SGLT2 inhibition modulates NLRP3 inflammasome activity via ketones and insulin in diabetes with cardiovascular disease

So Ra Kim, Sang-Guk Lee, Soo Hyun Kim, Jin Hee Kim, Eunhye Choi, Wonhee Cho, John Hoon Rim, Inhwa Hwang, Chan Joo Lee, Minyoung Lee, Chang-Myung Oh, Justin Y Jeon, Heon Yung Gee, Jeong-Ho Kim, Byung-Wan Lee, Eun Seok Kang, Bong-Soo Cha, Myung-Shik Lee, Je-Wook Yu, Jin Won Cho, Jung-Sun Kim, Yong-Ho Lee, So Ra Kim, Sang-Guk Lee, Soo Hyun Kim, Jin Hee Kim, Eunhye Choi, Wonhee Cho, John Hoon Rim, Inhwa Hwang, Chan Joo Lee, Minyoung Lee, Chang-Myung Oh, Justin Y Jeon, Heon Yung Gee, Jeong-Ho Kim, Byung-Wan Lee, Eun Seok Kang, Bong-Soo Cha, Myung-Shik Lee, Je-Wook Yu, Jin Won Cho, Jung-Sun Kim, Yong-Ho Lee

Abstract

Sodium-glucose cotransporter 2 (SGLT2) inhibitors reduce cardiovascular events in humans with type 2 diabetes (T2D); however, the underlying mechanism remains unclear. Activation of the NLR family, pyrin domain-containing 3 (NLRP3) inflammasome and subsequent interleukin (IL)-1β release induces atherosclerosis and heart failure. Here we show the effect of SGLT2 inhibitor empagliflozin on NLRP3 inflammasome activity. Patients with T2D and high cardiovascular risk receive SGLT2 inhibitor or sulfonylurea for 30 days, with NLRP3 inflammasome activation analyzed in macrophages. While the SGLT2 inhibitor's glucose-lowering capacity is similar to sulfonylurea, it shows a greater reduction in IL-1β secretion compared to sulfonylurea accompanied by increased serum β-hydroxybutyrate (BHB) and decreased serum insulin. Ex vivo experiments with macrophages verify the inhibitory effects of high BHB and low insulin levels on NLRP3 inflammasome activation. In conclusion, SGLT2 inhibitor attenuates NLRP3 inflammasome activation, which might help to explain its cardioprotective effects.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1. Participant flow diagram.
Fig. 1. Participant flow diagram.
Numbers of participants who were initially screened, underwent random allocation, withdrew, and were included in the final analysis. SGLT2 sodium–glucose cotransporter 2.
Fig. 2. Effects of SGLT2 inhibitor and…
Fig. 2. Effects of SGLT2 inhibitor and sulfonylurea on metabolic parameters.
ai Changes in metabolic parameters from baseline to end of treatment (sulfonylurea group: n = 32, SGLT2 inhibitor group: n = 29). †Statistical significance for the time × group interaction evaluated by repeat-measures analysis of variance (ANOVA) (Non-normally distributed variables were log transformed for analysis and back transformed for presentation). Data are represented as mean ± SEM or median (interquartile range). Two-sided paired t test or Wilcoxon signed rank test; *P < 0.05, **P < 0.01, and ***P < 0.001 versus baseline. BHB β-hydroxybutyrate, FFA free fatty acid, HOMA-IR homeostatic model assessment of insulin resistance, QUICKI quantitative insulin sensitivity check index, SEM standard error of the mean, SGLT2 sodium–glucose cotransporter 2. Source data are provided as a Source Data file.
Fig. 3. Effects of SGLT2 inhibitor and…
