Treatment of diabetic mice with the SGLT2 inhibitor TA-1887 antagonizes diabetic cachexia and decreases mortality

Taichi Sugizaki, Shunshun Zhu, Ge Guo, Akiko Matsumoto, Jiabin Zhao, Motoyoshi Endo, Haruki Horiguchi, Jun Morinaga, Zhe Tian, Tsuyoshi Kadomatsu, Keishi Miyata, Hiroshi Itoh, Yuichi Oike, Taichi Sugizaki, Shunshun Zhu, Ge Guo, Akiko Matsumoto, Jiabin Zhao, Motoyoshi Endo, Haruki Horiguchi, Jun Morinaga, Zhe Tian, Tsuyoshi Kadomatsu, Keishi Miyata, Hiroshi Itoh, Yuichi Oike

Abstract

A favorable effect of an inhibitor of the sodium-glucose cotransporter 2 (SGLT2i) on mortality of diabetic patients was recently reported, although mechanisms underlying that effect remained unclear. Here, we examine SGLT2i effects on survival of diabetic mice and assess factors underlying these outcomes. To examine SGLT2i treatment effects in a model of severe diabetes, we fed genetically diabetic db/db mice a high-fat diet and then assessed outcomes including diabetic complications between SGLT2i TA-1887-treated and control mice. We also compare effects of SGLT2i TA-1887 with those of lowering blood glucose levels via insulin treatment. Untreated db/db mice showed remarkable weight loss, or cachexia, while TA-1887-treated mice did not but rather continued to gain weight at later time points and decreased mortality. TA-1887 treatment prevented pancreatic beta cell death, enhanced preservation of beta cell mass and endogenous insulin secretion, and increased insulin sensitivity. Moreover, TA-1887 treatment attenuated inflammation, oxidative stress, and cellular senescence, especially in visceral white adipose tissue, and antagonized endothelial dysfunction. Insulin treatment of db/db mice also prevented weight loss and antagonized inflammation and oxidative stress. However, insulin treatment had less potent effects on survival and prevention of cellular senescence and endothelial dysfunction than did TA-1887 treatment. SGLT2i treatment prevents diabetic cachexia and death by preserving function of beta cells and insulin target organs and attenuating complications. SGLT2i treatment may be a promising therapeutic strategy for type 2 diabetes patients with morbid obesity and severe insulin resistance.

Conflict of interest statement

The authors declare that they have no competing financial interests.

