Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2008 Sep 2;105(35):13163-8.
doi: 10.1073/pnas.0801059105. Epub 2008 Aug 21.

Interleukin-6 regulates pancreatic alpha-cell mass expansion

Affiliations

Interleukin-6 regulates pancreatic alpha-cell mass expansion

Helga Ellingsgaard et al. Proc Natl Acad Sci U S A. .

Abstract

Interleukin-6 (IL-6) is systemically elevated in obesity and is a predictive factor to develop type 2 diabetes. Pancreatic islet pathology in type 2 diabetes is characterized by reduced beta-cell function and mass, an increased proportion of alpha-cells relative to beta-cells, and alpha-cell dysfunction. Here we show that the alpha cell is a primary target of IL-6 actions. Beginning with investigating the tissue-specific expression pattern of the IL-6 receptor (IL-6R) in both mice and rats, we find the highest expression of the IL-6R in the endocrine pancreas, with highest expression on the alpha-cell. The islet IL-6R is functional, and IL-6 acutely regulates both pro-glucagon mRNA and glucagon secretion in mouse and human islets, with no acute effect on insulin secretion. Furthermore, IL-6 stimulates alpha-cell proliferation, prevents apoptosis due to metabolic stress, and regulates alpha-cell mass in vivo. Using IL-6 KO mice fed a high-fat diet, we find that IL-6 is necessary for high-fat diet-induced increased alpha-cell mass, an effect that occurs early in response to diet change. Further, after high-fat diet feeding, IL-6 KO mice without expansion of alpha-cell mass display decreased fasting glucagon levels. However, despite these alpha-cell effects, high-fat feeding of IL-6 KO mice results in increased fed glycemia due to impaired insulin secretion, with unchanged insulin sensitivity and similar body weights. Thus, we conclude that IL-6 is necessary for the expansion of pancreatic alpha-cell mass in response to high-fat diet feeding, and we suggest that this expansion may be needed for functional beta-cell compensation to increased metabolic demand.

PubMed Disclaimer

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Fig. 1.
Fig. 1.
IL-6R is expressed in the pancreatic α-cell and is functionally coupled to STAT3 phosphorylation. (A and B) Tissue expression profile of mouse and rat IL-6R mRNA expression determined by Affymetrix gene array (n = 3–5). (C and D) Quantitative RT-PCR on RNA from FACS sorted rat α-cells and β-cells (purity ≈90% as assessed by insulin and glucagon staining respectively) normalized for 18S (n = 3). (E) Western blot analysis of the IL-6R in HeLa cell extracts (+ control), purified rat α-cells and β-cells, and whole mouse islets (representative of n = 3). (F) Western blot of pSTAT3 and total STAT3 in mouse and human islets after 15 min. exposure to 100 ng/ml IL-6 (representative of n = 3). *, P < 0.05.
Fig. 2.
Fig. 2.
Interleukin-6 regulates pro-glucagon mRNA and glucagon secretion with no effect on insulin mRNA and release. (A and C) Pro-glucagon and insulin mRNA in human islets after exposure to 200 ng/ml IL-6 (n = 3–4). (B and D) Glucagon and insulin release in culture medium of human islets after exposure to 200 ng/ml IL-6. (E) Glucagon secretion from human islets during 1 h static incubation in the presence of 20 mM glucose (white bars), 2 mM glucose (hatched bars), and 10 mM Arginine (black bars). Islets were pretreated with 200 ng/ml IL-6 for the indicated times (n = 4). (F) Circulating glucagon levels 2 h after 100 ng bolus IL-6 injection in mice during fed and fasted state (n = 3–5). All secretion experiments were performed on 20 islets per well in triplicate with the number of independent experiments indicated above. *, P < 0.05 vs. respective controls.
Fig. 3.
Fig. 3.
Interleukin-6 increases pancreatic α-cell proliferation and prevents α-cell apoptosis in vitro. (A) Ki67-positive human islet-cells per islet after 4 days' treatment in the absence (Ctrl) and presence of 200 ng/ml IL-6 (n = 3–5). (B) Ki67-positive human islet-cells per islet after 4 days treatment with the IL-6R antagonist, Sant7 (200 ng/ml; n = 3). (C and D) Percent BrdU-positive mouse α- and β-cells (glucagon and insulin positive, respectively) of total number of cells. Cells were treated in the absence (Ctrl) and presence of 100 ng/ml IL-6 for 24 and 96 h with BrdU present during the entire experiment (n = 3). (E and F) Percent TUNEL-positive mouse α-cells and β-cells after 12 h treatment with 33.3 mM glucose and 0.5 mM palmitate (gluc + palm), in the absence (Ctrl) and presence of 100 ng/ml IL-6 (n = 3). (G) Representative image of mouse islets on extracellular matrix coated dishes stained for BrdU after 4 days in the absence (control) and presence of 100 ng/ml IL-6, with BrdU present during the entire experiment. *, P < 0.05.
Fig. 4.
Fig. 4.
Impaired glucose tolerance in IL-6 KO mice after 18 weeks on HF diet. (A) Body weight, (G) ipGTT, (H) glucose-stimulated insulin secretion, and (I) ipITT in WT (solid line, open squares) and IL-6 KO (dashed line, closed circles) mice fed an HF diet for 18 weeks (n = 8 WT, n = 9 IL-6 KO). (B) Fasting blood glucose, (C) fasting plasma glucagon, (D) fasting plasma insulin, (E) glucagon during ipGTT, (F) fed blood glucose, (J) HOMA-IR, (K) a-cell mass, and (L) b-cell mass in WT (white bars) and IL-6 KO (black bars) mice after 18 weeks on HF diet (chow WT n = 5, chow IL-6 KO n = 8, HF WT n = 8, HF IL-6 KO n = 9) *, P < 0.05.

Similar articles

Cited by

References

    1. Unger RH, Orci L. The essential role of glucagon in the pathogenesis of diabetes mellitus. Lancet. 1975;1:14–16. - PubMed
    1. Deng S, et al. Structural and functional abnormalities in the islets isolated from type 2 diabetic subjects. Diabetes. 2004;53:624–632. - PubMed
    1. Yoon KH, et al. Selective beta-cell loss and alpha-cell expansion in patients with type 2 diabetes mellitus in Korea. J Clin Endocrinol Metab. 2003;88:2300–2308. - PubMed
    1. Donath MY, et al. Mechanisms of beta-cell death in type 2 diabetes. Diabetes. 2005;54(Suppl 2):S108–13. - PubMed
    1. Rahier J, Goebbels RM, Henquin JC. Cellular composition of the human diabetic pancreas. Diabetologia. 1983;24:366–371. - PubMed

Publication types

MeSH terms