Glucagon-like peptide 1 decreases lipotoxicity in non-alcoholic steatohepatitis

Matthew J Armstrong, Diana Hull, Kathy Guo, Darren Barton, Jonathan M Hazlehurst, Laura L Gathercole, Maryam Nasiri, Jinglei Yu, Stephen C Gough, Philip N Newsome, Jeremy W Tomlinson, Matthew J Armstrong, Diana Hull, Kathy Guo, Darren Barton, Jonathan M Hazlehurst, Laura L Gathercole, Maryam Nasiri, Jinglei Yu, Stephen C Gough, Philip N Newsome, Jeremy W Tomlinson

Abstract

Background & aims: Insulin resistance and lipotoxicity are pathognomonic in non-alcoholic steatohepatitis (NASH). Glucagon-like peptide-1 (GLP-1) analogues are licensed for type 2 diabetes, but no prospective experimental data exists in NASH. This study determined the effect of a long-acting GLP-1 analogue, liraglutide, on organ-specific insulin sensitivity, hepatic lipid handling and adipose dysfunction in biopsy-proven NASH.

Methods: Fourteen patients were randomised to 1.8mg liraglutide or placebo for 12-weeks of the mechanistic component of a double-blind, randomised, placebo-controlled trial (ClinicalTrials.gov-NCT01237119). Patients underwent paired hyperinsulinaemic euglycaemic clamps, stable isotope tracers, adipose microdialysis and serum adipocytokine/metabolic profiling. In vitro isotope experiments on lipid flux were performed on primary human hepatocytes.

Results: Liraglutide reduced BMI (-1.9 vs. +0.04kg/m(2); p<0.001), HbA1c (-0.3 vs. +0.3%; p<0.01), cholesterol-LDL (-0.7 vs. +0.05mmol/L; p<0.01), ALT (-54 vs. -4.0IU/L; p<0.01) and serum leptin, adiponectin, and CCL-2 (all p<0.05). Liraglutide increased hepatic insulin sensitivity (-9.36 vs. -2.54% suppression of hepatic endogenous glucose production with low-dose insulin; p<0.05). Liraglutide increased adipose tissue insulin sensitivity enhancing the ability of insulin to suppress lipolysis both globally (-24.9 vs. +54.8pmol/L insulin required to ½ maximally suppress serum non-esterified fatty acids; p<0.05), and specifically within subcutaneous adipose tissue (p<0.05). In addition, liraglutide decreased hepatic de novo lipogenesis in vivo (-1.26 vs. +1.30%; p<0.05); a finding endorsed by the effect of GLP-1 receptor agonist on primary human hepatocytes (24.6% decrease in lipogenesis vs. untreated controls; p<0.01).

Conclusions: Liraglutide reduces metabolic dysfunction, insulin resistance and lipotoxicity in the key metabolic organs in the pathogenesis of NASH. Liraglutide may offer the potential for a disease-modifying intervention in NASH.

Keywords: Adipose tissue; Glucagon-like peptide 1; Insulin sensitivity; Lipolysis; Non-alcoholic fatty liver.

Copyright © 2015 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved.

Figures

Graphical abstract
Graphical abstract
Fig. 1
Fig. 1
Liraglutide significantly reduces hepatic insulin resistance, but has no effect on muscle insulin sensitivity. (A) Tukey box-and-whisker plots highlight that liraglutide significantly increased the suppression of hepatic EGP with low-dose insulin compared to placebo. (B) Weight-adjusted Gd rates at low- and high-dose insulin phases of the euglycaemic clamp highlight that liraglutide did not change muscle insulin sensitivity (Gd) compared to placebo. ∗p <0.05 treatment vs. baseline (using paired Wilcoxon signed-rank tests). Unpaired Mann-Whitney tests were used to compare liraglutide vs. placebo.
Fig. 2
Fig. 2
Liraglutide significant reduces whole body lipolysis and adipose insulin resistance. (A) Tukey box-and-whisker plots representing NEFA concentrations at the basal and hyperinsulinaemic phases of euglycaemic clamp. Liraglutide reduced NEFA at every phase of the clamp compared to placebo. (B) Tukey box-and-whisker plots representing the effect of liraglutide vs. placebo on insulin concentration required to achieve ½ maximal suppression of circulating NEFA (INS-½-max NEFA). ∗p <0.05 treatment vs. baseline (using paired Wilcoxon signed-rank tests). Unpaired Mann-Whitney tests were used to compare liraglutide vs. placebo.
Fig. 3
Fig. 3
Liraglutide reduces abdominal SAT lipolysis and insulin resistance (IR). Line graphs (A, B) represent the mean ± SE concentrations of glycerol in the interstitial fluid measured from the SAT of each participant using in situ microdialysis throughout the 6 hour euglycaemic clamp. Liraglutide (A) decreased glycerol release throughout the clamp, whereas there were no clear differences after placebo treatment (B). (C) Tukey box-and-whisker plots (area under the curve analysis) highlight that liraglutide significantly reduced glycerol release from SAT in response to both low-dose and high-dose insulin compared to placebo, representing decreased abdominal SAT IR. ∗p <0.05 treatment vs. baseline (using paired Wilcoxon signed-rank tests). Unpaired Mann-Whitney tests were used to compare liraglutide vs. placebo.
Fig. 4
Fig. 4
GLP-1R analogues significantly reduce hepatic DNL in vivo and in vitro, respectively. (A) Tukey box-and-whisker plots demonstrate that 12 weeks treatment with liraglutide significantly reduces hepatic DNL compared to placebo in patients with NASH (data ∗p <0.05 vs. placebo). (B) Exendin-4 significantly reduces DNL in primary human hepatocytes in culture. DNL was defined by the amount of 14C acetate incorporated into intracellular lipid in primary human hepatocytes. Exendin-4 has no effect on NEFA uptake (3H-palmitate taken up by the cells) (C) and β-oxidation (amount of 3H water released from the cells) (D) in hepatoma cell lines (HuH7). In vitro data in (B, C, D) are presented as mean ± SE percentages of the untreated controls. Untreated control was DMEM with 0.5% BSA. Insulin 5 nM served as a positive control. In vitro experiments were performed four times with each treatment in quadruplicate. ∗p <0.05 vs. untreated control.
Fig. 5
Fig. 5
GLP-1R analogue, exendin-4, has direct anti-steatotic effects on hepatocytes in vitro. Exendin-4 (100 nM) reduces triglyceride content of NEFA-loaded HuH7 cells, as represented by (A) Oil Red O Staining (original magnification 10×, 40×) and (B) colorimetric triglyceride quantification assay. ∗p <0.05, ∗∗p <0.01 vs. untreated cells. (This figure appears in colour on the web.)

References

    1. Armstrong M.J., Adams L.A., Canbay A., et al. Extra-hepatic complications of nonalcoholic fatty liver disease. Hepatology. 2014;59:1174–1197.
    1. Sanyal A.J., Brunt E.M., Kleiner D.E., et al. Endpoints and clinical trial design for nonalcoholic steatohepatitis. Hepatology. 2011;54:344–353.
    1. Marchesini G., Brizi M., Bianchi G., et al. Nonalcoholic fatty liver disease: a feature of the metabolic syndrome. Diabetes. 2001;50:1844–1850.
    1. Lomonaco R., Ortiz-Lopez C., Orsak B., et al. Effect of adipose tissue insulin resistance on metabolic parameters and liver histology in obese patients with nonalcoholic fatty liver disease. Hepatology. 2012;55:1389–1397.
    1. Musso G., Cassader M., De Michieli F., et al. Nonalcoholic steatohepatitis versus steatosis: adipose tissue insulin resistance and dysfunctional response to fat ingestion predict liver injury and altered glucose and lipoprotein metabolism. Hepatology. 2012;56:933–942.
    1. Donnelly K.L., Smith C.I., Schwarzenberg S.J., et al. Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. J Clin Invest. 2005;115:1343–1351.
    1. Lambert J.E., Ramos-Roman M.A., Browning J.D., et al. Increased de novo lipogenesis is a distinct characteristic of individuals with nonalcoholic fatty liver disease. Gastroenterology. 2014;146:726–735.
    1. Cusi K. Role of obesity and lipotoxicity in the development of nonalcoholic steatohepatitis: pathophysiology and clinical implications. Gastroenterology. 2012;142:e6.
    1. Armstrong M.J., Houlihan D.D., Rowe I.A., et al. Safety and efficacy of liraglutide in patients with type 2 diabetes and elevated liver enzymes: individual patient data meta-analysis of the LEAD program. Aliment Pharmacol Ther. 2013;37:234–242.
    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. Sharma S., Mells J.E., Fu P.P., et al. GLP-1 analogs reduce hepatocyte steatosis and improve survival by enhancing the unfolded protein response and promoting macroautophagy. PLoS One. 2011;6:e25269.
    1. Shirakawa J., Fujii H., Ohnuma K., et al. Diet-induced adipose tissue inflammation and liver steatosis are prevented by DPP-4 inhibition in diabetic mice. Diabetes. 2011;60:1246–1257.
    1. Mells J.E., Fu P.P., Sharma S., et al. Glp-1 analog, liraglutide, ameliorates hepatic steatosis and cardiac hypertrophy in C57BL/6J mice fed a Western diet. Am J Physiol Gastrointest Liver Physiol. 2012;302:G225–G235.
    1. Ding X., Saxena N.K., Lin S., et al. Exendin-4, a glucagon-like protein-1 (GLP-1) receptor agonist, reverses hepatic steatosis in ob/ob mice. Hepatology. 2006;43:173–181.
    1. Lee J., Hong S.-W., Chae S.W., et al. Exendin-4 improves steatohepatitis by increasing Sirt1 expression in high-fat diet-induced obese C57BL/6J mice. PLoS One. 2012;7:e31394.
    1. Trevaskis J.L., Griffin P.S., Wittmer C., et al. Glucagon-like peptide-1 receptor agonism improves metabolic, biochemical, and histopathological indices of nonalcoholic steatohepatitis in mice. Am J Physiol Gastrointest Liver Physiol. 2012;302:G762–G772.
    1. Lee Y.-S., Shin S., Shigihara T., et al. Glucagon-like peptide-1 gene therapy in obese diabetic mice results in long-term cure of diabetes by improving insulin sensitivity and reducing hepatic gluconeogenesis. Diabetes. 2007;56:1671–1679.
    1. Zhang L., Yang M., Ren H., et al. GLP-1 analogue prevents NAFLD in ApoE KO mice with diet and Acrp30 knockdown by inhibiting c-JNK. Liver Int. 2013;33:794–804.
    1. D’Alessio D.A., Kahn S.E., Leusner C.R., et al. Glucagon-like peptide 1 enhances glucose tolerance both by stimulation of insulin release and by increasing insulin-independent glucose disposal. J Clin Invest. 1994;93:2263–2266.
