Liraglutide treatment improves postprandial lipid metabolism and cardiometabolic risk factors in humans with adequately controlled type 2 diabetes: A single-centre randomized controlled study

Niina Matikainen, Sanni Söderlund, Elias Björnson, Kirsi Pietiläinen, Antti Hakkarainen, Nina Lundbom, Marja-Riitta Taskinen, Jan Borén, Niina Matikainen, Sanni Söderlund, Elias Björnson, Kirsi Pietiläinen, Antti Hakkarainen, Nina Lundbom, Marja-Riitta Taskinen, Jan Borén

Abstract

Aims: Patients with type 2 diabetes and non-alcoholic fatty liver disease (NAFLD) exhibit considerable residual risk for cardiovascular disease (CVD). There is, therefore, increasing interest in targeting postprandial lipid metabolism and remnant cholesterol. Treatment with the glucagon-like peptide 1 (GLP-1) analogue liraglutide reduces CVD risk by mechanisms that remain unexplained in part. Here we investigated the effects of liraglutide intervention on ectopic fat depots, hepatic lipogenesis and fat oxidation, postprandial lipid metabolism and glycaemia in humans with type 2 diabetes.

Methods: The effect of liraglutide was investigated in 22 patients with adequately controlled type 2 diabetes. Patients were randomly allocated, in a single-blind fashion, to either liraglutide 1.8 mg or placebo once daily for 16 weeks. Because liraglutide is known to promote weight loss, the study included dietary counselling to achieve similar weight loss in the liraglutide and placebo groups. Cardiometabolic responses to a high-fat mixed meal were measured before and at the end of the liraglutide intervention.

Results: Weight loss at Week 16 was similar between the groups: -2.4 kg (-2.5%) in the liraglutide group and -2.1 kg (-2.2%) in the placebo group. HBA1c improved by 6.4 mmol/mol (0.6%) in the liraglutide group (P = 0.005). Liver fat decreased in both groups, by 31% in the liraglutide group and by 18% in the placebo group, but there were no significant changes in the rate of hepatic de novo lipogenesis or β-hydroxybutyrate levels, a marker of fat oxidation. We observed significant postprandial decreases in triglycerides only in plasma, chylomicrons and VLDL, and remnant particle cholesterol after treatment in the liraglutide group. Fasting and postprandial apoCIII concentrations decreased after liraglutide intervention and these changes were closely related to reduced glycaemia. In relative importance analysis, approximately half of the changes in postprandial lipids were explained by reductions in apoCIII concentrations, whereas less than 10% of the variation in postprandial lipids was explained by reductions in weight, glycaemic control, liver fat or postprandial insulin responses.

Conclusions: Intervention with liraglutide for 16 weeks produces multiple improvements in cardiometabolic risk factors that were not seen in the placebo group, despite similar weight loss. Of particular importance was a marked reduction in postprandial atherogenic remnant particles. The underlying mechanism may be improved glycaemic control, which leads to reduced expression of apoCIII, a key regulator of hypertriglyceridaemia in hyperglycaemic patients.

Keywords: GLP-1-agonist; apolipoprotein C3; atherogenic dyslipidaemia; de novo lipogenesis; liraglutide; liver fat; postprandial lipids; remnant lipoproteins.

Conflict of interest statement

The authors report no duality of interest.

© 2018 The Authors. Diabetes, Obesity and Metabolism published by John Wiley & Sons Ltd.

Figures

Figure 1
Figure 1
Responses of plasma glucose, insulin, β‐hydroxybutyrate and NEFA after a fat‐rich mixed meal before treatment (open squares) and at week 16 (filled squares) in patients with type 2 diabetes being treated with liraglutide (n = 15) or receiving placebo (n = 7). P values were calculated using the ANOVA repeated measures test for within‐group differences
Figure 2
Figure 2
Responses of plasma triglycerides (TG), chylomicron TG, VLDL1 TG and plasma apoB48 after a fat‐rich mixed meal before treatment (open squares) and at week 16 (filled squares) in subjects with type 2 diabetes treated with liraglutide (n = 15) or placebo (n = 7). P values were calculated using the ANOVA repeated measures test for within‐group differences
Figure 3
Figure 3
Responses of plasma apoCIII, remnant‐like particle cholesterol (RLP‐C) and triglyceride‐rich lipoprotein cholesterol (TRL‐C) after a fat‐rich mixed meal before treatment (open squares) and at week 16 (filled squares) in subjects with type 2 diabetes treated with liraglutide (n = 15) or placebo (n = 7). P values were calculated using the ANOVA repeated measures test for within‐group differences
Figure 4
Figure 4
Relative importance analysis demonstrating the percentage contribution of changes in various metabolic parameters to reduction (delta, Δ) in A, plasma TG area under the curve (AUC); B, plasma remnant‐like particle cholesterol (RLP‐C) AUC; C, triglyceride‐rich lipoprotein cholesterol (TRL‐C); and D, plasma apoCIII AUC. Height of bar shows the absolute percentage contribution, calculated using an average of four different methods (see the Material and Methods section). Variables to be included in the regression model were chosen based on hypothesized physiological relevance to the trait of interest. Exclusion of Matsuda index in favour of HOMA2‐IR did not significantly affect the results