Figures

Fig. 1
Fig. 1
TA-1887-treated or insulin-treated severely diabetic mice show enhanced survival and increased body weight but does not alter food intake or energy expenditure. a left: Changes in body weight in db/db mice fed a high-fat diet (HF) or comparable mice treated with saline (HF + saline) or with TA-1887(HF + TA) or insulin (HF + Ins) (n = 20–30). right: AUC for body weight at indicated periods. b Food intake of each group at 13, 37, and 72 days (n = 6). c Proportion of animals surviving 4 months (n = 20–30). d Representative appearance of mice in indicated groups at day 80. e Energy expenditure (EE) after 9 weeks of each treatment, as determined by indirect calorimetry (n = 6). f Volume of lean body mass, total fat, visceral fat (v fat) and subcutaneous fat (s fat), as measured by computed tomography (CT) after 3 months of each treatment (n = 6). Values shown are means ± SEM. *p < 0.05, **p < 0.01 versus HF, and #p < 0.05, ##p < 0.01 versus HF + saline, and ¶¶p < 0.01 versus HF + TA and ††p < 0.01 versus HF + Ins
Fig. 2
Fig. 2
Protective effect of TA-1887 on pancreas and insulin sensitivity in db/db mice fed a high-fat (HF) diet. a, b Blood glucose and plasma insulin (n = 8) levels at 0, 1, and 2 months of indicated treatments. c Image showing insulin immunostaining in pancreatic tissue of representative mice after 4 months of indicated treatment. Squares in upper panels are magnified in corresponding lower panels. Scale bars: 500 μm (upper), 200 μm (lower). d Analysis of INS1 and INS2 mRNA expression in mouse pancreatic tissue after 4 months of treatment (n = 5–9). e Representative double immunostaining of insulin and active caspase 3 in pancreatic tissue after 4 months of treatment. Scale bars: 100 μm. f Insulin tolerance test after 10 weeks of drug treatment and corresponding AUC (n = 5–9). g levels of mRNAs encoding glycolytic enzymes (n = 5–9) in eWAT, iWAT, MG, MS and liver after 4 months of treatment. DAPI, 4',6-diamidino-2-phenylindole; INS1, insulin I; INS2, insulin II; HK2, hexokinase 2; Eno1, enolase 1; PFKFB1, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 1; Gpi1, glucose phosphate isomerase 1. Values shown are means ± SEM. *p < 0.05 and **p < 0.01 versus HF
Fig. 3
Fig. 3
Expression of inflammatory and senescence markers in TA-1887-treated db/db mice fed a high-fat diet. a mRNA levels of inflammatory mediators in eWAT, iWAT, MG and MS after 4 months of indicated treatment (n = 5-9). b Plasma IL-6 concentration after 2 months of drug treatment, as determined by ELISA (n = 8). c mRNA levels of the senescence markers p21 and p16INK4a in eWAT, iWAT, MG and MS after 4 months of indicated treatment (n = 5–9). d Senescence-associated staining for SPiDER beta-Gal in frozen sections of eWAT, iWAT and MG tissues after 4 months of treatment. Scale bars: 200 μm. IL-6, interleukin-6; IL-1b, interleukin-1b; PAI-1, plasminogen activator inhibitor-1; MCP1, monocyte chemoattractant protein-1; mmp9, matrix metalloproteinase 9. Values shown are means ± SEM. *p < 0.05 and **p < 0.01 versus HF
Fig. 4
Fig. 4
Effect of TA-1887 on urinary excretion and tissue expression of 8-OHdG, expression of antioxidative enzymes and vascular function in db/db mice fed a high-fat diet. a Urinary excretion of 8-OHdG in db/db mice fed a high-fat (HF) diet and treated for 4 months as indicated, as measured by ELISA (n = 5). b Immunohistochemistry with an 8-OHdG antibody of representative eWAT, iWAT and MG samples of mice treated 4 months as indicated. Scale bars: 200 μm (eWAT, iWAT), 100 μm (MG). c levels of mRNAs encoding the antioxidative enzymes MnSOD and catalase in