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
. 2018 Sep 1;159(9):3132-3142.
doi: 10.1210/en.2018-00433.

α7 Nicotinic Acetylcholine Receptor Regulates the Function and Viability of L Cells

Affiliations

α7 Nicotinic Acetylcholine Receptor Regulates the Function and Viability of L Cells

Dawei Wang et al. Endocrinology. .

Abstract

Enteroendocrine L cells secrete the incretin hormone glucagon-like peptide-1 (GLP-1), and they also express the α7 nicotinic acetylcholine receptor (α7nAChR), which may regulate GLP-1 secretion. Here, GTS-21, a selective α7nAChR agonist, was used to examine the effect of α7nAChR activation in L-cell lines, mouse intestinal primary cell cultures, and C57BL/6 mice. GTS-21 stimulated GLP-1 secretion in vitro, and this effect was attenuated by an α7nAChR antagonist or by α7nAChR-specific small interfering RNA. Under in vitro cell culture conditions of glucotoxicity, GTS-21 restored GLP-1 secretion and improved L-cell viability while also acting in vivo to raise levels of circulating GLP-1 in mice. To assess potential signaling mechanisms underlying these actions of GTS-21, we first monitored Ca2+, cAMP, and phosphatidylinositol 3-kinase (PI3K) activity. As expected for a GLP-1 secretagogue promoting Ca2+ influx through α7nAChR cation channels, [Ca2+]i increased in response to GTS-21, but [cAMP]i was unchanged. Surprisingly, pharmacological inhibition of growth factor signaling pathways revealed that GTS-21 also acts on the PI3K-protein kinase B-mammalian target of rapamycin pathway to promote L-cell viability. Moreover, the Ca2+ chelator BAPTA-AM counteracted GTS-21‒stimulated PI3K activity, thereby indicating unexpected crosstalk of L-cell Ca2+ and growth factor signaling pathways. Collectively, these data demonstrate that α7nAChR activation enhances GLP-1 secretion by increasing levels of cytosolic Ca2+ while also revealing Ca2+- and PI3K-dependent processes of α7nAChR activation that promote L-cell survival.

