GABA coordinates with insulin in regulating secretory function in pancreatic INS-1 β-cells

Paul Bansal, Shuanglian Wang, Shenghao Liu, Yun-Yan Xiang, Wei-Yang Lu, Qinghua Wang, Paul Bansal, Shuanglian Wang, Shenghao Liu, Yun-Yan Xiang, Wei-Yang Lu, Qinghua Wang

Abstract

Pancreatic islet β-cells produce large amounts of γ-aminobutyric acid (GABA), which is co-released with insulin. GABA inhibits glucagon secretion by hyperpolarizing α-cells via type-A GABA receptors (GABA(A)Rs). We and others recently reported that islet β-cells also express GABA(A)Rs and that activation of GABA(A)Rs increases insulin release. Here we investigate the effects of insulin on the GABA-GABA(A)R system in the pancreatic INS-1 cells using perforated-patch recording. The results showed that GABA produces a rapid inward current and depolarizes INS-1 cells. However, pre-treatment of the cell with regular insulin (1 µM) suppressed the GABA-induced current (I(GABA)) by 43%. Zinc-free insulin also suppressed I(GABA) to the same extent of inhibition by regular insulin. The inhibition of I(GABA) occurs within 30 seconds after application of insulin. The insulin-induced inhibition of I(GABA) persisted in the presence of PI3-kinase inhibitor, but was abolished upon inhibition of ERK, indicating that insulin suppresses GABA(A)Rs through a mechanism that involves ERK activation. Radioimmunoassay revealed that the secretion of C-peptide was enhanced by GABA, which was blocked by pre-incubating the cells with picrotoxin (50 µM, p<0.01) and insulin (1 µM, p<0.01), respectively. Together, these data suggest that autocrine GABA, via activation of GABA(A)Rs, depolarizes the pancreatic β-cells and enhances insulin secretion. On the other hand, insulin down-regulates GABA-GABA(A)R signaling presenting a feedback mechanism for fine-tuning β-cell secretion.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. GABA depolarizes membrane potential and…
Figure 1. GABA depolarizes membrane potential and increases intracellular Ca2+ in INS-1 cells.
(A) Perfusion of ECS containing 28 mM glucose induces a gradual and sustained depolarization of the membrane potential (Vm) (n = 5). (B) GABA induces a rapid and GABAAR inhibition-sensitive depolarization of Vm under the current-clamp conditions at 1.4 mM glucose (n = 5). (C) Cells cultured in 96-well plates pre-loaded with Fluo-3 AM were treated with GABA (30 µM), or 5 mM KCl as positive control. Changes in relative fluorescence units (RFU) were monitored with a fluorescent plate reader. Data are Mean±SE, n = 6.
Figure 2. GABA-evoked currents (I GABA )…
Figure 2. GABA-evoked currents (IGABA) is inhibited by insulin in INS-1 cells.
GABA-evoked inward current was measured by means of a computer-controlled multi-barrelled perfusion system, in two-minute intervals, under voltage-clamp conditions. Representative traces of GABA-evoked currents in the absence and presence of insulin (100 nM, (A), 1 µM, (B)) in the same INS-1 cell. A' and B' represents the average of IGABA from separated experiments. (C) Normalized average IGABA during the course of experiment (control = average of first 4 IGABA, insulin = average of IGABA in the presence insulin at indicated concentrations). Data were mean ± SE. *p<0.05 ** p<0.01, n = 6.
Figure 3. Zinc-free insulin inhibits GABA-evoked currents…
Figure 3. Zinc-free insulin inhibits GABA-evoked currents in INS-1 cells.
(A) Representative traces of GABA-evoked currents in the absence and presence of zinc-free insulin and regular insulin in the same INS-1 cell. (B) The average of IGABA from separated experiments. (C) Normalized average IGABA was separately recorded during the course of experiment (control = average of first 4 IGABA, ZFI = average of IGABA in the presence zinc-free insulin, insulin = average of IGABA in the presence of insulin after washing out). (D) Representative traces of IGABA obtained from when GABA was applied simultaneously with insulin or 30 seconds after insulin pre-treatment. (E) Normalized average IGABA of separated experiments as described in (C). Data were mean ± SE. *p<0.05, n = 5.
Figure 4. Insulin-induced inhibition of I GABA…
Figure 4. Insulin-induced inhibition of IGABA in INS-1 cells is PI3-K/Akt independent.
(A) Representative traces of GABA-evoked currents in the absence and presence of zinc-free insulin (1 µM) along with PI3-K inhibitor wortmannin (100 nM). (B) Normalized average of IGABA from separated experiments. (C) Akt activity determined by Western Blotting using anti-phospho Akt (S473) in cells treated without or with wortmannin (Wort), or in the cells transfected with dominant-negative Akt (DN-Akt). (D) Representative traces of GABA-evoked currents in cells expressing DN-Akt in the absence and presence of zinc-free insulin (ZFI,1 µM). (E) Average IGABA from separated time-course experiments. Data were mean ± SE. *p<0.05, ** p<0.01, n = 5.
Figure 5. Insulin suppresses I GABA which…
Figure 5. Insulin suppresses IGABA which is not associated with GABAAR membrane relocalization and is ERK-dependent.
(A) Confocal microscopic image of INS-1 cells immunostained for GABAARs using anti-GABAAR β2/3 mouse IgG and Cy3-conjugated secondary antibody (red) with DAPI-nuclear staining (blue). Cells were treated with or without insulin, in the presence or absence of PI3-K inhibitor wortmannin. (B) Insulin (100 nM, 5 min) stimulated ERK phosphorylation in INS-1 cells, which was blocked by pre-treatment of the cells with PD98059 (20 µM, 10 min). (C) Representative traces of GABA-evoked currents in the absence and presence of zinc-free insulin (0.6 µM) with or without PD98059 (20 µM). (D) Normalized average of IGABA from separated experiments. Data were mean ± SE. *p<0.05, ** p<0.01, n = 5–6.
Figure 6. GABA enhances insulin secretion which…
Figure 6. GABA enhances insulin secretion which is attenuated by insulin in INS-1 cells.
Insulin secretion was evaluated by C-peptide RIA. Cells were serum-starved in KRB buffer containing 1.4 mM glucose for 60 min prior to the RIA. The RIA was conducted using cells which had their culture medium replaced with fresh KRB buffer containing 1.4 mM glucose (or 11.1 mM glucose as positive control, PC), in the presence of GABA (30 µM), with or without either picrotoxin (Pic, 50 µM) or insulin (Ins, 1 µM) for 120 min. Data were mean ± SE, from three independent experiments with each sample counted in triplication. ** p

References

    1. Luscher B, Keller CA. Regulation of GABAA receptor trafficking, channel activity, and functional plasticity of inhibitory synapses. Pharmacol Ther. 2004;102:195–221.
    1. Okada Y, Taniguchi H, Shimada C. High Concentration of GABA and High Glutamate Decarboxylase Activity in Rat Pancreatic Islets and Human Insulinoma. Science. 1976;194:620–622.
    1. Gerber JC, III, Hare TA. Gamma-aminobutyric acid in peripheral tissue, with emphasis on the endocrine pancreas: presence in two species and reduction by streptozotocin. Diabetes. 1979;28:1073–1076.
    1. Vincent SR, Hokfelt T, Wu JY, Elde RP, Morgan LM, et al. Immunohistochemical studies of the GABA system in the pancreas. Neuroendocrinology. 1983;36:197–204.
    1. Reetz A, Solimena M, Matteoli M, Folli F, Takei K, et al. GABA and pancreatic beta-cells: colocalization of glutamic acid decarboxylase (GAD) and GABA with synaptic-like microvesicles suggests their role in GABA storage and secretion. EMBO J. 1991;10:1275–1284.
    1. Braun M, Ramracheya R, Bengtsson M, Clark A, Walker JN, et al. Gamma-Aminobutyric Acid (GABA) Is an Autocrine Excitatory Transmitter in Human Pancreatic Beta-Cells. Diabetes. 2010;59:1694–1701.
    1. Rorsman P, Berggren PO, Bokvist K, Ericson H, Mohler H, et al. Glucose-inhibition of glucagon secretion involves activation of GABAA-receptor chloride channels. Nature. 1989;341:233–236.
    1. Smismans A, Schuit F, Pipeleers D. Nutrient regulation of gamma-aminobutyric acid release from islet beta cells. Diabetologia. 1997;40:1411–1415. 10.1007/s001250050843.
    1. Winnock F, Ling Z, De Proft R, Dejonghe S, Schuit F, et al. Correlation between GABA release from rat islet beta -cells and their metabolic state. Am J Physiol Endocrinol Metab. 2002;282:E937–E942.
    1. Wendt A, Birnir B, Buschard K, Gromada J, Salehi A, et al. Glucose Inhibition of Glucagon Secretion From Rat [alpha]-Cells Is Mediated by GABA Released From Neighboring [beta]-Cells. Diabetes. 2004;53:1038–1045.
    1. Bailey SJ, Ravier MA, Rutter GA. Glucose-dependent regulation of gamma-aminobutyric acid (GABA A) receptor expression in mouse pancreatic islet alpha-cells. Diabetes. 2007;56:320–327.
    1. Xu E, Kumar M, Zhang Y, Ju W, Obata T, et al. Intra-islet insulin suppresses glucagon release via GABA-GABAA receptor system. Cell Metab. 2006;3:47–58.
    1. Glassmeier G, Hopfner M, Buhr H, Lemmer K, Riecken EO, et al. Expression of functional GABAA receptors in isolated human insulinoma cells. Ann N Y Acad Sci. 1998;859:241–248.
    1. Blankenfeld G, Enkvist MOK, Kettenmann H, Turner J, Ahnert-Hilger G, et al. Expression of functional GABAA receptors in neuroendocrine gastropancreatic cells. Pflugers Archiv European Journal of Physiology. 1995;430:381–388. 10.1007/BF00373913.
    1. Dong H, Kumar M, Zhang Y, Gyulkhandanyan A, Xiang YY, et al. Gamma-aminobutyric acid up- and downregulates insulin secretion from beta cells in concert with changes in glucose concentration. Diabetologia. 2006;49:697–705.
    1. Leibiger IB, Leibiger B, Berggren PO. Insulin Signaling in the Pancreatic [beta]-Cell. Annual Review of Nutrition. 2008;28:233–251. doi: .
    1. Wang Q, Jin T. The role of insulin signaling in the development of beta-cell dysfunction and diabetes. Islets. 2009;1:95–101.
    1. Soltani N, Qiu H, Aleksic M, Glinka Y, Zhao F, et al. GABA exerts protective and regenerative effects on islet beta cells and reverses diabetes. Proc Natl Acad Sci U S A. 2011;108:11692–11697.
    1. Sugita M, Hirono C, Shiba Y. Gramicidin-perforated Patch Recording Revealed the Oscillatory Nature of Secretory Cl- Movements in Salivary Acinar Cells. The Journal of General Physiology. 2004;124:59–69.
    1. Li J, Zhang N, Ye B, Ju W, Orser B, et al. Non-steroidal anti-inflammatory drugs increase insulin release from beta cells by inhibiting ATP-sensitive potassium channels. Br J Pharmacol. 2007;151:483–493. 0707259 [pii];10.1038/sj.bjp.0707259 [doi]
    1. Smith GD, Swenson DC, Dodson EJ, Dodson GG, Reynolds CD. Structural stability in the 4-zinc human insulin hexamer. Proc Natl Acad Sci U S A. 1984;81:7093–7097.
    1. Aguayo LG, Alarcon JM. Modulation of the developing rat sympathetic GABAA receptor by Zn++, benzodiazepines, barbiturates and ethanol. Journal of Pharmacology and Experimental Therapeutics. 1993;267:1414–1422.
    1. Hosie AM, Dunne EL, Harvey RJ, Smart TG. Zinc-mediated inhibition of GABAA receptors: discrete binding sites underlie subtype specificity. Nat Neurosci. 2003;6:362–369. 10.1038/nn1030.
    1. Taniguchi CM, Emanuelli B, Kahn CR. Critical nodes in signalling pathways: insights into insulin action. Nat Rev Mol Cell Biol. 2006;7:85–96. 10.1038/nrm1837.
    1. Wang Q, Liu L, Pei L, Ju W, Ahmadian G, et al. Neuron. 38; 2003. Control of Synaptic Strength, a Novel Function of Akt. pp. 915–928. doi: .
    1. Assmann A, Ueki K, Winnay JN, Kadowaki T, Kulkarni RN. Glucose effects on beta-cell growth and survival require activation of insulin receptors and insulin receptor substrate 2. Mol Cell Biol. 2009;29:3219–3228.
    1. Adeghate E, Ponery AS. GABA in the endocrine pancreas: cellular localization and function in normal and diabetic rats. Tissue and Cell. 2002;34:1–6. doi: .
    1. Ligon B, Yang J, Morin SB, Ruberti MF, Steer ML. Regulation of pancreatic islet cell survival and replication by gamma-aminobutyric acid. Diabetologia. 2007;50:764–773.
    1. Bormann J. Electrophysiology of GABAA and GABAB receptor subtypes. Trends in Neurosciences. 1988;11:112–116. doi: .
    1. Chen G, Trombley PQ, van den Pol AN. Excitatory actions of GABA in developing rat hypothalamic neurones. The Journal of Physiology. 1996;494:451–464.
    1. Owens DF, Kriegstein AR. Is there more to gaba than synaptic inhibition? Nat Rev Neurosci. 2002;3:715–727. 10.1038/nrn919.
    1. Ludwig A, Li H, Saarma M, Kaila K, Rivera C. Developmental up-regulation of KCC2 in the absence of GABAergic and glutamatergic transmission. Eur J Neurosci. 2003;18 3199-3206. 3069 [pii]
    1. Davies SL, Roussa E, Le Rouzic P, Thevenod F, Alper SL, et al. Expression of K+-Cl- cotransporters in the [alpha]-cells of rat endocrine pancreas. Biochimica et Biophysica Acta (BBA) - Biomembranes. 2004;1667:7–14. doi: .
    1. Takayama C, Inoue Y. Developmental localization of potassium chloride co-transporter 2 (KCC2), GABA and vesicular GABA transporter (VGAT) in the postnatal mouse somatosensory cortex. Neurosci Res. 2010;67:137–148.
    1. Chausmer AB. Zinc, Insulin and Diabetes. J Am Coll Nutr. 1998;17:109–115.
    1. Williams DB. A novel, rapid, inhibitory effect of insulin on [alpha]1[beta]2[gamma]2s [gamma]-aminobutyric acid type A receptors. Neuroscience Letters. 2008;443:27–31. doi: .
    1. Bell-Horner CL, Dohi A, Nguyen Q, Dillon GH, Singh M. ERK/MAPK pathway regulates GABAA receptors. J Neurobiol. 2006;66:1467–1474.
    1. Liu F, Wan Q, Pristupa ZB, Yu XM, Wang YT, et al. Direct protein-protein coupling enables cross-talk between dopamine D5 and [gamma]-aminobutyric acid A receptors. Nature. 2000;403:274–280. 10.1038/35002014.
    1. Persaud SJ, Asare-Anane H, Jones PM. Insulin receptor activation inhibits insulin secretion from human islets of Langerhans. FEBS Letters. 2002;510:225–228. doi: .
    1. Collier JJ, White SM, Dick GM, Scott DK. Phosphatidylinositol 3-kinase inhibitors reveal a unique mechanism of enhancing insulin secretion in 832/13 rat insulinoma cells. Biochem Biophys Res Commun. 2004;324:1018–1023.
    1. Eto K, Yamashita T, Tsubamoto Y, Terauchi Y, Hirose K, et al. Phosphatidylinositol 3-kinase suppresses glucose-stimulated insulin secretion by affecting post-cytosolic [Ca(2+)] elevation signals. Diabetes. 2002;51:87–97.
    1. Khan FA, Goforth PB, Zhang M, Satin LS. Insulin Activates ATP-Sensitive K+ Channels in Pancreatic [beta]-Cells Through a Phosphatidylinositol 3-Kinase–Dependent Pathway. Diabetes. 2001;50:2192–2198.
    1. Jimenez-Feltstrom J, Lundquist I, Obermuller S, Salehi A. Insulin feedback actions: complex effects involving isoforms of islet nitric oxide synthase. Regul Pept. 2004;122:109–118.
    1. Aspinwall CA, Lakey JRT, Kennedy RT. Insulin-stimulated Insulin Secretion in Single Pancreatic Beta Cells. J Biol Chem. 1999;274:6360–6365.
    1. Aspinwall CA, Qian WJ, Roper MG, Kulkarni RN, Kahn CR, et al. Roles of Insulin Receptor Substrate-1, Phosphatidylinositol 3-Kinase, and Release of Intracellular Ca2+ Stores in Insulin-stimulated Insulin Secretion in [beta]-Cells. J Biol Chem. 2000;275:22331–22338.
    1. Kulkarni RN, Bruning JC, Winnay JN, Postic C, Magnuson MA, et al. Tissue-Specific Knockout of the Insulin Receptor in Pancreatic [beta] Cells Creates an Insulin Secretory Defect Similar to that in Type 2 Diabetes. Cell. 1999;96:329–339. doi: .
    1. Otani K, Kulkarni RN, Baldwin AC, Krutzfeldt J, Ueki K, et al. Reduced {beta}-cell mass and altered glucose sensing impair insulin-secretory function in {beta}IRKO mice. Am J Physiol Endocrinol Metab. 2004;286:E41–E49.
    1. Xu GG, Rothenberg PL. Insulin receptor signaling in the beta-cell influences insulin gene expression and insulin content: evidence for autocrine beta-cell regulation. Diabetes. 1998;47:1243–1252.
    1. Bansal P, Wang Q. Insulin as a physiological modulator of glucagon secretion. Am J Physiol Endocrinol Metab. 2008;295:E751–E761.
    1. Elahi D, Nagulesparan M, Hershcopf RJ, Muller DC, Tobin JD, et al. Feedback inhibition of insulin secretion by insulin: relation to the hyperinsulinemia of obesity. N Engl J Med. 1982;306:1196–1202.
    1. Zhang Y, Zhang N, Gyulkhandanyan A, Xu E, Gaisano H, et al. Presence of functional hyperpolarisation-activated cyclic nucleotide-gated channels in clonal alpha cell lines and rat islet alpha cells. Diabetologia. 2008;51:2290–2298.
    1. Elahi D, Nagulesparan M, Hershcopf RJ, Muller DC, Tobin JD, et al. Feedback inhibition of insulin secretion by insulin: relation to the hyperinsulinemia of obesity. N Engl J Med. 1982;306:1196–1202.
    1. Ueki K, Okada T, Hu J, