Effect of Dapagliflozin on Urine Metabolome in Patients with Type 2 Diabetes

Evdoxia Bletsa, Sebastien Filippas-Dekouan, Christina Kostara, Panagiotis Dafopoulos, Aikaterini Dimou, Eleni Pappa, Styliani Chasapi, Georgios Spyroulias, Anastasios Koutsovasilis, Eleni Bairaktari, Ele Ferrannini, Vasilis Tsimihodimos, Evdoxia Bletsa, Sebastien Filippas-Dekouan, Christina Kostara, Panagiotis Dafopoulos, Aikaterini Dimou, Eleni Pappa, Styliani Chasapi, Georgios Spyroulias, Anastasios Koutsovasilis, Eleni Bairaktari, Ele Ferrannini, Vasilis Tsimihodimos

Abstract

Context: Inhibitors of sodium-glucose cotransporters-2 have cardio- and renoprotective properties. However, the underlying mechanisms remain indeterminate.

Objective: To evaluate the effect of dapagliflozin on renal metabolism assessed by urine metabolome analysis in patients with type 2 diabetes.

Design: Prospective cohort study.

Setting: Outpatient diabetes clinic of a tertiary academic center.

Patients: Eighty patients with hemoglobin A1c > 7% on metformin monotherapy were prospectively enrolled.

Intervention: Fifty patients were treated with dapagliflozin for 3 months. To exclude that the changes observed in urine metabolome were merely the result of the improvement in glycemia, 30 patients treated with insulin degludec were used for comparison.

Main outcome measure: Changes in urine metabolic profile before and after the administration of dapagliflozin and insulin degludec were assessed by proton-nuclear magnetic resonance spectroscopy.

Results: In multivariate analysis urine metabolome was significantly altered by dapagliflozin (R2X = 0.819, R2Y = 0.627, Q2Y = 0.362, and coefficient of variation analysis of variance, P < 0.001) but not insulin. After dapagliflozin, the urine concentrations of ketone bodies, lactate, branched chain amino acids (P < 0.001), betaine, myo-inositol (P < 0001), and N-methylhydantoin (P < 0.005) were significantly increased. Additionally, the urine levels of alanine, creatine, sarcosine, and citrate were also increased (P < 0001, P <0.0001, and P <0.0005, respectively) whereas anserine decreased (P < 0005).

Conclusions: Dapagliflozin significantly affects urine metabolome in patients with type 2 diabetes in a glucose lowering-independent way. Most of the observed changes can be considered beneficial and may contribute to the renoprotective properties of dapagliflozin.

Trial registration: ClinicalTrials.gov NCT02798757.

Keywords: branched chain amino acids; dapagliflozin; kidney; metabolomics; osmolytes.

© The Author(s) 2021. Published by Oxford University Press on behalf of the Endocrine Society.

Figures

Figure 1.
Figure 1.
(A) OPLS-DA and validation (B)of the corresponding OPLS-DA models via permutations tests (n = 100) of the urine metabolic data from patients with type 2 diabetes mellitus before (black squares) and 3 months after (red triangles) administration of dapagliflozin. The regression line of Q2 (goodness of prediction, blue squares) with intercept at −0.306 and R2 (goodness of fit, green circles) regression line intercept at 0.242 indicate that the resulting OPLS-DA model has higher R2 (goodness of fit) and Q2 (goodness of prediction) values in the validation test than the permutated models generated. (C) The corresponding OPLS-DA regression coefficient plot of the urine metabolites, upper (black): higher before and down (red): higher after treatment. Abbreviations: 1MH, 1-methylhistidine; 1MNA, 1-methylnicotinamide; 2HIBA, 2-hydroxyisobutyrate; 3ClTyr, 3-chlorotyrosine; 3HIVA, 3-hydroxyisovalerate; 3IS, 3-indoxylsulfate; 3M2OV, 3-methyl-2-oxovalerate; 3MH, 3-methylhistidine; Ala, alanine; Ans, anserine; Bet, betaine; Car, carnitine; Cho, choline; cisAco, cis-aconitate; Cit, citrate; Cr, creatine; Creat, creatinine; Fum, fumarate; GLA, gluconate; Gluc, glucose; Hip, hippurate; Hpx, hypoxanthine; ILA, indole-3-lactate; Ile, isoleucine; Imid, imidazole; Lys, lysine; Mtl, mannitol; Meth, methanol; N-IVG, N-isovaleroylglycine; N-PAG, N-phenylacetylglycine; Suc, succinate; TG, trigonelline; Thr, threonine; TMAO, trimethylamine N-oxide; transAco, trans-aconitate; Val, valine; *, drug metabolites; Un, unknown.

