Skip to main content
Top
Published in: Diabetologia 3/2018

01-03-2018 | Article

Evidence of a liver–alpha cell axis in humans: hepatic insulin resistance attenuates relationship between fasting plasma glucagon and glucagonotropic amino acids

Authors: Nicolai J. Wewer Albrechtsen, Kristine Færch, Troels M. Jensen, Daniel R. Witte, Jens Pedersen, Yuvaraj Mahendran, Anna E. Jonsson, Katrine D. Galsgaard, Marie Winther-Sørensen, Signe S. Torekov, Torsten Lauritzen, Oluf Pedersen, Filip K. Knop, Torben Hansen, Marit E. Jørgensen, Dorte Vistisen, Jens J. Holst

Published in: Diabetologia | Issue 3/2018

Login to get access

Abstract

Aims/hypothesis

The secretion of glucagon is controlled by blood glucose and inappropriate secretion of glucagon contributes to hyperglycaemia in diabetes. Besides its role in glucose regulation, glucagon regulates amino acid metabolism in hepatocytes by increasing ureagenesis. Disruption of this mechanism causes hyperaminoacidaemia, which in turn increases glucagon secretion. We hypothesised that hepatic insulin resistance (secondary to hepatic steatosis) via defective glucagon signalling/glucagon resistance would lead to impaired ureagenesis and, hence, increased plasma concentrations of glucagonotropic amino acids and, subsequently, glucagon.

Methods

To examine the association between glucagon and amino acids, and to explore whether this relationship was modified by hepatic insulin resistance, we studied a well-characterised cohort of 1408 individuals with normal and impaired glucose regulation. In this cohort, we have previously reported insulin resistance to be accompanied by increased plasma concentrations of glucagon. We now measure plasma levels of amino acids in the same cohort. HOMA-IR was calculated as a marker of hepatic insulin resistance.

Results

Fasting levels of glucagonotropic amino acids and glucagon were significantly and inversely associated in linear regression models (persisting after adjustment for age, sex and BMI). Increasing levels of hepatic, but not peripheral insulin resistance (p > 0.166) attenuated the association between glucagon and circulating levels of alanine, glutamine and tyrosine, and was significantly associated with hyperaminoacidaemia and hyperglucagonaemia. A doubling of the calculated glucagon–alanine index was significantly associated with a 30% increase in hepatic insulin resistance, a 7% increase in plasma alanine aminotransferase levels, and a 14% increase in plasma γ-glutamyltransferase levels.

