Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Resolution of NASH and hepatic fibrosis by the GLP-1R and GCGR dual-agonist cotadutide via modulating mitochondrial function and lipogenesis

Abstract

Non-alcoholic fatty liver disease and steatohepatitis are highly associated with obesity and type 2 diabetes mellitus. Cotadutide, a glucagon-like protein-1 receptor (GLP-1R) and glucagon receptor (GCGR) agonist, was shown to reduce blood glycaemia, body weight and hepatic steatosis in people with type 2 diabetes mellitus. Here, we demonstrate that the effects of cotadutide in reducing body weight and food intake and improving glucose control are predominantly mediated through Glp-1 signalling, whereas its action on the liver to reduce lipid content, drive glycogen flux and improve mitochondrial turnover and function are directly mediated through Gcg signalling. This was confirmed by the identification of phosphorylation sites on key lipogenic and glucose metabolism enzymes in liver of mice treated with cotadutide. Complementary metabolomic and transcriptomic analyses implicated lipogenic, fibrotic and inflammatory pathways, consistent with a unique therapeutic contribution of GCGR agonism by cotadutide in vivo. Notably, cotadutide also alleviated fibrosis to a greater extent than liraglutide or obeticholic acid, despite dose adjustment to achieve similar weight loss in two preclinical mouse models of NASH. Thus, cotadutide, via direct hepatic (GcgR) and extrahepatic (Glp-1R) effects, exerts multifactorial improvement in liver function and is a promising therapeutic option for the treatment of steatohepatitis.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Metabolic and hepatic parameters following 2-week treatment of DIO Glp-1R WT or KO mice with cotadutide, liraglutide, g1437 or liraglutide+g1437.
Fig. 2: Temporal changes in metabolic and hepatic parameters following 1-week treatment of DIO C57Bl6/J mice with cotadutide, liraglutide, g1437 or liraglutide+g1437.
Fig. 3: Hepatic glycogen flux following 1-week treatment of DIO C57Bl6/J mice with cotadutide, liraglutide or g1437.
Fig. 4: Cotadutide-induced changes in the hepatic phosphoproteome of carbohydrate-metabolism- and lipid-metabolism-related molecules.
Fig. 5: Cotadutide induces mitochondrial turnover and improves mitochondrial respiration.
Fig. 6: Superior efficacy of cotadutide on NASH end points in C57BL6/J mice fed an AMLN diet for 29 weeks, compared with liraglutide and OCA treatment.
Fig. 7: Superior reductions in liver lipids with cotadutide versus liraglutide in an ob/ob AMLN mouse model of NASH.
Fig. 8: Cotadutide further reduces hepatic fibrosis and inflammation compared with that after liraglutide or diet switch in an ob/ob AMLN mouse model of NASH.
Fig. 9: Summary of cotadutide mechanism of action at target organs.

Similar content being viewed by others

Data availability

The datasets generated during these studies are available from the corresponding author on reasonable request. These datasets include: in vivo data for the mouse studies and encompass metabolic, biochemical and histological data/images; mass spectrometry data from mouse liver and/or serum, mouse hepatocytes or human hepatocytes; imaging and quantitation data for mitochondrial analyses; qRT–PCR data for select gene expression analyses. RNA-seq datasets are available in the SRA database with accession number PRJNA574649.

References

  1. Younossi, Z. et al. Global burden of NAFLD and NASH: trends, predictions, risk factors and prevention. Nat. Rev. Gastroenterol. Hepatol. 15, 11–20 (2018).

    Article  PubMed  Google Scholar 

  2. Ibrahim, S. H., Hirsova, P. & Gores, G. J. Non-alcoholic steatohepatitis pathogenesis: sublethal hepatocyte injury as a driver of liver inflammation. Gut 67, 963–972 (2018).