References

    1. Zinman B, Wanner C, Lachin JM, et al. ; EMPA-REG OUTCOME Investigators . Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373(22):2117-2128.
    1. Neal B, Perkovic V, Mahaffey KW, et al. ; CANVAS Program Collaborative Group . Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J Med. 2017;377(7):644-657.
    1. Wiviott SD, Raz I, Bonaca MP, et al. ; DECLARE–TIMI 58 Investigators . Dapagliflozin and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2019;380(4):347-357.
    1. Heerspink HJL, Stefánsson BV, Correa-Rotter R, et al. ; DAPA-CKD Trial Committees and Investigators . Dapagliflozin in patients with chronic kidney disease. N Engl J Med. 2020;383(15):1436-1446.
    1. Cosentino F, Grant PJ, Aboyans V, et al. ; ESC Scientific Document Group . 2019 ESC guidelines on diabetes, pre-diabetes, and cardiovascular diseases developed in collaboration with the EASD. Eur Heart J. 2020;41(2):255-323.
    1. Ferrannini E, Mark M, Mayoux E. CV protection in the EMPA-REG OUTCOME trial: a “thrifty substrate” hypothesis. Diabetes Care. 2016;39(7):1108-1114.
    1. Perrone-Filardi P, Avogaro A, Bonora E, et al. . Mechanisms linking empagliflozin to cardiovascular and renal protection. Int J Cardiol. 2017;241:450-456.
    1. Newgard CB. Metabolomics and metabolic diseases: where do we stand? Cell Metab. 2017;25(1):43-56.
    1. Gall WE, Beebe K, Lawton KA, et al. ; RISC Study Group . alpha-hydroxybutyrate is an early biomarker of insulin resistance and glucose intolerance in a nondiabetic population. PLoS One. 2010;5(5):e10883.
    1. Wang TJ, Ngo D, Psychogios N, et al. . 2-Aminoadipic acid is a biomarker for diabetes risk. J Clin Invest. 2013;123(10):4309-4317.
    1. Liggi S, Griffin JL. Metabolomics applied to diabetes-lessons from human population studies. Int J Biochem Cell Biol. 2017;93:136-147.
    1. Kappel BA, Lehrke M, Schütt K, et al. . Effect of empagliflozin on the metabolic signature of patients with type 2 diabetes mellitus and cardiovascular disease. Circulation. 2017;136(10):969-972.
    1. Mulder S, Heerspink HJL, Darshi M, et al. . Effects of dapagliflozin on urinary metabolites in people with type 2 diabetes. Diabetes Obes Metab. 2019;21(11):2422-2428.
    1. Savorani F, Tomasi G, Engelsen SB. icoshift: A versatile tool for the rapid alignment of 1D NMR spectra. J Magn Reson. 2010;202(2):190-202.
    1. Eriksson L, Trygg J, Wold S. CV-ANOVA for significance testing of PLS and OPLS (R) models. J Chemom. 2008(22):594-600.
    1. Bletsa Ε, Filippas-Dekouan S, Kostara C, et al. . Effect of dapagliflozin on urine metabolome in patients with type 2 diabetes. Dryad data set, ProMED-mail website. 10.5061/dryad.3n5tb2rg9. Accessed March 9, 2021.
    1. Ferrannini E, Baldi S, Frascerra S, et al. . Renal handling of ketones in response to sodium-glucose cotransporter 2 inhibition in patients with type 2 diabetes. Diabetes Care. 2017;40(6):771-776.
    1. White PJ, Newgard CB. Branched-chain amino acids in disease. Science. 2019;363(6427):582-583.
    1. Bloomgarden Z. Diabetes and branched-chain amino acids: what is the link? J Diabetes. 2018;10(5):350-352.
    1. Park SH, Choi HJ, Lee JH, Woo CH, Kim JH, Han HJ. High glucose inhibits renal proximal tubule cell proliferation and involves PKC, oxidative stress, and TGF-beta 1. Kidney Int. 2001;59(5):1695-1705.