References

    1. Cholesterol Treatment Trialists' (CTT) Collaborators , Mihaylova B, Emberson J, et al. The effects of lowering LDL cholesterol with statin therapy in people at low risk of vascular disease: meta‐analysis of individual data from 27 randomised trials. Lancet. 2012;380:581‐590.
    1. Gaede P, Oellgaard J, Carstensen B, et al. Years of life gained by multifactorial intervention in patients with type 2 diabetes mellitus and microalbuminuria: 21 years follow‐up on the Steno‐2 randomised trial. Diabetologia. 2016;59:2298‐2307.
    1. Wild SH, Walker JJ, Morling JR, et al. Cardiovascular disease, cancer, and mortality among people with type 2 diabetes and alcoholic or nonalcoholic fatty liver disease hospital admission. Diabetes Care. 2018;41:341‐347.
    1. Varbo A, Freiberg JJ, Nordestgaard BG. Remnant cholesterol and myocardial infarction in normal weight, overweight, and obese individuals from the Copenhagen General Population Study. Clin Chem. 2018;64:219‐230.
    1. Varbo A, Benn M, Nordestgaard BG. Remnant cholesterol as a cause of ischemic heart disease: evidence, definition, measurement, atherogenicity, high risk patients, and present and future treatment. Pharmacol Ther. 2014;141:358‐367.
    1. Taskinen MR, Boren J. New insights into the pathophysiology of dyslipidemia in type 2 diabetes. Atherosclerosis. 2015;239:483‐495.
    1. Nordestgaard BG, Langsted A, Mora S, et al. Fasting is not routinely required for determination of a lipid profile: clinical and laboratory implications including flagging at desirable concentration cut‐points‐a joint consensus statement from the European Atherosclerosis Society and European Federation of Clinical Chemistry and Laboratory Medicine. Eur Heart J. 2015;37:1944‐1958.
    1. Cohn JS, Johnson EJ, Millar JS, et al. Contribution of apoB‐48 and apoB‐100 triglyceride‐rich lipoproteins (TRL) to postprandial increases in the plasma concentration of TRL triglycerides and retinyl esters. J Lipid Res. 1993;34:2033‐2040.
    1. Boren J, Matikainen N, Adiels M, Taskinen MR. Postprandial hypertriglyceridemia as a coronary risk factor. Clin Chim Acta. 2014;431:131‐142.
    1. Brunzell JD, Hazzard WR, Porte D Jr, Bierman EL. Evidence for a common, saturable, triglyceride removal mechanism for chylomicrons and very low density lipoproteins in man. J Clin Invest. 1973;52:1578‐1585.
    1. Adiels M, Matikainen N, Westerbacka J, et al. Postprandial accumulation of chylomicrons and chylomicron remnants is determined by the clearance capacity. Atherosclerosis. 2012;222:222‐228.
    1. Matikainen N, Adiels M, Soderlund S, et al. Hepatic lipogenesis and a marker of hepatic lipid oxidation, predict postprandial responses of triglyceride‐rich lipoproteins. Obesity (Silver Spring). 2014;22:1854‐1859.
    1. Prasad‐Reddy L, Isaacs D. A clinical review of GLP‐1 receptor agonists: efficacy and safety in diabetes and beyond. Drugs Context. 2015;4:212283.
    1. Marso SP, Daniels GH, Brown‐Frandsen K, et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2016;375:311‐322.
    1. Hermansen K, Baekdal TA, During M, et al. Liraglutide suppresses postprandial triglyceride and apolipoprotein B48 elevations after a fat‐rich meal in patients with type 2 diabetes: a randomized, double‐blind, placebo‐controlled, cross‐over trial. Diabetes Obes Metab. 2013;15:1040‐1048.
    1. Bunck MC, Corner A, Eliasson B, et al. One‐year treatment with exenatide vs. insulin glargine: effects on postprandial glycemia, lipid profiles, and oxidative stress. Atherosclerosis. 2010;212:223‐229.