    Article  CAS  PubMed  Google Scholar 

  3. Friedman, S. L., Neuschwander-Tetri, B. A., Rinella, M. & Sanyal, A. J. Mechanisms of NAFLD development and therapeutic strategies. Nat. Med. 24, 908–922 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Cusi, K. Treatment of patients with type 2 diabetes and non-alcoholic fatty liver disease: current approaches and future directions. Diabetologia 59, 1112–1120 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Bueter, M. & le Roux, C. W. Gastrointestinal hormones, energy balance and bariatric surgery. Int. J. Obes. 35(Suppl. 3), S35–S393 (2011).

    Article  CAS  Google Scholar 

  6. Ionut, V., Burch, M., Youdim, A. & Bergman, R. N. Gastrointestinal hormones and bariatric surgery-induced weight loss. Obesity 21, 1093–1103 (2013).

    Article  CAS  PubMed  Google Scholar 

  7. Meek, C. L., Lewis, H. B., Reimann, F., Gribble, F. M. & Park, A. J. The effect of bariatric surgery on gastrointestinal and pancreatic peptide hormones. Peptides 77, 28–37 (2016).

    Article  CAS  PubMed  Google Scholar 

  8. Cohen, M. A. et al. Oxyntomodulin suppresses appetite and reduces food intake in humans. J. Clin. Endocrinol. Metab. 88, 4696–4701 (2003).

    Article  CAS  PubMed  Google Scholar 

  9. Wynne, K. et al. Oxyntomodulin increases energy expenditure in addition to decreasing energy intake in overweight and obese humans: a randomised controlled trial. Int. J. Obes. 30, 1729–1736 (2006).

    Article  CAS  Google Scholar 

  10. Scott, R., Minnion, J., Tan, T. & Bloom, S. R. Oxyntomodulin analogue increases energy expenditure via the Gcg receptor. Peptides 104, 70–77 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Ambery, P. et al. Cotadutide, a GLP-1 and Gcg receptor dual agonist, in obese or overweight patients with type 2 diabetes: a randomised, controlled, double-blind, ascending dose and phase 2a study. Lancet 10140, 2607–2618 (2018).

    Article  Google Scholar 

  12. Day, J. W. et al. A new Gcg and GLP-1 co-agonist eliminates obesity in rodents. Nat. Chem. Biol. 5, 749–757 (2009).

    Article  CAS  PubMed  Google Scholar 

  13. Henderson, S. J. et al. Robust anti-obesity and metabolic effects of a dual GLP-1/Gcg receptor peptide agonist in rodents and non-human primates. Diabetes Obes. Metab. 18, 1176–1190 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Khajavi, N., Biebermann, H., Tschop, M. & DiMarchi, R. Treatment of diabetes and obesity by rationally designed peptide agonists functioning at multiple metabolic receptors. Endocr. Dev. 32, 165–182 (2017).

    Article  PubMed  Google Scholar 

  15. Tan, T. M. et al. Coadministration of Gcg-like peptide-1 during Gcg infusion in humans results in increased energy expenditure and amelioration of hyperglycemia. Diabetes 62, 1131–1138 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Pocai, A. et al. Glucagon-like peptide 1/Gcg receptor dual agonism reverses obesity in mice. Diabetes 58, 2258–2266 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Patel, V. et al. Coagonist of GLP-1 and Gcg receptor ameliorates development of non-alcoholic fatty liver disease. Cardiovasc. Hematol. Agents Med. Chem. 16, 35–43 (2018).

    Article  CAS  PubMed  Google Scholar 

  18. Patel, V. et al. Coagonist of Gcg-like peptide-1 and Gcg receptors ameliorates nonalcoholic fatty liver disease. Can. J. Physiol. Pharmacol. 96, 587–596 (2018).

    Article  CAS  PubMed  Google Scholar 

  19. Valdecantos, M. P. et al. A novel Gcg-like peptide 1/Gcg receptor dual agonist improves steatohepatitis and liver regeneration in mice. Hepatology 65, 950–968 (2017).

    Article  CAS  PubMed  Google Scholar 

  20. Hansen, H. H. et al. Mouse models of nonalcoholic steatohepatitis in preclinical drug development. Drug Discov. Today 22, 1707–1718 (2017).

    Article  CAS  PubMed  Google Scholar 

  21. Koliaki, C. et al. Adaptation of hepatic mitochondrial function in humans with non-alcoholic fatty liver is lost in steatohepatitis. Cell Metab. 21, 739–746 (2015).

    Article  CAS  PubMed  Google Scholar 

  22. Boland, M. L. et al. Nonalcoholic steatohepatitis severity is defined by a failure in compensatory antioxidant capacity in the setting of mitochondrial dysfunction. World J. Gastroenterol. 24, 1748–1765 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Shaw, R. J. et al. The kinase LKB1 mediates glucose homeostasis in liver and therapeutic effects of metformin. Science 310, 1642–1646 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lawitz, E. J. et al. Acetyl-CoA carboxylase inhibitor GS-0976 for 12 weeks reduces hepatic de novo lipogenesis and steatosis in patients with nonalcoholic steatohepatitis. Clin. Gastroenterol. Hepatol. 16, 1983–1991 (2018).

    Article  CAS  PubMed  Google Scholar 

  25. Koo, S.-H. et al. The CREB coactivator TORC2 is a key regulator of fasting glucose metabolism. Nature 437, 1109–1113 (2005).

    Article  CAS  PubMed  Google Scholar 

  26. Dulai, P. S. et al. Increased risk of mortality by fibrosis stage in nonalcoholic fatty liver disease: systematic review and meta-analysis. Hepatology 65, 1557–1565 (2017).

    Article  CAS  PubMed  Google Scholar 

  27. Armstrong, M. J. et al. Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): a multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet 387, 679–690 (2016).

    Article  CAS  PubMed  Google Scholar 

  28. Loomba, R. et al. GS-0976 reduces hepatic steatosis and fibrosis markers in patients with nonalcoholic fatty liver disease. Gastroenterology 155, 1463–1473 (2018).

    Article  CAS  PubMed  Google Scholar 

  29. Neuschwander-Tetri, B. A. et al. Farnesoid X nuclear receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic steatohepatitis (FLINT): a multicentre, randomised, placebo-controlled trial. Lancet 385, 956–965 (2015).

    Article  CAS  PubMed  Google Scholar 

  30. Ayala, J. E. et al. Hyperinsulinemic–euglycemic clamps in conscious, unrestrained mice. J. Vis. Exp. e3188 (2011).

  31. Steele, R., Wall, J. S., De Bodo, R. C. & Altszuler, N. Measurement of size and turnover rate of body glucose pool by the isotope dilution method. Am. J. Physiol. 187, 15–24 (1956).

    Article  CAS  PubMed  Google Scholar 

  32. Bederman, I. R., Foy, S., Chandramouli, V., Alexander, J. C. & Previs, S. F. Triglyceride synthesis in epididymal adipose tissue: contribution of glucose and non-glucose carbon sources. J. Biol. Chem. 284, 6101–6108 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Hasenour, C. M. et al. Mass spectrometry-based microassay of 2H and 13C plasma glucose labeling to quantify liver metabolic fluxes in vivo. Am. J. Physiol. Endocrinol. Metab. 309, E191–E203 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Young, J. D. INCA: a computational platform for isotopically non-stationary metabolic flux analysis. Bioinformatics 30, 1333–1335 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Hughey, C. C. et al. Loss of hepatic AMP-activated protein kinase impedes the rate of glycogenolysis but not gluconeogenic fluxes in exercising mice. J. Biol. Chem. 292, 20125–20140 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Chan, T. M. & Exton, J. H. A method for the determination of glycogen content and radioactivity in small quantities of tissues or isolated hepatocytes. Anal. Biochem. 71, 96–105 (1976).

    Article  CAS  PubMed  Google Scholar 

  37. Yang, D. et al. Assay of low deuterium enrichment of water by isotopic exchange with [U-13C3]acetone and gas chromatography-mass spectrometry. Anal. Biochem. 258, 315–321 (1998).