    1. Peñaranda C, García-Ocaña A, Sarasa JL, Esbrit P. Hypertrophy of rabbit proximal tubule cells is associated with overexpression of TGF beta. Life Sci. 1996;59(21):1773-1782.
    1. Johnson DW, Saunders HJ, Poronnik P, Cook DI, Field MJ, Pollock CA. Physiological changes in extracellular sodium directly control human proximal tubule growth and transport. Pflugers Arch. 1998;435(2):211-218.
    1. Onishi A, Fu Y, Darshi M, et al. . Effect of renal tubule-specific knockdown of the Na+/H+ exchanger NHE3 in Akita diabetic mice. Am J Physiol Renal Physiol. 2019;317(2):F419-F434.
    1. Vallon V, Gerasimova M, Rose MA, et al. . SGLT2 inhibitor empagliflozin reduces renal growth and albuminuria in proportion to hyperglycemia and prevents glomerular hyperfiltration in diabetic Akita mice. Am J Physiol Renal Physiol. 2014;306(2):F194-F204.
    1. Garcia-Perez A, Burg MB. Renal medullary organic osmolytes. Physiol Rev. 1991;71(4):1081-1115.
    1. Ziyadeh FN, Simmons DA, Snipes ER, Goldfarb S. Effect of myo-inositol on cell proliferation and collagen transcription and secretion in proximal tubule cells cultured in elevated glucose. J Am Soc Nephrol. 1991;1(11):1220-1229.
    1. Chang HH, Chao HN, Walker CS, Choong SY, Phillips A, Loomes KM. Renal depletion of myo-inositol is associated with its increased degradation in animal models of metabolic disease. Am J Physiol Renal Physiol. 2015;309(9):F755-F763.
    1. Nayak B, Kondeti VK, Xie P, et al. . Transcriptional and post-translational modulation of myo-inositol oxygenase by high glucose and related pathobiological stresses. J Biol Chem. 2011;286(31):27594-27611.
    1. Zhao G, He F, Wu C, et al. . Betaine in inflammation: mechanistic aspects and applications. Front Immunol. 2018;9:1070.
    1. Ejaz A, Martinez-Guino L, Goldfine AB, et al. . Dietary betaine supplementation increases Fgf21 levels to improve glucose homeostasis and reduce hepatic lipid accumulation in mice. Diabetes. 2016;65(4):902-912.
    1. Zhang L, Qi Y, ALuo Z, Liu S, Zhang Z, Zhou L. Betaine increases mitochondrial content and improves hepatic lipid metabolism. Food Funct. 2019;10(1):216-223.
    1. Hasegawa G, Nakano K, Ienaga K. Serum accumulation of a creatinine oxidative metabolite (NZ-419: 5-hydroxy-1- methylhydatoin) as an intrinsic antioxidant in diabetic patients with or without chronic kidney disease. Clin Nephrol. 2011;76(4):284-289.
    1. Ienaga K, Park CH, Yokozawa T. Protective effect of an intrinsic antioxidant, HMH (5-hydroxy-1-methylhydantoin; NZ-419), against cellular damage of kidney tubules. Exp Toxicol Pathol. 2013;65(5):559-566.
    1. Lee DM, Battson ML, Jarrell DK, et al. . SGLT2 inhibition via dapagliflozin improves generalized vascular dysfunction and alters the gut microbiota in type 2 diabetic mice. Cardiovasc Diabetol. 2018;17(1):62.
    1. Lever M, McEntyre CJ, George PM, Chambers ST. Is N,N-dimethylglycine N-oxide a choline and betaine metabolite? Biol Chem. 2017;398(7):775-784.
    1. Huo T, Xiong Z, Lu X, Cai S. Metabonomic study of biochemical changes in urinary of type 2 diabetes mellitus patients after the treatment of sulfonylurea antidiabetic drugs based on ultra-performance liquid chromatography/mass spectrometry. Biomed Chromatogr. 2015;29(1):115-122.
    1. Schalkwijk CG, Stehouwer CDA. Methylglyoxal, a highly reactive dicarbonyl compound, in diabetes, its vascular complications, and other age-related diseases. Physiol Rev. 2020;100(1):407-461.
    1. Ravichandran M, Priebe S, Grigolon G, et al. . Impairing L-threonine catabolism promotes healthspan through methylglyoxal-mediated proteohormesis. Cell Metab. 2018;27(4):914-925.e5.
    1. Jessen H. Taurine and beta-alanine transport in an established human kidney cell line derived from the proximal tubule. Biochim Biophys Acta. 1994;1194(1):44-52.
    1. van de Poll MC, Soeters PB, Deutz NE, Fearon KC, Dejong CH. Renal metabolism of amino acids: its role in interorgan amino acid exchange. Am J Clin Nutr. 2004;79(2):185-197.
    1. Tizianello A, De Ferrari G, Garibotto G, Gurreri G, Robaudo C. Renal metabolism of amino acids and ammonia in subjects with normal renal function and in patients with chronic renal insufficiency. J Clin Invest. 1980;65(5):1162-1173.
    1. Latva-Rasku A, Honka MJ, Kullberg J, et al. . The SGLT2 inhibitor dapagliflozin reduces liver fat but does not affect tissue insulin sensitivity: a randomized, double-blind, placebo-controlled study with 8-week treatment in type 2 diabetes patients. Diabetes Care. 2019;42(5):931-937.
    1. Martinez R, Al-Jobori H, Ali AM, et al. . Endogenous glucose production and hormonal changes in response to canagliflozin and liraglutide combination therapy. Diabetes. 2018;67(6):1182-1189.
    1. Ferrannini E, Baldi S, Frascerra S, et al. . Shift to fatty substrate utilization in response to sodium-glucose cotransporter 2 inhibition in subjects without diabetes and patients with type 2 diabetes. Diabetes. 2016;65(5):1190-1195.
    1. Paller MS, Patten M. Protective effects of glutathione, glycine, or alanine in an in vitro model of renal anoxia. J Am Soc Nephrol. 1992;2(8):1338-1344.
    1. Ghule AE, Jadhav SS, Bodhankar SL. Trigonelline ameliorates diabetic hypertensive nephropathy by suppression of oxidative stress in kidney and reduction in renal cell apoptosis and fibrosis in streptozotocin induced neonatal diabetic (nSTZ) rats. Int Immunopharmacol. 2012;14(4):740-748.
    1. Huang CF, Cheng ML, Fan CM, Hong CY, Shiao MS. Nicotinuric acid: a potential marker of metabolic syndrome through a metabolomics-based approach. Diabetes Care. 2013;36(6):1729-1731.
    1. Liu JJ, Liu S, Gurung RL, et al. . Urine tricarboxylic acid cycle metabolites predict progressive chronic kidney disease in Type 2 diabetes. J Clin Endocrinol Metab. 2018;103(12):4357-4364.
    1. Forbes JM, Thorburn DR. Mitochondrial dysfunction in diabetic kidney disease. Nat Rev Nephrol. 2018;14(5):291-312.
    1. Lees HJ, Swann JR, Wilson ID, Nicholson JK, Holmes E. Hippurate: the natural history of a mammalian-microbial cometabolite. J Proteome Res. 2013;12(4):1527-1546.
    1. Nochi Z, Olsen RKJ, Gregersen N. Short-chain acyl-CoA dehydrogenase deficiency: from gene to cell pathology and possible disease mechanisms. J Inherit Metab Dis. 2017;40(5):641-655.
    1. Afshinnia F, Zeng L, Byun J, et al. ; Michigan Kidney Translational Core CPROBE Investigator Group . Myeloperoxidase levels and its product 3-chlorotyrosine predict chronic kidney disease severity and associated coronary artery disease. Am J Nephrol. 2017;46(1):73-81.

Source: PubMed

3
Sottoscrivi