Fig. 3. Effects of SGLT2 inhibitor and sulfonylurea on secretion of IL-1β and TNF-α from macrophages.
ELISA assay measurement of IL-1β secretion from macrophages exposed to 2 mM ATP (a) or 0.2 mM palmitate (b) with 0.1 μg/mL LPS priming (sulfonylurea group: n = 32, SGLT2 inhibitor group: n = 29). TNF-α secretion from macrophages exposed to ATP (c) or palmitate (d). Experiments were repeated twice or three times per sample; bar graphs are drawn using mean values of those results per sample, whereas the statistical significances are derived from raw data. Data are represented as mean ± SEM. Two-sided paired t test; *P < 0.05, **P < 0.01, and ***P < 0.001 versus baseline. mRNA levels encoding IL1B (e), TNFA (f), and NLRP3 (g) (n = 6 per group). Two-sided paired t test or Wilcoxon signed rank test. †Statistical significance for the time × group interaction evaluated by repeat-measures analysis of variance (ANOVA) (Non-normally distributed variables were log transformed for analysis and back transformed for presentation). h Representative protein levels of molecules regarding NLRP3 inflammasome activation with or without LPS and ATP stimulation. IL-1β interleukin-1β, NF-κB nuclear factor kappa-light-chain-enhancer of activated B cells, NLRP3 NLR family, pyrin domain-containing 3, PA palmitate, SEM standard error of the mean, SGLT2 sodium–glucose cotransporter 2, TNF-α tumor necrosis factor-α. Source data are provided as a Source Data file.
Fig. 4. Effects of BHB, glucose, and…
Fig. 4. Effects of BHB, glucose, and insulin on NLRP3 inflammasome activation in human macrophages by ELISA assay.
IL-1β secretion when exposed to vehicle or 2 mM ATP with 0.1 μg/mL LPS priming and increased BHB (1, 10, 20, and 30 mM) or 100 nM MCC950, a small-molecule inhibitor of NLRP3 inflammasome (administered for the last 5 hours as a positive control) (a); increased 2-DG (b); increased glucose (11.5 mM vs. 25 mM) (c); 11.5 mM glucose with/without 10 nM insulin (d); increased BHB (10 mM vs. 20 mM) with/without 10 nM insulin (e). 5–64 biologically independent samples per treatment; experiments on each treatment were repeated up to three times per sample. Symbols are data points from independent experiments: n = 137, 90, 58, 64, 28, 24, 5 (a, left to right), 66, 44, 12, 12 (b, left to right), 39, 27, 28, 11 (c, left to right), 48, 33, 21 (d, left to right), 39, 26, 18, 14, 14, 8 (e, left to right). Data are represented as mean ± SEM. One-way analysis of variance (ANOVA) using Tukey’s test or a two-tailed Student’s t test with the Bonferroni method for adjusting P values; *P < 0.05, **P < 0.01, and ***P < 0.001. BHB β-hydroxybutyrate, IL-1β interleukin-1β, NLRP3 NLR family, pyrin domain-containing 3, SEM standard error of the mean, 2-DG 2-deoxyglucose. Source data are provided as a Source Data file.
Fig. 5. Scheme representing the proposed effects…
Fig. 5. Scheme representing the proposed effects of SGLT2 inhibitor on NLRP3 inflammasome activation.
BHB β-hydroxybutyrate, FFA free fatty acid, IL-1β interleukin-1β, NLRP3 NLR family, pyrin domain-containing 3, SGLT2 sodium–glucose cotransporter 2.

References

    1. Eckel RH, Kahn R, Robertson RM, Rizza RA. Preventing cardiovascular disease and diabetes: a call to action from the American Diabetes Association and the American Heart Association. Circulation. 2006;113:2943–2946. doi: 10.1161/CIRCULATIONAHA.106.176583.
    1. Guo H, Callaway JB, Ting JP. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat. Med. 2015;21:677–687. doi: 10.1038/nm.3893.
    1. Siwik DA, Chang DL, Colucci WS. Interleukin-1beta and tumor necrosis factor-alpha decrease collagen synthesis and increase matrix metalloproteinase activity in cardiac fibroblasts in vitro. Circ. Res. 2000;86:1259–1265. doi: 10.1161/01.RES.86.12.1259.
    1. Duewell P, et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature. 2010;464:1357–1361. doi: 10.1038/nature08938.
    1. Van Tassell BW, Toldo S, Mezzaroma E, Abbate A. Targeting interleukin-1 in heart disease. Circulation. 2013;128:1910–1923. doi: 10.1161/CIRCULATIONAHA.113.003199.
    1. Kim SH, et al. Ezetimibe ameliorates steatohepatitis via AMP activated protein kinase-TFEB-mediated activation of autophagy and NLRP3 inflammasome inhibition. Autophagy. 2017;13:1767–1781. doi: 10.1080/15548627.2017.1356977.
    1. Grebe A, Hoss F, Latz E. NLRP3 inflammasome and the IL-1 pathway in atherosclerosis. Circ. Res. 2018;122:1722–1740. doi: 10.1161/CIRCRESAHA.118.311362.
    1. Ferrannini E. Sodium-glucose co-transporters and their inhibition: clinical physiology. Cell Metab. 2017;26:27–38. doi: 10.1016/j.cmet.2017.04.011.
    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. Neal B, et al. Canagliflozin and cardiovascular and renal events in type 2 diabetes. N. Engl. J. Med. 2017;377:644–657. doi: 10.1056/NEJMoa1611925.
    1. Ferrannini E, Mark M, Mayoux E. CV protection in the EMPA-REG OUTCOME Trial: a “thrifty substrate” hypothesis. Diabetes Care. 2016;39:1108–1114. doi: 10.2337/dc16-0330.
    1. Youm YH, et al. The ketone metabolite beta-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat. Med. 2015;21:263–269. doi: 10.1038/nm.3804.
    1. Traba J, et al. Fasting and refeeding differentially regulate NLRP3 inflammasome activation in human subjects. J. Clin. Investig. 2015;125:4592–4600. doi: 10.1172/JCI83260.
    1. McKellar GE, McCarey DW, Sattar N, McInnes IB. Role for TNF in atherosclerosis? Lessons from autoimmune disease. Nat. Rev. Cardiol. 2009;6:410–417. doi: 10.1038/nrcardio.2009.57.
    1. Maki-Petaja KM, et al. Rheumatoid arthritis is associated with increased aortic pulse-wave velocity, which is reduced by anti-tumor necrosis factor-alpha therapy. Circulation. 2006;114:1185–1192. doi: 10.1161/CIRCULATIONAHA.105.601641.
    1. Mooradian AD, Reed RL, Meredith KE, Scuderi P. Serum levels of tumor necrosis factor and IL-1 alpha and IL-1 beta in diabetic patients. Diabetes Care. 1991;14:63–65. doi: 10.2337/diacare.14.1.63.
    1. Doganay S, et al. Comparison of serum NO, TNF-alpha, IL-1beta, sIL-2R, IL-6 and IL-8 levels with grades of retinopathy in patients with diabetes mellitus. Eye. 2002;16:163–170. doi: 10.1038/sj/eye/6700095.
    1. Gustavsson C, Agardh E, Bengtsson B, Agardh CD. TNF-alpha is an independent serum marker for proliferative retinopathy in type 1 diabetic patients. J. Diabetes Complicat. 2008;22:309–316. doi: 10.1016/j.jdiacomp.2007.03.001.
    1. Tan HY, et al. Aberrant inflammasome activation characterizes tuberculosis-associated immune reconstitution inflammatory syndrome. J. Immunol. 2016;196:4052–4063. doi: 10.4049/jimmunol.1502203.
    1. Ikonomidis I, et al. Increased proinflammatory cytokines in patients with chronic stable angina and their reduction by aspirin. Circulation. 1999;100:793–798. doi: 10.1161/01.CIR.100.8.793.
    1. Toldo S, et al. Independent roles of the priming and the triggering of the NLRP3 inflammasome in the heart. Cardiovasc. Res. 2015;105:203–212. doi: 10.1093/cvr/cvu259.
    1. Devlin CM, Kuriakose G, Hirsch E, Tabas I. Genetic alterations of IL-1 receptor antagonist in mice affect plasma cholesterol level and foam cell lesion size. Proc. Natl Acad. Sci. USA. 2002;99:6280–6285. doi: 10.1073/pnas.092324399.
    1. Chamberlain J, et al. Interleukin-1 regulates multiple atherogenic mechanisms in response to fat feeding. PLoS ONE. 2009;4:e5073. doi: 10.1371/journal.pone.0005073.
    1. Kang HT, Hwang ES. 2-Deoxyglucose: an anticancer and antiviral therapeutic, but not any more a low glucose mimetic. Life Sci. 2006;78:1392–1399. doi: 10.1016/j.lfs.2005.07.001.
    1. Nicklin MJ, Hughes DE, Barton JL, Ure JM, Duff GW. Arterial inflammation in mice lacking the interleukin 1 receptor antagonist gene. J. Exp. Med. 2000;191:303–312. doi: 10.1084/jem.191.2.303.
    1. Paramel Varghese G, et al. NLRP3 inflammasome expression and activation in human atherosclerosis. J. Am. Heart Assoc. 2016;5:e003031. doi: 10.1161/JAHA.115.003031.
    1. Sandanger O, et al. The NLRP3 inflammasome is up-regulated in cardiac fibroblasts and mediates myocardial ischaemia-reperfusion injury. Cardiovasc. Res. 2013;99:164–174. doi: 10.1093/cvr/cvt091.
    1. Ridker PM, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N. Engl. J. Med. 2017;377:1119–1131. doi: 10.1056/NEJMoa1707914.
    1. Monami M, Dicembrini I, Mannucci E. Effects of SGLT-2 inhibitors on mortality and cardiovascular events: a comprehensive meta-analysis of randomized controlled trials. Acta Diabetol. 2017;54:19–36. doi: 10.1007/s00592-016-0892-7.
    1. Kosiborod M, et al. Cardiovascular events associated with SGLT-2 inhibitors versus other glucose-lowering drugs: the CVD-REAL 2 study. J. Am. Coll. Cardiol. 2018;71:2628–2639. doi: 10.1016/j.jacc.2018.03.009.
    1. Dror E, et al. Postprandial macrophage-derived IL-1beta stimulates insulin, and both synergistically promote glucose disposal and inflammation. Nat. Immunol. 2017;18:283–292. doi: 10.1038/ni.3659.
    1. DeFronzo RA, Norton L, Abdul-Ghani M. Renal, metabolic and cardiovascular considerations of SGLT2 inhibition. Nat. Rev. Nephrol. 2017;13:11–26. doi: 10.1038/nrneph.2016.170.
    1. Goff, D.C. et al. 2013 ACC/AHA guideline on the assessment of cardiovascular risk. Circulation. 10.1161/01.cir.0000437741.48606.98 (2013).
    1. Mancia G, et al. 2007 Guidelines for the management of arterial hypertension: The Task Force for the Management of Arterial Hypertension of the European Society of Hypertension (ESH) and of the European Society of Cardiology (ESC) Eur. Heart J. 2007;28:1462–1536.
    1. Matthews DR, et al. Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia. 1985;28:412–419. doi: 10.1007/BF00280883.
    1. Katz A, et al. Quantitative insulin sensitivity check index: a simple, accurate method for assessing insulin sensitivity in humans. J. Clin. Endocrinol. Metab. 2000;85:2402–2410. doi: 10.1210/jcem.85.7.6661.
    1. Levey AS, et al. A new equation to estimate glomerular filtration rate. Ann. Intern. Med. 2009;150:604–612. doi: 10.7326/0003-4819-150-9-200905050-00006.

Source: PubMed

3
購読する