eWAT, iWAT, MG and MS after 4 months of treatment (n = 5–9). d Endothelium-dependent vasorelaxation in response to acetylcholine in aorta after 4 months of treatment (n = 5–9). e Expression of transcripts associated with vascular inflammation in aorta of mice treated as indicated for 4 months (n = 5–9). f Expression of senescence marker transcripts in aorta after 4 months of indicated treatment (n = 5–9). g Expression of transcripts encoding antioxidative enzymes in aorta after 4 months of indicated treatment (n = 5–9). Values shown are means ± SEM. *p < 0.05 and **p < 0.01 versus HF
Fig. 5
Fig. 5
Effects of insulin on tissue composition, pancreatic beta cell, glucose metabolism and inflammation in db/db mice fed a high-fat (HF) diet. a Volume of lean body mass, total fat, visceral fat (v fat) and subcutaneous fat (s fat), as measured by Computed Tomography after 3 months of indicated treatment (n = 5). b Analysis of INS1 and INS2 mRNAs in samples of pancreatic tissue after 4 months of indicated treatment (n = 5–7). c Immunostaining for insulin in representative pancreatic tissue samples after 4 months of indicated treatment. Squares in upper panels are magnified in corresponding lower panels. Scale bars: 200 μm. d Double immunostaining for insulin and active caspase 3 in representative pancreatic tissues after 4 months of treatment. Scale bars: 100 μm. e, f Blood glucose and plasma insulin levels at 0, 1, and 2 months of drug treatment (n = 8). g levels of mRNAs encoding glycolytic enzymes in eWAT, iWAT, MG and MS after 4 months of indicated treatment (n = 5–7). h Plasma IL-6 concentrations as measured by ELISA after 2 months of drug treatment (n = 8). i levels of inflammatory mRNAs in eWAT, iWAT, MG and MS after 4 months of indicated treatment (n = 5–7). DAPI, 4',6-diamidino-2-phenylindole; INS1, insulin I; INS2, insulin II; HK2, hexokinase 2; Eno1, enolase 1; PFKFB1, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 1; Gpi1, glucose phosphate isomerase 1; IL-6, interleukin-6; IL-1b, interleukin-1b; PAI-1, plasminogen activator inhibitor-1; MCP1, monocyte chemoattractant protein-1; mmp9, matrix metalloproteinase 9. Values shown are means ± SEM. *p < 0.05 and **p < 0.01 versus HF + saline
Fig. 6
Fig. 6
Effects of insulin treatment on senescence and oxidative stress markers and on vascular function in db/db mice fed a high-fat (HF) diet. a left: 8-OHdG concentration in urine as determined by ELISA after 4 months of treatment. right: Immunohistochemistry with an 8-OHdG antibody of representative eWAT, iWAT and MG samples after 4 months of indicated treatment. Scale bars: 200 μm (eWAT, iWAT), 100 μm (MG). b Levels of transcripts encoding the antioxidative enzymes MnSOD or catalase in mouse eWAT, iWAT, MG and MS following 4 months of indicated treatments (n = 5–7). c mRNA levels of the senescence markers p21 and p16INK4a in eWAT, iWAT, MG and MS after 4 months of indicated treatment (n = 5–7). d Senescence-associated staining for SPiDER beta-Gal in frozen sections of eWAT, iWAT and MG tissues after 4 months of indicated treatment. Scale bars: 200 μm. e Endothelium-dependent vasorelaxation in response to acetylcholine in aorta after 4 months of treatment (n = 5–7). f Expression of transcripts associated with vascular inflammation in aorta after 4 months of treatment (n = 5–7). g Expression of transcripts of senescence markers in aorta after 4 months of indicated treatment (n = 5–7). h Expression of transcripts encoding antioxidative enzymes in aorta after 4 months of indicated treatment (n = 5–7). Values shown are means ± SEM. *p < 0.05 and **p < 0.01 versus HF + saline