    1. Zander M., Madsbad S., Madsen J.L., et al. Effect of 6-week course of glucagon-like peptide 1 on glycaemic control, insulin sensitivity, and beta-cell function in type 2 diabetes: a parallel-group study. Lancet. 2002;359:824–830.
    1. Prigeon R.L., Quddusi S., Paty B., et al. Suppression of glucose production by GLP-1 independent of islet hormones: a novel extrapancreatic effect. Am J Physiol Endocrinol Metab. 2003;285:E701–E707.
    1. Orskov L., Holst J.J., Møller J., et al. GLP-1 does not acutely affect insulin sensitivity in healthy man. Diabetologia. 1996;39:1227–1232.
    1. Armstrong M.J., Barton D., Gaunt P., et al. Liraglutide efficacy and action in non-alcoholic steatohepatitis (LEAN): study protocol for a phase II multicentre, double-blinded, randomised, controlled trial. BMJ Open. 2013;3:e003995.
    1. Armstrong M.J., Hazlehurst J.M., Hull D. Abdominal subcutaneous adipose tissue insulin resistance and lipolysis in patients with non-alcoholic steatohepatitis. Diabetes Obes Metab. 2014;16:651–660.
    1. Hazlehurst J.M., Gathercole L.L., Nasiri M., et al. Glucocorticoids fail to cause insulin resistance in human subcutaneous adipose tissue in vivo. J Clin Endocrinol Metab. 2013;98:1631–1640.
    1. Gathercole L.L., Morgan S.A., Bujalska I.J., et al. Regulation of lipogenesis by glucocorticoids and insulin in human adipose tissue. PLoS One. 2011;6:e26223.
    1. Seghieri M., Rebelos E., Gastaldelli A., et al. Direct effect of GLP-1 infusion on endogenous glucose production in humans. Diabetologia. 2013;56:156–161.
    1. Gutniak M., Orskov C., Holst J.J., et al. Antidiabetogenic effect of glucagon-like peptide-1 (7–36)amide in normal subjects and patients with diabetes mellitus. N Engl J Med. 1992;326:1316–1322.
    1. Toft-Nielson M., Madsbad S., Holst J.J. The effect of glucagon-like peptide I (GLP-I) on glucose elimination in healthy subjects depends on the pancreatic glucoregulatory hormones. Diabetes. 1996;45:552–556.
    1. Gedulin B.R., Nikoulina S.E., Smith P.A., et al. Exenatide (exendin-4) improves insulin sensitivity and {beta}-cell mass in insulin-resistant obese fa/fa Zucker rats independent of glycemia and body weight. Endocrinology. 2005;146:2069–2076.
    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. Campbell J.E., Drucker D.J. Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab. 2013;17:819–837.
    1. Boden G. Fatty acid-induced inflammation and insulin resistance in skeletal muscle and liver. Curr Diab Rep. 2006;6:177–181.
    1. Gao H., Wang X., Zhang Z., et al. GLP-1 amplifies insulin signaling by up-regulation of IRbeta, IRS-1 and Glut4 in 3T3-L1 adipocytes. Endocrine. 2007;32:90–95.
    1. Vendrell J., El Bekay R., Peral B., et al. Study of the potential association of adipose tissue GLP-1 receptor with obesity and insulin resistance. Endocrinology. 2011;152:4072–4079.
    1. Perea A., Viñambres C., Clemente F., et al. GLP-1 (7–36) amide: effects on glucose transport and metabolism in rat adipose tissue. Horm Metab Res. 1997;29:417–421.
    1. Villanueva-Peñacarrillo M.L., Márquez L., González N., et al. Effect of GLP-1 on lipid metabolism in human adipocytes. Horm Metab Res. 2001;33:73–77.
    1. Gastaldelli A., Harrison S.A., Belfort-Aguilar R., et al. Importance of changes in adipose tissue insulin resistance to histological response during thiazolidinedione treatment of patients with nonalcoholic steatohepatitis. Hepatology. 2009;50:1087–1093.
    1. Bertin E., Arner P., Bolinder J., et al. Action of glucagon and glucagon-like peptide-1-(7–36) amide on lipolysis in human subcutaneous adipose tissue and skeletal muscle in vivo. J Clin Endocrinol Metab. 2001;86:1229–1234.
    1. Gastaldelli A., Harrison S., Belfort-Aguiar R., et al. Pioglitazone in the treatment of NASH: the role of adiponectin. Aliment Pharmacol Ther. 2010;32:769–775.

Source: PubMed

3
Iratkozz fel