J., Li D., Zhou H., Chen Y. Melatonin protected cardiac microvascular endothelial cells against oxidative stress injury via suppression of IP3R-[Ca(2+)]c/VDAC-[Ca(2+)]m axis by activation of MAPK/ERK signaling pathway. Cell Stress Chaperon. 2017
    1. Suarez-Rivero J.M., Villanueva-Paz M., De La Cruz-Ojeda P., De La Mata M., Cotan D., Oropesa-Avila M., De Lavera I., Alvarez-Cordoba M., Luzon-Hidalgo R., Sanchez-Alcazar J.A. Mitochondrial dynamics in mitochondrial diseases. Diseases. 2016;5(1)
    1. Onphachanh X., Lee H.J., Lim J.R., Jung Y.H., Kim J.S., Chae C.W., Lee S.J., Gabr A.A., Han H.J. Enhancement of high glucose-induced PINK1 expression by melatonin stimulates neuronal cell survival: involvement of MT2/Akt/NF-kappaB pathway. J. Pineal Res. 2017;63(2)
    1. Bertero E., Prates Roma L., Ameri P., Maack C. Cardiac effects of SGLT2 inhibitors: the sodium hypothesis. Cardiovasc. Res. 2017
    1. Shi X., Verma S., Yun J., Brand-Arzamendi K., Singh K.K., Liu X., Garg A., Quan A., Wen X.Y. Effect of empagliflozin on cardiac biomarkers in a zebrafish model of heart failure: clues to the EMPA-REG OUTCOME trial? Mol. Cell. Biochem. 2017;433(1–2):97–102.

Source: PubMed

3
Abonnieren