    Article  CAS  PubMed  Google Scholar 

  38. Previs, S. F. et al. Using [2H]water to quantify the contribution of de novo palmitate synthesis in plasma: enabling back-to-back studies. Am. J. Physiol. Endocrinol. Metab. 315, E63–e71 (2018).

    Article  CAS  PubMed  Google Scholar 

  39. Burgess, S. C. et al. Impaired tricarboxylic acid cycle activity in mouse livers lacking cytosolic phosphoenolpyruvate carboxykinase. J. Biol. Chem. 279, 48941–48949 (2004).

    Article  CAS  PubMed  Google Scholar 

  40. Antoniewicz, M. R., Kelleher, J. K. & Stephanopoulos, G. Measuring deuterium enrichment of glucose hydrogen atoms by gas chromatography/mass spectrometry. Anal. Chem. 83, 3211–3216 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Kristiansen, M. N. et al. Obese diet-induced mouse models of nonalcoholic steatohepatitis-tracking disease by liver biopsy. World J. Hepatol. 8, 673–684 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Barascuk, N. et al. A novel assay for extracellular matrix remodeling associated with liver fibrosis: an enzyme-linked immunosorbent assay (ELISA) for a MMP-9 proteolytically revealed neo-epitope of type III collagen. Clin. Biochem. 43, 899–904 (2010).

    Article  CAS  PubMed  Google Scholar 

  43. Leeming, D. J. et al. Enzyme-linked immunosorbent serum assay specific for the 7S domain of Collagen Type IV (P4NP 7S): a marker related to the extracellular matrix remodeling during liver fibrogenesis. Hepatol. Res. 42, 482–493 (2012).

    Article  CAS  PubMed  Google Scholar 

  44. Vassiliadis, E. et al. Immunological detection of the type V collagen propeptide fragment, PVCP-1230, in connective tissue remodeling associated with liver fibrosis. Biomarkers 16, 426–433 (2011).

    Article  CAS  PubMed  Google Scholar 

  45. Ouberai, M. M. et al. Controlling the bioactivity of a peptide hormone in vivo by reversible self-assembly. Nat. Commun. 8, 1026 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Kleiner, D. E. et al. Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology 41, 1313–1321 (2005).

    Article  PubMed  Google Scholar 

  47. Glick, D. et al. BNip3 regulates mitochondrial function and lipid metabolism in the liver. Mol. Cell Biol. 32, 2570–2584 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Michopoulos, F. et al. Targeted profiling of polar intracellular metabolites using ion-pair-high performance liquid chromatography and -ultra high performance liquid chromatography coupled to tandem mass spectrometry: applications to serum, urine and tissue extracts. J. Chromatogr. A. 1349, 60–68 (2014).

    Article  CAS  PubMed  Google Scholar 

  49. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Liberzon, A. et al. Molecular signatures database (MSigDB) 3.0. Bioinformatics 27, 1739–1740 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Liberzon, A. et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 1, 417–425 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Hanzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 14, 7 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Kang, T. et al. Characterization of the molecular mechanisms underlying glucose stimulated insulin secretion from isolated pancreatic beta-cells using post-translational modification specific proteomics (PTMomics). Mol. Cell Proteomics 17, 95–110 (2018).

    Article  CAS  PubMed  Google Scholar 

  55. The, N. et al. Fast and accurate proteim false discovery rates on large-scale proteomics data sets with percolator 3.0. J. Am. Soc. Mass Spectrom. 27, 1719–1727 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Käll, L. et al. Semi-supervised learning for peptide identification from shotgun proteomics datasets. Nat. Methods 4, 923–925 (2007).

    Article  PubMed  CAS  Google Scholar 

  57. Marx, H. et al. A large synthetic peptide and phosphopeptide reference library for mass spectrometry-based proteomics. Nat. Biotechnol. 13, 557–566 (2013).

    Article  CAS  Google Scholar 

  58. Navarro, P. et al. General statistical framework for quantitative proteomics by stabe isotope labeling. J. Proteome Res. 13, 1234–1247 (2014).