References

    1. Stratton IM, et al. Association of glycaemia with macrovascular and microvascular complications of type 2 diabetes (UKPDS 35): prospective observational study. BMJ. 2000;321:405–412. doi: 10.1136/bmj.321.7258.405.
    1. Emerging Risk Factors Collaboration. et al. Diabetes mellitus, fasting blood glucose concentration, and risk of vascular disease: a collaborative meta-analysis of 102 prospective studies. Lancet. 2010;375:2215–2222. doi: 10.1016/S0140-6736(10)60484-9.
    1. UK Prospective Diabetes Study (UKPDS Group Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33) Lancet. 1998;352:837–853. doi: 10.1016/S0140-6736(98)07019-6.
    1. UK Prospective Diabetes Study (UKPDS Group Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34) Lancet. 1998;352:854–865. doi: 10.1016/S0140-6736(98)07037-8.
    1. Holman RR, et al. 10-year follow-up of intensive glucose control in type 2 diabetes. N. Engl. J. Med. 2008;359:1577–1589. doi: 10.1056/NEJMoa0806470.
    1. Group AC, et al. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N. Engl. J. Med. 2008;358:2560–2572. doi: 10.1056/NEJMoa0802987.
    1. Zinman B, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N. Engl. J. Med. 2015;373:2117–2128. doi: 10.1056/NEJMoa1504720.
    1. Cardoso CR, Ferreira MT, Leite NC, Salles GF. Prognostic impact of aortic stiffness in high-risk type 2 diabetic patients: the Rio deJaneiro type 2 diabetes cohort study. Diabetes Care. 2013;36:3772–3778. doi: 10.2337/dc13-0506.
    1. Bakris GL, Molitch M. Microalbuminuria as a risk predictor in diabetes: the continuing saga. Diabetes Care. 2014;37:867–875. doi: 10.2337/dc13-1870.
    1. Tikkanen I, et al. Empagliflozin reduces blood pressure in patients with type 2 diabetes and hypertension. Diabetes Care. 2015;38:420–428. doi: 10.2337/dc14-1096.
    1. Nomura S, et al. Novel Indole-N-glucoside, TA-1887 as a sodium glucose cotransporter 2 inhibitor for treatment of type 2 diabetes. ACS Med Chem. Lett. 2014;5:51–55. doi: 10.1021/ml400339b.
    1. Kuriyama C, et al. Analysis of the effect of canagliflozin on renal glucose reabsorption and progression of hyperglycemia in zucker diabetic Fatty rats. J. Pharmacol. Exp. Ther. 2014;351:423–431. doi: 10.1124/jpet.114.217992.
    1. Bolinder J, et al. Effects of dapagliflozin on body weight, total fat mass, and regional adipose tissue distribution in patients with type 2 diabetes mellitus with inadequate glycemic control on metformin. J. Clin. Endocrinol. Metab. 2012;97:1020–1031. doi: 10.1210/jc.2011-2260.
    1. Suzuki M, et al. Tofogliflozin, a sodium/glucose cotransporter 2 inhibitor, attenuates body weight gain and fat accumulation in diabetic and obese animal models. Nutr. Diabetes. 2014;4:e125. doi: 10.1038/nutd.2014.20.
    1. Zhang HM, et al. Geldanamycin derivative ameliorates high fat diet-induced renal failure in diabetes. PLoS One. 2012;7:e32746. doi: 10.1371/journal.pone.0032746.
    1. Okada-Iwabu M, et al. A small-molecule AdipoR agonist for type 2 diabetes and short life in obesity. Nature. 2013;503:493–499. doi: 10.1038/nature12656.
    1. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444:860–867. doi: 10.1038/nature05485.
    1. Shoelson SE, Lee J, Goldfine AB. Inflammation and insulin resistance. J. Clin. Invest. 2006;116:1793–1801. doi: 10.1172/JCI29069.
    1. Tominaga K. The emerging role of senescent cells in tissue homeostasis and pathophysiology. Pathobiol. Aging Age Relat. Dis. 2015;5:27743. doi: 10.3402/pba.v5.27743.
    1. Childs BG, Durik M, Baker DJ, van Deursen JM. Cellular senescence in aging and age-related disease: from mechanisms to therapy. Nat. Med. 2015;21:1424–1435. doi: 10.1038/nm.4000.
    1. Doura T, et al. Detection of LacZ-positive cells in living tissue with single-cell resolution. Angew. Chem. Int. Ed. Engl. 2016;55:9620–9624. doi: 10.1002/anie.201603328.
    1. Ceriello A, Motz E. Is oxidative stress the pathogenic mechanism underlying insulin resistance, diabetes, and cardiovascular disease? The common soil hypothesis revisited. Arterioscler. Thromb. Vasc. Biol. 2004;24:816–823. doi: 10.1161/01.ATV.0000122852.22604.78.
    1. Salmon, A. B. Beyond diabetes: does obesity-induced oxidative stress drive the aging process? Antioxidants (Basel) 5, 24 (2016).
    1. Valavanidis A, Vlachogianni T, Fiotakis C. 8-hydroxy-2’ -deoxyguanosine (8-OHdG): A critical biomarker of oxidative stress and carcinogenesis. J. Environ. Sci. Health C Environ. Carcinog. Ecotoxicol. Rev. 2009;27:120–139. doi: 10.1080/10590500902885684.
    1. Hodgson JM, Marshall JJ. Direct vasoconstriction and endothelium-dependent vasodilation. Mechanisms of acetylcholine effects on coronary flow and arterial diameter in patients with nonstenotic coronary arteries. Circulation. 1989;79:1043–1051. doi: 10.1161/01.CIR.79.5.1043.
    1. Matsubara J, et al. A dipeptidyl peptidase-4 inhibitor, des-fluoro-sitagliptin, improves endothelial function and reduces atherosclerotic lesion formation in apolipoprotein E-deficient mice. J. Am. Coll. Cardiol. 2012;59:265–276. doi: 10.1016/j.jacc.2011.07.053.
    1. Stienstra R, et al. Inflammasome is a central player in the induction of obesity and insulin resistance. Proc. Natl. Acad. Sci. USA. 2011;108:15324–15329. doi: 10.1073/pnas.1100255108.
    1. Kanda H, et al. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J. Clin. Invest. 2006;116:1494–1505. doi: 10.1172/JCI26498.
    1. Raz I, Eldor R, Cernea S, Shafrir E. Diabetes: insulin resistance and derangements in lipid metabolism. Cure through intervention in fat transport and storage. Diabetes Metab. Res. Rev. 2005;21:3–14. doi: 10.1002/dmrr.493.
    1. Wajchenberg BL. beta-cell failure in diabetes and preservation by clinical treatment. Endocr. Rev. 2007;28:187–218. doi: 10.1210/10.1210/er.2006-0038.
    1. Guillausseau PJ, et al. Abnormalities in insulin secretion in type 2 diabetes mellitus. Diabetes Metab. 2008;34:S43–48. doi: 10.1016/S1262-3636(08)73394-9.
    1. Kimura KD, Tissenbaum HA, Liu Y, Ruvkun G. daf-2, an insulin receptor-like gene that regulates longevity and diapause in Caenorhabditis elegans. Science. 1997;277:942–946. doi: 10.1126/science.277.5328.942.
    1. Clancy DJ, et al. Extension of life-span by loss of CHICO, a Drosophila insulin receptor substrate protein. Science. 2001;292:104–106. doi: 10.1126/science.1057991.
    1. Blazer S, et al. High glucose-induced replicative senescence: point of no return and effect of telomerase. Biochem. Biophys. Res. Commun. 2002;296:93–101. doi: 10.1016/S0006-291X(02)00818-5.
    1. Ksiazek K, Passos JF, Olijslagers S, von Zglinicki T. Mitochondrial dysfunction is a possible cause of accelerated senescence of mesothelial cells exposed to high glucose. Biochem. Biophys. Res. Commun. 2008;366:793–799. doi: 10.1016/j.bbrc.2007.12.021.
    1. Liu J, et al. Receptor for advanced glycation end-products promotes premature senescence of proximal tubular epithelial cells via activation of endoplasmic reticulum stress-dependent p21 signaling. Cell. Signal. 2014;26:110–121. doi: 10.1016/j.cellsig.2013.10.002.
    1. Niu S, et al. Broad infiltration of macrophages leads to a proinflammatory state in streptozotocin-induced hyperglycemic mice. J. Immunol. 2016;197:3293–3301. doi: 10.4049/jimmunol.1502494.
    1. Nelson G, et al. A senescent cell bystander effect: senescence-induced senescence. Aging Cell. 2012;11:345–349. doi: 10.1111/j.1474-9726.2012.00795.x.
    1. Boucher J, Tseng YH, Kahn CR. Insulin and insulin-like growth factor-1 receptors act as ligand-specific amplitude modulators of a common pathway regulating gene transcription. J. Biol. Chem. 2010;285:17235–17245. doi: 10.1074/jbc.M110.118620.
    1. Katic M, Kahn CR. The role of insulin and IGF-1 signaling in longevity. Cell. Mol. Life Sci. 2005;62:320–343. doi: 10.1007/s00018-004-4297-y.
    1. Handayaningsih AE, et al. IGF-I enhances cellular senescence via the reactive oxygen species-p53 pathway. Biochem. Biophys. Res. Commun. 2012;425:478–484. doi: 10.1016/j.bbrc.2012.07.140.
    1. Clark MA, Perks CM, Winters ZE, Holly JM. DNA damage uncouples the mitogenic response to IGF-I in MCF-7 malignant breast cancer cells by switching the roles of PI3 kinase and p21WAF1/Cip1. Int. J. Cancer. 2005;116:506–513. doi: 10.1002/ijc.21029.
    1. Tran D, et al. Insulin-like growth factor-1 regulates the SIRT1-p53 pathway in cellular senescence. Aging Cell. 2014;13:669–678. doi: 10.1111/acel.12219.
    1. Nguyen TT, Sheppard AM, Kaye PL, Noakes PG. IGF-I and insulin activate mitogen-activated protein kinase via the type 1 IGF receptor in mouse embryonic stem cells. Reproduction. 2007;134:41–49. doi: 10.1530/REP-06-0087.
    1. Cagnol S, Chambard JC. ERK and cell death: mechanisms of ERK-induced cell death--apoptosis, autophagy and senescence. FEBS J. 2010;277:2–21. doi: 10.1111/j.1742-4658.2009.07366.x.
    1. Hwangbo DS, et al. Drosophila dFOXO controls lifespan and regulates insulin signalling in brain and fat body. Nature. 2004;429:562–566. doi: 10.1038/nature02549.
    1. Bluher M, Kahn BB, Kahn CR. Extended longevity in mice lacking the insulin receptor in adipose tissue. Science. 2003;299:572–574. doi: 10.1126/science.1078223.
    1. Giannakou ME, et al. Long-lived Drosophila with overexpressed dFOXO in adult fat body. Science. 2004;305:361. doi: 10.1126/science.1098219.
    1. Ponugoti B, Dong G, Graves DT. Role of forkhead transcription factors in diabetes-induced oxidative stress. Exp. Diabetes Res. 2012;2012:939751. doi: 10.1155/2012/939751.
    1. Bakker W, Eringa EC, Sipkema P, van Hinsbergh VW. Endothelial dysfunction and diabetes: roles of hyperglycemia, impaired insulin signaling and obesity. Cell. Tissue Res. 2009;335:165–189. doi: 10.1007/s00441-008-0685-6.
    1. Sato Y, et al. Increased expression of CCAAT/enhancer binding protein-beta and -delta and monocyte chemoattractant protein-1 genes in aortas from hyperinsulinaemic rats. Diabetologia. 2007;50:481–489. doi: 10.1007/s00125-006-0480-4.
    1. Park CW, et al. High glucose-induced intercellular adhesion molecule-1 (ICAM-1) expression through an osmotic effect in rat mesangial cells is PKC-NF-kappa B-dependent. Diabetologia. 2000;43:1544–1553. doi: 10.1007/s001250051567.
    1. Baumgartner-Parzer SM, et al. Modulation by high glucose of adhesion molecule expression in cultured endothelial cells. Diabetologia. 1995;38:1367–1370. doi: 10.1007/BF00401771.
    1. Li G, et al. Insulin and insulin-like growth factor-I receptors differentially mediate insulin-stimulated adhesion molecule production by endothelial cells. Endocrinology. 2009;150:3475–3482. doi: 10.1210/en.2009-0172.
    1. Kuo T, Harris CA, Wang JC. Metabolic functions of glucocorticoid receptor in skeletal muscle. Mol. Cell. Endocrinol. 2013;380:79–88. doi: 10.1016/j.mce.2013.03.003.
    1. Sandri M. Autophagy in skeletal muscle. FEBS Lett. 2010;584:1411–1416. doi: 10.1016/j.febslet.2010.01.056.
    1. Sano, M., Meguro S., Kawai T., Suzuki Y. Increased grip strength with sodium-glucose cotransporter 2 inhibitors. J. Diabetes8, 736–737 (2016).
    1. Kouyama R, et al. Attenuation of diet-induced weight gain and adiposity through increased energy expenditure in mice lacking angiotensin II type 1a receptor. Endocrinology. 2005;146:3481–3489. doi: 10.1210/en.2005-0003.

Source: PubMed

3
Tilaa