Conclusions/interpretation

This cross-sectional study supports the existence of a liver–alpha cell axis in humans: glucagon regulates plasma levels of amino acids, which in turn feedback to regulate the secretion of glucagon. With hepatic insulin resistance, reflecting hepatic steatosis, the feedback cycle is disrupted, leading to hyperaminoacidaemia and hyperglucagonaemia. The glucagon–alanine index is suggested as a relevant marker for hepatic glucagon signalling.
Literature
1.
go back to reference Gromada J, Franklin I, Wollheim CB (2007) α-cells of the endocrine pancreas: 35 years of research but the enigma remains. Endocr Rev 28:84–116CrossRefPubMed Gromada J, Franklin I, Wollheim CB (2007) α-cells of the endocrine pancreas: 35 years of research but the enigma remains. Endocr Rev 28:84–116CrossRefPubMed
2.
go back to reference Kazda CM, Garhyan P, Kelly RP et al (2015) A randomized, double-blind, placebo-controlled phase 2 study of the glucagon receptor antagonist LY2409021 in patients with type 2 diabetes. Diabetes Care 37:1241–1249 Kazda CM, Garhyan P, Kelly RP et al (2015) A randomized, double-blind, placebo-controlled phase 2 study of the glucagon receptor antagonist LY2409021 in patients with type 2 diabetes. Diabetes Care 37:1241–1249
3.
go back to reference Petersen KF, Sullivan JT (2001) Effects of a novel glucagon receptor antagonist (Bay 27-9955) on glucagon-stimulated glucose production in humans. Diabetologia 44:2018–2024CrossRefPubMed Petersen KF, Sullivan JT (2001) Effects of a novel glucagon receptor antagonist (Bay 27-9955) on glucagon-stimulated glucose production in humans. Diabetologia 44:2018–2024CrossRefPubMed
4.
go back to reference Johnson DG, Goebel CU, Hruby VJ, Bregman MD, Trivedi D (1982) Hyperglycemia of diabetic rats decreased by a glucagon receptor antagonist. Science 215:1115–1116CrossRefPubMed Johnson DG, Goebel CU, Hruby VJ, Bregman MD, Trivedi D (1982) Hyperglycemia of diabetic rats decreased by a glucagon receptor antagonist. Science 215:1115–1116CrossRefPubMed
6.
go back to reference Assan R, Marre M, Gormley M (1983) The amino acid-induced secretion of glucagon. In: Lefebvre PJ (ed) Glucagon II. Spinger, Heidelberg, pp 19–41CrossRef Assan R, Marre M, Gormley M (1983) The amino acid-induced secretion of glucagon. In: Lefebvre PJ (ed) Glucagon II. Spinger, Heidelberg, pp 19–41CrossRef
7.
go back to reference Solloway Mark J, Madjidi A, Gu C et al (2015) Glucagon couples hepatic amino acid catabolism to mTOR-dependent regulation of α-cell mass. Cell Rep 12:495–510CrossRefPubMed Solloway Mark J, Madjidi A, Gu C et al (2015) Glucagon couples hepatic amino acid catabolism to mTOR-dependent regulation of α-cell mass. Cell Rep 12:495–510CrossRefPubMed
8.
go back to reference Holst JJ, Wewer Albrechtsen NJ, Pedersen J, Knop FK (2017) Glucagon and amino acids are linked in a mutual feedback cycle: the liver-alpha-cell axis. Diabetes 66:235–240CrossRefPubMed Holst JJ, Wewer Albrechtsen NJ, Pedersen J, Knop FK (2017) Glucagon and amino acids are linked in a mutual feedback cycle: the liver-alpha-cell axis. Diabetes 66:235–240CrossRefPubMed
9.
10.
go back to reference Larger E, Wewer Albrechtsen NJ, Hansen LH et al (2016) Pancreatic alpha-cell hyperplasia and hyperglucagonemia due to a glucagon receptor splice mutation. Endocrinol Diabetes Metab Case Rep 2016:16–0081PubMedPubMedCentral Larger E, Wewer Albrechtsen NJ, Hansen LH et al (2016) Pancreatic alpha-cell hyperplasia and hyperglucagonemia due to a glucagon receptor splice mutation. Endocrinol Diabetes Metab Case Rep 2016:16–0081PubMedPubMedCentral
11.
go back to reference Norton JA, Kahn CR, Schiebinger R, Gorschboth C, Brennan MF (1979) Amino acid deficiency and the skin rash associated with glucagonoma. Ann Intern Med 91:213–215CrossRefPubMed Norton JA, Kahn CR, Schiebinger R, Gorschboth C, Brennan MF (1979) Amino acid deficiency and the skin rash associated with glucagonoma. Ann Intern Med 91:213–215CrossRefPubMed
12.
go back to reference Alexander EK, Robinson M, Staniec M, Dluhy RG (2002) Peripheral amino acid and fatty acid infusion for the treatment of necrolytic migratory erythema in the glucagonoma syndrome. Clin Endocrinol 57:827–831CrossRef Alexander EK, Robinson M, Staniec M, Dluhy RG (2002) Peripheral amino acid and fatty acid infusion for the treatment of necrolytic migratory erythema in the glucagonoma syndrome. Clin Endocrinol 57:827–831CrossRef
13.
go back to reference Samuel VT, Liu ZX, Qu X et al (2004) Mechanism of hepatic insulin resistance in non-alcoholic fatty liver disease. J Biol Chem 279:32345–32353CrossRefPubMed Samuel VT, Liu ZX, Qu X et al (2004) Mechanism of hepatic insulin resistance in non-alcoholic fatty liver disease. J Biol Chem 279:32345–32353CrossRefPubMed
14.
go back to reference Isokuortti E, Zhou Y, Peltonen M et al (2017) Use of HOMA-IR to diagnose non-alcoholic fatty liver disease: a population-based and inter-laboratory study. Diabetologia 60:1873–1882CrossRefPubMed Isokuortti E, Zhou Y, Peltonen M et al (2017) Use of HOMA-IR to diagnose non-alcoholic fatty liver disease: a population-based and inter-laboratory study. Diabetologia 60:1873–1882CrossRefPubMed
15.
go back to reference Johansen NB, Hansen AL, Jensen TM et al (2012) Protocol for ADDITION-PRO: a longitudinal cohort study of the cardiovascular experience of individuals at high risk for diabetes recruited from Danish primary care. BMC Public Health 12:1078CrossRefPubMedPubMedCentral Johansen NB, Hansen AL, Jensen TM et al (2012) Protocol for ADDITION-PRO: a longitudinal cohort study of the cardiovascular experience of individuals at high risk for diabetes recruited from Danish primary care. BMC Public Health 12:1078CrossRefPubMedPubMedCentral
16.
go back to reference Lauritzen T, Griffin S, Borch-Johnsen K, Wareham NJ, Wolffenbuttel BH, Rutten G (2000) The ADDITION study: proposed trial of the cost-effectiveness of an intensive multifactorial intervention on morbidity and mortality among people with type 2 diabetes detected by screening. Int J Obes Relat Metab Disord 24(Suppl 3):S6–S11CrossRefPubMed Lauritzen T, Griffin S, Borch-Johnsen K, Wareham NJ, Wolffenbuttel BH, Rutten G (2000) The ADDITION study: proposed trial of the cost-effectiveness of an intensive multifactorial intervention on morbidity and mortality among people with type 2 diabetes detected by screening. Int J Obes Relat Metab Disord 24(Suppl 3):S6–S11CrossRefPubMed
17.
go back to reference Færch K, Torekov SS, Vistisen D et al (2015) Glucagon-like peptide-1 (GLP-1) response to oral glucose is reduced in pre-diabetes, screen-detected type 2 diabetes and obesity, and influenced by sex: the ADDITION-PRO study. Diabetes 64:2513–2525CrossRefPubMed Færch K, Torekov SS, Vistisen D et al (2015) Glucagon-like peptide-1 (GLP-1) response to oral glucose is reduced in pre-diabetes, screen-detected type 2 diabetes and obesity, and influenced by sex: the ADDITION-PRO study. Diabetes 64:2513–2525CrossRefPubMed
18.
19.
go back to reference Lund A, Bagger JI, Wewer Albrechtsen NJ et al (2015) Evidence of extrapancreatic glucagon secretion in man. Diabetes 65:585–597CrossRefPubMed Lund A, Bagger JI, Wewer Albrechtsen NJ et al (2015) Evidence of extrapancreatic glucagon secretion in man. Diabetes 65:585–597CrossRefPubMed
20.
go back to reference Soininen P, Kangas AJ, Wurtz P et al (2009) High-throughput serum NMR metabonomics for cost-effective holistic studies on systemic metabolism. Analyst 134:1781–1785CrossRefPubMed Soininen P, Kangas AJ, Wurtz P et al (2009) High-throughput serum NMR metabonomics for cost-effective holistic studies on systemic metabolism. Analyst 134:1781–1785CrossRefPubMed
21.
go back to reference Matthews DR, Hosker JP, Rudenski AS, Naylor BA, Treacher DF, Turner RC (1985) Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia 28:412–419CrossRefPubMed Matthews DR, Hosker JP, Rudenski AS, Naylor BA, Treacher DF, Turner RC (1985) Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia 28:412–419CrossRefPubMed
22.
go back to reference Abdul-Ghani MA, Matsuda M, Balas B, DeFronzo RA (2007) Muscle and liver insulin resistance indexes derived from the oral glucose tolerance test. Diabetes Care 30:89–94CrossRefPubMed Abdul-Ghani MA, Matsuda M, Balas B, DeFronzo RA (2007) Muscle and liver insulin resistance indexes derived from the oral glucose tolerance test. Diabetes Care 30:89–94CrossRefPubMed
23.
go back to reference Gutt M, Davis CL, Spitzer SB et al (2000) Validation of the insulin sensitivity index (ISI0,120): comparison with other measures. Diabetes Res Clin Pract 47:177–184CrossRefPubMed Gutt M, Davis CL, Spitzer SB et al (2000) Validation of the insulin sensitivity index (ISI0,120): comparison with other measures. Diabetes Res Clin Pract 47:177–184CrossRefPubMed
24.