    1. Zhong J, Maiseyeu A, Rajagopalan S. Lipoprotein effects of incretin analogs and dipeptidyl peptidase 4 inhibitors. Clin Lipidol. 2015;10:103‐112.
    1. Junker AE, Gluud L, Holst JJ, Knop FK, Vilsboll T. Diabetic and nondiabetic patients with nonalcoholic fatty liver disease have an impaired incretin effect and fasting hyperglucagonaemia. J Intern Med. 2016;279:485‐493.
    1. Matikainen N, Bogl LH, Hakkarainen A, et al. GLP‐1 responses are heritable and blunted in acquired obesity with high liver fat and insulin resistance. Diabetes Care. 2014;37:242‐251.
    1. Petit JM, Verges B. GLP‐1 receptor agonists in NAFLD. Diabetes Metab. 2017;43(suppl 1):2S28‐2S33.
    1. Armstrong MJ, Gaunt P, Aithal GP, et al. Liraglutide safety and efficacy in patients with non‐alcoholic steatohepatitis (LEAN): a multicentre, double‐blind, randomised, placebo‐controlled phase 2 study. Lancet. 2016;387:679‐690.
    1. Kenny PR, Brady DE, Torres DM, Ragozzino L, Chalasani N, Harrison SA. Exenatide in the treatment of diabetic patients with non‐alcoholic steatohepatitis: a case series. Am J Gastroenterol. 2010;105:2707‐2709.
    1. Drucker DJ, Habener JF, Holst JJ. Discovery, characterization, and clinical development of the glucagon‐like peptides. J Clin Invest. 2017;127:4217‐4227.
    1. Jendle J, Nauck MA, Matthews DR, et al. Weight loss with liraglutide, a once‐daily human glucagon‐like peptide‐1 analogue for type 2 diabetes treatment as monotherapy or added to metformin, is primarily as a result of a reduction in fat tissue. Diabetes Obes Metab. 2009;11:1163‐1172.
    1. Nauck M, Frid A, Hermansen K, et al. Long‐term efficacy and safety comparison of liraglutide, glimepiride and placebo, all in combination with metformin in type 2 diabetes: 2‐year results from the LEAD‐2 study. Diabetes Obes Metab. 2013;15:204‐212.
    1. Nauck M, Frid A, Hermansen K, et al. Efficacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin, in type 2 diabetes: the LEAD (liraglutide effect and action in diabetes)‐2 study. Diabetes Care. 2009;32:84‐90.
    1. Taskinen MR, Adiels M, Westerbacka J, et al. Dual metabolic defects are required to produce hypertriglyceridemia in obese subjects. Arterioscler Thromb Vasc Biol. 2011;31:2144‐2150.
    1. Ryysy L, Hakkinen AM, Goto T, et al. Hepatic fat content and insulin action on free fatty acids and glucose metabolism rather than insulin absorption are associated with insulin requirements during insulin therapy in type 2 diabetic patients. Diabetes. 2000;49:749‐758.
    1. Matikainen N, Manttari S, Schweizer A, et al. Vildagliptin therapy reduces postprandial intestinal triglyceride‐rich lipoprotein particles in patients with type 2 diabetes. Diabetologia. 2006;49:2049‐2057.
    1. Lindeman R, Merenda P. Gold R Introduction to Bivariate and Multidimensional Analysis. Glenview, IL: Scott Foresman; 1980.
    1. Pratt JW. Dividing the indivisible: using simple symmetry to partition variance explained Proceedings of the Second International Tampere Conference in Statistics. Tampere, Finland: Department of Mathematical Sciences, University of Tampere; 1987:245‐260.
    1. Grömping U. Relative importance for linear regression in R: the package relaimpo. J Stat Softw. 2006;17:1‐27.
    1. Taskinen MR, Boren J. Why is apolipoprotein CIII emerging as a novel therapeutic target to reduce the burden of cardiovascular disease? Curr Atheroscler Rep. 2016;18:59.
    1. Nauck MA, Meier JJ, Cavender MA, Abd El Aziz M, Drucker DJ. Cardiovascular actions and clinical outcomes with glucagon‐like peptide‐1 receptor agonists and dipeptidyl peptidase‐4 inhibitors. Circulation. 2017;136:849‐870.
    1. Eliasson B, Moller‐Goede D, Eeg‐Olofsson K, et al. Lowering of postprandial lipids in individuals with type 2 diabetes treated with alogliptin and/or pioglitazone: a randomised double‐blind placebo‐controlled study. Diabetologia. 2012;55:915‐925.
    1. Matikainen N, Taskinen MR. The effect of vildagliptin therapy on atherogenic postprandial remnant particles and LDL particle size in subjects with type 2 diabetes. Diabet Med. 2013;30:756‐757.
    1. Schwartz EA, Koska J, Mullin MP, Syoufi I, Schwenke DC, Reaven PD. Exenatide suppresses postprandial elevations in lipids and lipoproteins in individuals with impaired glucose tolerance and recent onset type 2 diabetes mellitus. Atherosclerosis. 2010;212:217‐222.
    1. Tabas I, Williams KJ, Boren J. Subendothelial lipoprotein retention as the initiating process in atherosclerosis: update and therapeutic implications. Circulation. 2007;116:1832‐1844.
    1. Boren J, Williams KJ. The central role of arterial retention of cholesterol‐rich apolipoprotein‐B‐containing lipoproteins in the pathogenesis of atherosclerosis: a triumph of simplicity. Curr Opin Lipidol. 2016;27:473‐483.
    1. Boren J, Watts GF, Adiels M, et al. Kinetic and related determinants of plasma triglyceride concentration in abdominal obesity: multicenter tracer kinetic study. Arterioscler Thromb Vasc Biol. 2015;35:2218‐2224.
    1. Jorgensen AB, Frikke‐Schmidt R, West AS, Grande P, Nordestgaard BG, Tybjaerg‐Hansen A. Genetically elevated non‐fasting triglycerides and calculated remnant cholesterol as causal risk factors for myocardial infarction. Eur Heart J. 2013;34:1826‐1833.
    1. Adiels M, Taskinen MR, Packard C, et al. Overproduction of large VLDL particles is driven by increased liver fat content in man. Diabetologia. 2006;49:755‐765.
    1. Ben‐Shlomo S, Zvibel I, Shnell M, et al. Glucagon‐like peptide‐1 reduces hepatic lipogenesis via activation of AMP‐activated protein kinase. J Hepatol. 2011;54:1214‐1223.
    1. Svegliati‐Baroni G, Saccomanno S, Rychlicki C, et al. Glucagon‐like peptide‐1 receptor activation stimulates hepatic lipid oxidation and restores hepatic signalling alteration induced by a high‐fat diet in nonalcoholic steatohepatitis. Liver Int. 2011;31:1285‐1297.
    1. Armstrong MJ, Hull D, Guo K, et al. Glucagon‐like peptide 1 decreases lipotoxicity in non‐alcoholic steatohepatitis. J Hepatol. 2016;64:399‐408.
    1. Pyke C, Heller RS, Kirk RK, et al. GLP‐1 receptor localization in monkey and human tissue: novel distribution revealed with extensively validated monoclonal antibody. Endocrinology. 2014;155:1280‐1290.
    1. Hiukka A, Fruchart‐Najib J, Leinonen E, Hilden H, Fruchart JC, Taskinen MR. Alterations of lipids and apolipoprotein CIII in very low density lipoprotein subspecies in type 2 diabetes. Diabetologia. 2005;48:1207‐1215.
    1. Hiukka A, Stahlman M, Pettersson C, et al. ApoCIII‐enriched LDL in type 2 diabetes displays altered lipid composition, increased susceptibility for sphingomyelinase, and increased binding to biglycan. Diabetes. 2009;58:2018‐2026.
    1. Caron S, Verrijken A, Mertens I, et al. Transcriptional activation of apolipoprotein CIII expression by glucose may contribute to diabetic dyslipidemia. Arterioscler Thromb Vasc Biol. 2011;31:513‐519.
    1. Mardinoglu A, Wu H, Bjornson E, et al. An integrated understanding of the rapid metabolic benefits of a carbohydrate‐restricted diet on hepatic steatosis in humans. Cell Metab. 2018;27:559‐571.

Source: PubMed

3
購読する