    Article  CAS  PubMed  Google Scholar 

  59. Zierer, J. et al. The fecal metabolome as a functional readout of the gut microbiome. Nat. Genet. 50, 790–795 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Zhang, M. et al. Adipocyte-derived lipids mediate melanoma progression via FATP proteins. Cancer Discov. 8, 1006–1025 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Lofgren, L. et al. The BUME method: a novel automated chloroform-free 96-well total lipid extraction method for blood plasma. J. Lipid. Res. 53, 1690–1700 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

The authors wish to thank the Lab Animal Resource staff at MedImmune/AstraZeneca for their assistance with animal husbandry and care. We thank D. Leeming and M. Karsdal (Nordic Biosciences, Denmark) for assistance with circulating collagen fragment detection and interpretation. We thank K. Hightower (Metabolon, Durham, NC) for assistance with metabolomics analyses and interpretation. The authors also wish to thank M. Jain (AstraZeneca, Cambridge, UK) for critical review and comments on the manuscript during preparation. The Villum Center for Bioanalytical Sciences at University of Southern Denmark is acknowledged for access to high-end MS instruments. Vanderbilt MMPC and their NIH funding (DK059637).

Author information

Authors and Affiliations

Authors

Contributions

M.L.B., R.C.L., A.N., M.F., M.R.L., L.L., O.P.M. and J.L.T. designed experiments; M.L.B., R.C.L., A.N., S.O., B.B.B., H.L., J.C., K.M., J.N., M.F., M.R.L., L.L., O.P.M., C.J.R. and J.L.T. collected and/or analysed and interpreted experimental data; S.G. and S.S.V. provided pathology analysis of mouse NASH studies; M.L.B, R.C.L., J.G., M.R.L, C.J.R. and J.L.T. wrote the paper; C.M.R. and L.J. reviewed and edited the manuscript.

Corresponding author

Correspondence to Christopher J. Rhodes.

Ethics declarations

Competing interests

The authors declare competing interests as defined by Nature Research. Employee of AstraZeneca (R.C.L., K.M., S.O., J.C., J.N., J.G., L.J., C.J.R.). Owns stock in AstraZeneca (K.M., S.O., J.C., J.N., J.G., L.J., C.M.R., J.L.T., C.J.R.).

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Metabolic and hepatic parameters following six-week treatment of NASH C57Bl6/J mice.

Mice were treated with cotadutide, liraglutide, g1437 or liraglutide+g1437 at equimolar dosing (10 nmol/kg, SC, QD for 42 days) compared to vehicle. Animals were given ad libitum access to food for the entirety of the study except on day 14 mice were fasted for 6 h prior to ipPTT. a, Reduction in body weight throughout the 42-day dosing period shown as % change. b, Blood glucose profile during ipPTT (pyruvate given at dose of 2g/kg) and (c) area under the ipPTT curve. d, Fasting plasma insulin, (e) fasting blood glucose and (f) plasma ALT levels at the end of the study. g, Terminal liver glycogen content, (h) triglycerides and (i) cholesterol. Chow vehicle (n=10); NASH vehicle (n=11); Cotadutide (n=12); Liraglutide (n=12); g1437 (n=12); Liraglutide+g1437 (n=12). Data shown as the mean ± SEM. (a and b) Two-way ANOVA, Tukey’s multiple comparisons post-hoc test. In (a and b) colored lines and p-values indicated differences for the corresponding treatment group compared with NASH vehicle at each time point. c-i, Two-sided student’s t-test for chow controls vs. NASH vehicle to determine effect of NASH diet; One-way ANOVA, Tukey’s multiple comparisons post-hoc test, chow group excluded.

Extended Data Fig. 2 Table of phosphopeptides.

Description of hepatic phosphopeptides detected in primary mouse and human hepatocytes following treatment with g1437.

Extended Data Fig. 3 Table of phosphopeptides.

Description of hepatic phosphopeptides detected in primary mouse and human hepatocytes following treatment with g1437.