go back to reference Yamada C, Kondo M, Kishimoto N et al (2015) Association between insulin resistance and plasma amino acid profile in non-diabetic Japanese subjects. J Diabetes Investig 6:408–415CrossRefPubMedPubMedCentral Yamada C, Kondo M, Kishimoto N et al (2015) Association between insulin resistance and plasma amino acid profile in non-diabetic Japanese subjects. J Diabetes Investig 6:408–415CrossRefPubMedPubMedCentral
25.
go back to reference Connelly MA, Wolak-Dinsmore J, Dullaart RPF (2017) Branched chain amino acids are associated with insulin resistance independent of leptin and adiponectin in subjects with varying degrees of glucose tolerance. Metab Syndr Relat Disord 15:183–186CrossRefPubMed Connelly MA, Wolak-Dinsmore J, Dullaart RPF (2017) Branched chain amino acids are associated with insulin resistance independent of leptin and adiponectin in subjects with varying degrees of glucose tolerance. Metab Syndr Relat Disord 15:183–186CrossRefPubMed
26.
go back to reference Mahendran Y, Jonsson A, Have CT et al (2017) Genetic evidence of a causal effect of insulin resistance on branched-chain amino acid levels. Diabetologia 60:873–878CrossRefPubMed Mahendran Y, Jonsson A, Have CT et al (2017) Genetic evidence of a causal effect of insulin resistance on branched-chain amino acid levels. Diabetologia 60:873–878CrossRefPubMed
30.
go back to reference Longuet C, Robledo AM, Dean ED et al (2013) Liver-specific disruption of the murine glucagon receptor produces α-cell hyperplasia: evidence for a circulating α-cell growth factor. Diabetes 62:1196–1120CrossRefPubMedPubMedCentral Longuet C, Robledo AM, Dean ED et al (2013) Liver-specific disruption of the murine glucagon receptor produces α-cell hyperplasia: evidence for a circulating α-cell growth factor. Diabetes 62:1196–1120CrossRefPubMedPubMedCentral
31.
32.
go back to reference Zhou C, Dhall D, Nissen NN, Chen C-R, Yu R (2009) A homozygous P86S mutation of the human glucagon receptor is associated with hyperglucagonemia, α cell hyperplasia, and islet cell tumor. Pancreas 38:941–946CrossRefPubMedPubMedCentral Zhou C, Dhall D, Nissen NN, Chen C-R, Yu R (2009) A homozygous P86S mutation of the human glucagon receptor is associated with hyperglucagonemia, α cell hyperplasia, and islet cell tumor. Pancreas 38:941–946CrossRefPubMedPubMedCentral
33.
go back to reference Miller HC, Kidd M, Modlin IM et al (2015) Glucagon receptor gene mutations with hyperglucagonemia but without the glucagonoma syndrome. World J Gastrointest Surg 7:60–66CrossRefPubMedPubMedCentral Miller HC, Kidd M, Modlin IM et al (2015) Glucagon receptor gene mutations with hyperglucagonemia but without the glucagonoma syndrome. World J Gastrointest Surg 7:60–66CrossRefPubMedPubMedCentral
34.
go back to reference Gelling RW, Du XQ, Dichmann DS et al (2003) Lower blood glucose, hyperglucagonemia, and pancreatic α cell hyperplasia in glucagon receptor knockout mice. Proc Natl Acad Sci U S A 100:1438–1443CrossRefPubMedPubMedCentral Gelling RW, Du XQ, Dichmann DS et al (2003) Lower blood glucose, hyperglucagonemia, and pancreatic α cell hyperplasia in glucagon receptor knockout mice. Proc Natl Acad Sci U S A 100:1438–1443CrossRefPubMedPubMedCentral
35.
go back to reference Junker AE, Gluud L, Holst JJ, Knop FK, Vilsboll T (2016) Diabetic and nondiabetic patients with nonalcoholic fatty liver disease have an impaired incretin effect and fasting hyperglucagonaemia. J Intern Med 279:485–493CrossRefPubMed Junker AE, Gluud L, Holst JJ, Knop FK, Vilsboll T (2016) Diabetic and nondiabetic patients with nonalcoholic fatty liver disease have an impaired incretin effect and fasting hyperglucagonaemia. J Intern Med 279:485–493CrossRefPubMed
36.
go back to reference Wewer Albrechtsen NJ, Junker AE, Christensen M et al (2017) Hyperglucagonemia correlates with plasma levels of non-branched chained amino acids in patients with liver disease independent of type 2 diabetes. Am J Physiol Gastrointest Liver Physiol. https://doi.org/10.1152/ajpgi.00216.2017 Wewer Albrechtsen NJ, Junker AE, Christensen M et al (2017) Hyperglucagonemia correlates with plasma levels of non-branched chained amino acids in patients with liver disease independent of type 2 diabetes. Am J Physiol Gastrointest Liver Physiol. https://​doi.​org/​10.​1152/​ajpgi.​00216.​2017
37.
go back to reference Khan K, Elia M (1991) Factors affecting the stability of L-glutamine in solution. Clin Nutr 10:186–192CrossRefPubMed Khan K, Elia M (1991) Factors affecting the stability of L-glutamine in solution. Clin Nutr 10:186–192CrossRefPubMed
39.
go back to reference Dean ED, Li M, Prasad N et al (2017) Interrupted glucagon signaling reveals hepatic alpha cell axis and role for L-glutamine in α cell proliferation. Cell Metab 25:1362–1373CrossRefPubMed Dean ED, Li M, Prasad N et al (2017) Interrupted glucagon signaling reveals hepatic alpha cell axis and role for L-glutamine in α cell proliferation. Cell Metab 25:1362–1373CrossRefPubMed
40.
go back to reference Kim J, Okamoto H, Huang Z et al (2017) Amino acid transporter Slc38a5 controls glucagon receptor inhibition-induced pancreatic α cell hyperplasia in mice. Cell Metab 25:1348–1361CrossRefPubMed Kim J, Okamoto H, Huang Z et al (2017) Amino acid transporter Slc38a5 controls glucagon receptor inhibition-induced pancreatic α cell hyperplasia in mice. Cell Metab 25:1348–1361CrossRefPubMed
41.
42.
go back to reference Ahren B (2006) Glucagon secretion in relation to insulin sensitivity in healthy subjects. Diabetologia 49:117–122CrossRefPubMed Ahren B (2006) Glucagon secretion in relation to insulin sensitivity in healthy subjects. Diabetologia 49:117–122CrossRefPubMed
43.
go back to reference Newgard CB, An J, Bain JR et al (2009) A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab 9:311–326CrossRefPubMedPubMedCentral Newgard CB, An J, Bain JR et al (2009) A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab 9:311–326CrossRefPubMedPubMedCentral
44.
go back to reference Holst JJ, Holland W, Gromada J et al (2017) Insulin and glucagon: partners for life. Endocrinology 158:696–701CrossRefPubMed Holst JJ, Holland W, Gromada J et al (2017) Insulin and glucagon: partners for life. Endocrinology 158:696–701CrossRefPubMed
45.
go back to reference Faerch K, Vistisen D, Pacini G et al (2016) Insulin resistance is accompanied by increased fasting glucagon and delayed glucagon suppression in individuals with normal and impaired glucose regulation. Diabetes 65:3473–3481CrossRefPubMed Faerch K, Vistisen D, Pacini G et al (2016) Insulin resistance is accompanied by increased fasting glucagon and delayed glucagon suppression in individuals with normal and impaired glucose regulation. Diabetes 65:3473–3481CrossRefPubMed
47.
go back to reference Dixon JB, Bhathal PS, O'Brien PE (2006) Weight loss and non-alcoholic fatty liver disease: falls in gamma-glutamyl transferase concentrations are associated with histologic improvement. Obes Surg 16:1278–1286CrossRefPubMed Dixon JB, Bhathal PS, O'Brien PE (2006) Weight loss and non-alcoholic fatty liver disease: falls in gamma-glutamyl transferase concentrations are associated with histologic improvement. Obes Surg 16:1278–1286CrossRefPubMed
48.
go back to reference Wewer Albrechtsen N, Hartmann B, Veedfald S et al (2014) Hyperglucagonaemia analysed by glucagon sandwich ELISA: nonspecific interference or truly elevated levels? Diabetologia 57:1919–1926CrossRefPubMed Wewer Albrechtsen N, Hartmann B, Veedfald S et al (2014) Hyperglucagonaemia analysed by glucagon sandwich ELISA: nonspecific interference or truly elevated levels? Diabetologia 57:1919–1926CrossRefPubMed
49.
go back to reference Guzman CB, Zhang XM, Liu R et al (2017) Treatment with LY2409021, a glucagon receptor antagonist, increases liver fat in patients with type 2 diabetes. Diabetes Obes Metab 19:1521–1528CrossRefPubMed Guzman CB, Zhang XM, Liu R et al (2017) Treatment with LY2409021, a glucagon receptor antagonist, increases liver fat in patients with type 2 diabetes. Diabetes Obes Metab 19:1521–1528CrossRefPubMed
Metadata
Title
Evidence of a liver–alpha cell axis in humans: hepatic insulin resistance attenuates relationship between fasting plasma glucagon and glucagonotropic amino acids
Authors
Nicolai J. Wewer Albrechtsen
Kristine Færch
Troels M. Jensen
Daniel R. Witte
Jens Pedersen
Yuvaraj Mahendran
Anna E. Jonsson
Katrine D. Galsgaard
Marie Winther-Sørensen
Signe S. Torekov
Torsten Lauritzen
Oluf Pedersen
Filip K. Knop
Torben Hansen
Marit E. Jørgensen
Dorte Vistisen
Jens J. Holst
Publication date
01-03-2018
Publisher
Springer Berlin Heidelberg
Published in
Diabetologia / Issue 3/2018
Print ISSN: 0012-186X
Electronic ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-017-4535-5

Other articles of this Issue 3/2018

Diabetologia 3/2018 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.