PubMed Disclaimer

Figures

Figure 1.
Figure 1.
α7nAChR expression in intestinal L cells. (A) α7nAChR mRNA expression was analyzed by RT-PCR. A 199-bp band corresponding to mouse α7nAChR was specifically amplified in mouse brain tissue (positive control), GLUTag cells, and STC-1 cells. An 843-bp band corresponding to human α7nAChR was amplified in NCI-H716 cells. No contamination of genomic DNA was detected in the lane labeled “RT−,” which corresponds to PCR reactions run in the absence of reverse transcription. (B) α7nAChR protein expression was detected in mouse brain (positive control), GLUTag, STC-1, and NCI-H716 cells, as evaluated by Western blot analysis (20 μg of protein loaded per lane) using anti-α7nAChR antisera. RT−, reverse transcription‒negative.
Figure 2.
Figure 2.
Immunofluorescent staining of α7nAChR and GLP-1. Top two panels show α7nAChR (green) and GLP-1 (red) immunoreactivity in sections of mouse ileum and in primary intestinal (Intest.) cell cultures that contain L-cells (arrows). Merged overlays demonstrate codetection of both immunoreactivities in single L cells (yellow). Bottom two panels show the same as for top panels except that findings are for GLUTag and STC-1 cells. Calibration bars, 100 μm for ileum, GLUTag, and STC-1 or 10 μm for primary intestinal cell cultures.
Figure 3.
Figure 3.
Effects of GTS-21 and α-bungarotoxin (α-BgTX) on GLP-1 secretion. (A) GLUTag, STC-1, NCI-H716, and mouse primary intestinal cell cultures were treated with PBS (control) or increasing concentrations of GTS-21 for 2 h. Secreted GLP-1 was measured using ELISA assays and is expressed as the percentage change relative to control for each cell type. *P < 0.05, **P < 0.01, ***P < 0.001 vs. control group (n = 8 to 10). (B) GLUTag, STC-1, NCI-H716, and mouse primary intestinal cell cultures were preincubated with 100 nM of α-BgTX for 30 min before 2-h stimulation with GTS-21. **P < 0.01, ***P < 0.001 vs. control group; ##P < 0.01, ###P < 0.001 vs. GTS-21 group (n = 8 to 10). Box and whisker plots show the mean (solid box), 25% to 75% range (open box), median (horizontal line within open box), and minimum and maximum values (whiskers).
Figure 4.
Figure 4.
α7nAChR siRNA regulates α7nAChR expression and GLP-1 secretion. (A and B) Cells were transfected with (+) or without (−) α7nAChR (α7-si) or nontargeting [scrambled (Scr)] siRNA for 48 h. Levels of α7nAChR (A) mRNA and (B) protein (α7-R) were measured by RT-PCR and Western blot analysis. Cells transfected with α7nAChR or Scr siRNA (48 h) were treated with GTS-21 (75μM) for 2 h, and GLP-1 secretion was determined. (C and D) Knockdown of α7nAChR reduced GLP-1 secretion induced by GTS-21 in (C) GLUTag and (D) STC-1 cells. ***P < 0.001 vs. scrambled siRNA group; ###P < 0.001 vs. scrambled siRNA plus GTS-21 group (n = 7 to 8).
Figure 5.
Figure 5.
Effects of glucotoxicity (Glucotox.). (A) GLUTag cells were incubated for 72 h in either 5.6 mM (control) or 25.6 mM of glucose, washed and pretreated ± α-BgTX for 30 min, then stimulated with PBS or GTS-21 (75 μM) for 2 h. The amount of GLP-1 released into the medium was measured by ELISA. ***P < 0.001 vs. control group; ###P < 0.001 vs. PBS group; §§§P < 0.001 vs. GTS-21 group (n = 7). (B) GLUTag cells were preincubated with 100 nM of α-BgTX for 30 min and then were treated with PBS or GTS-21 (1 μM) for 24 h. Cell viability was determined by the MTT assay. ***P < 0.001 vs. PBS group; ###P < 0.001 vs. GTS-21 group (n = 8). (C) GLUTag cells were treated with 5.6 mM (control) or 30.6 mM (Glucotox.) of glucose for 72 h. GLUTag cells were then stimulated with GTS-21 (1 μM) for 24 h with (+) or without (−) preincubation using α-BgTX (100 nM). Cell viability was then measured by MTT assay. ***P < 0.001 vs. control group; ###P < 0.001 vs. PBS group; §§§P < 0.001 vs. 10-μM GTS-21 alone group (n = 7).
Figure 6.
Figure 6.
Assays for cytosolic Ca2+ and cAMP. (A) GLUTag cells transduced with the Ca2+ reporter YC3.6 were perifused with 5.6 mM of glucose standard extracellular solution at 37°C. GTS-21 (100 μM) was applied (indicated by bars) and induced an oscillatory rise of [Ca2+]i. Data are plotted as mean ± SEM for 9 cells from a single experiment and are representative of 64 cells from three independent transductions. (B) Single cell [Ca2+]i response to GTS-21 (100 μM) taken from the record shown in (A). The single-cell record highlights the oscillatory [Ca2+]i response. (C) GLUTag cells transduced with the cAMP reporter H188 were perifused with GTS-21 (100 μM) followed by forskolin (Fsk; 5 μM) + 3-isobutyl-1-methylxanthine (IBMX; 100 μM). GTS-21 had no effect on [cAMP]i, whereas the positive control (Fsk + IBMX) reversibly elevated [cAMP]. Data are plotted as mean ± SEM for 24 cells from three experiments and are representative of 62 cells from three independent viral transductions.
Figure 7.
Figure 7.
Regulation of GLP-1 secretion by the PI3K/AKT/mTOR signaling pathway. (A‒C) Stimulation of α7nAChR by GTS-21 enhanced GLP-1 secretion through PI3K/AKT/mTOR activation in GLUTag cells. (A) PI3K/AKT/mTOR activation. Cells were pretreated with α-BgTX (100 nM) for 30 min, then treated with GTS-21 (75 μM) for 2 h. The cell lysate was subjected to Western blotting. (B) Effects of GTS-21 on GLP-1 secretion were abrogated by the inhibitor of the PI3K/AKT/mTOR signaling pathway. Cells were pretreated with LY294002 (10 μM), Akt1/2 kinase inhibitor (5 μM), and rapamycin (2 nM) for 30 min, then treated with GTS-21 (75 μM) for 2 h. The amount of GLP-1 released into the medium was measured, and the cell lysate was subjected to Western blot analysis. (C) Effect of BAPTA-AM on GLP-1 secretion. Cells were pretreated with BAPTA-AM (10 µM) or PO4-AM3 (3.3 µM) for 30 min, then treated with GTS-21 (75 μM) for 2 h. Cell lysates were subjected to Western blotting. *P < 0.05, ***P < 0.001 vs. control group; #P < 0.05, ###P < 0.001 vs. GTS-21 group (n = 5 to 8).
Figure 8.
Figure 8.
Effects of GTS-21 on circulating GLP-1. (A) Mice were injected intraperitoneally with 2 to 8 mg/kg of GTS-21 or normal saline (vehicle). Blood samples were collected 1 h later, and GLP-1 levels were measured by ELISA. *P < 0.05, **P < 0.01 vs. control group (n = 4 to 5). (B) Mice were injected with 4 mg/kg of GTS-21, and circulating GLP-1 was measured at 1, 2, 4, and 8 h later by ELISA. *P < 0.05, **P < 0.01 vs. control group (n = 4 or 5).
Figure 9.
Figure 9.
A model for α7nAChR coupling to L-cell function, growth, and survival. Acetylcholine (ACh) or GTS-21 activates α7nAChR and promotes Ca2+ influx. Activation of the PI3K/Akt/mTOR pathway downstream of α7nAChR is involved in regulating GLP-1 secretion and cell viability, as revealed by the use of LY294002 (PI3K inhibitor), Akt1/2 kinase inhibitor, or rapamycin (mTOR inhibitor). Coupling of α7nAChR to PI3K activation can occur through several pathways including Ca2+-calmodulin (CaM) (41) and Janus kinase 2 (JAK2) (42). Ca2+ can also promote GLP-1 secretion directly or through Ca2+/calmodulin‒dependent kinase II (CaMKII) (43). The Ca2+ chelator BAPTA-AM inhibits all depicted effects of GST-21. Numerous downstream effectors of the metabolically regulated mTOR complex 1 (mTORC1) (44) are involved in cell viability, and determining which pathways are important to L-cell biology requires further investigation.

Similar articles

Cited by

References

    1. Habib AM, Richards P, Rogers GJ, Reimann F, Gribble FM. Co-localisation and secretion of glucagon-like peptide 1 and peptide YY from primary cultured human L cells. Diabetologia. 2013;56(6):1413–1416. - PMC - PubMed
    1. Vilsbøll T, Holst JJ. Incretins, insulin secretion and type 2 diabetes mellitus. Diabetologia. 2004;47(3):357–366. - PubMed
    1. Abbott CR, Monteiro M, Small CJ, Sajedi A, Smith KL, Parkinson JR, Ghatei MA, Bloom SR. The inhibitory effects of peripheral administration of peptide YY(3-36) and glucagon-like peptide-1 on food intake are attenuated by ablation of the vagal-brainstem-hypothalamic pathway. Brain Res. 2005;1044(1):127–131. - PubMed
    1. Krieger JP, Arnold M, Pettersen KG, Lossel P, Langhans W, Lee SJ. Knockdown of GLP-1 receptors in vagal afferents affects normal food intake and glycemia. Diabetes. 2016;65(1):34–43. - PubMed
    1. Courtney H, Nayar R, Rajeswaran C, Jandhyala R. Long-term management of type 2 diabetes with glucagon-like peptide-1 receptor agonists. Diabetes Metab Syndr Obes. 2017;10:79–87. - PMC - PubMed

Publication types

MeSH terms