Extended Data Fig. 4 Cotadutide improves mitochondrial respiratory function in primary mouse hepatocytes through Gcg signaling mechanisms.

a, Mitochondrial oxygen consumption rate (OCR) during mitochondrial stress test of healthy primary murine hepatocytes treated ex vivo for 4h with 100 nM cotadutide, g1437 or liraglutide compared to vehicle control hepatocytes (n=3 biologically independent samples/group). b, Basal respiration and (c) maximal respiration measured following injection of uncoupler FCCP. d, Oxygen consumption of primary mouse hepatocytes shown as the percentage of total respiration that is driven by the oxidation of indicated substrates (n=2 biologically independent samples/group). e-i, Oxygen consumption of mouse primary hepatocytes, during mitochondrial stress test, treated with cotadutide +/- Ampk inhibitor, Compound C (e; 20 uM), p38Mapk inhibitor, SB203580 (f; 20 um), mTor inhibitor, Rapamycin (g; 1 uM), Pi3 inhibitor, LY294 (h; 10 uM) or Mapkk inhibitor, PD98 (i; 20 uM) compared with control treated hepatocytes. n=6 replicates from 1 biological samples for all except for Cotadutide; Cotadutide+SB203580; Rapamycin; Cotadutide+Rapamycin; PD98; Cotadutide+PD98 in which 5 replicates were performed. The experiment was performed on 3 separate occasions with similar results. All data shown as the mean ± SEM. b-d, One-way ANOVA with Dunnett’s multiple comparisons post-hoc test.

Extended Data Fig. 5 Cotadutide reduces hepatic fibrosis and inflammation corresponding to animals shown in Figs. 7 and 8.

a, representative αSma stained liver sections, quantification is provided in Fig. 8d. Scale bar = 100 µm. b, representative PSR stained liver sections, quantification is provided in Fig. 8e. Scale bar = 100 µm. These experiments were performed in ob/ob mice with LFD (n=8); AMLN vehicle (n=11); Cotadutide (n=7); Liraglutide (n= 10); AMLN-to-LFD (n=10) mice/group.

Extended Data Fig. 6 Grades of histopathological NASH features corresponding to animals in Figs. 7 and 8.

a, steatosis grade, (b) lobular inflammation, (c) biliary hyperplasia and (d) hepatocyte ballooning. LFD (n=8); Vehicle (n=11); Cotadutide (n=7); Liraglutide (n=10); AMLN-to-LFD (n=10). Data represented as the mean ± SEM. Two-sided student’s t-test for LFD vs. vehicle to determine effect of NASH diet; one-way ANOVA, Tukey’s multiple comparison post-hoc test, LFD group excluded.

Extended Data Fig. 7 CD68, a marker of immune cell infiltration, is reduced by Cotadutide in animals corresponding to Figs. 7 and 8.

a, Representative images of CD68 IHC staining of mouse liver and the (b) pathologist graded scoring. LFD control slides are set to baseline of 1 to account for resident Kupffer cells (dark stained spots disperse equally between hepatocytes). Infiltration of immune cells identified by accumulation around vacuoles we scored relative to the baseline. Scale bar = 200 µm. LFD (n=8); Vehicle (n=11); Cotadutide (n=7); Liraglutide (n=10); AMLN-to-LFD (n=10). Data represented as the mean ± SEM. Two-sided student’s t-test for LFD vs. vehicle to determine effect of NASH diet; One-way ANOVA, Tukey’s multiple comparisons post-hoc test, LFD group excluded.

Supplementary information

Supplementary Information

Supplementary Figures 1–13, Tables 1–10 and References

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Boland, M.L., Laker, R.C., Mather, K. et al. Resolution of NASH and hepatic fibrosis by the GLP-1R and GCGR dual-agonist cotadutide via modulating mitochondrial function and lipogenesis. Nat Metab 2, 413–431 (2020). https://doi.org/10.1038/s42255-020-0209-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-020-0209-6

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing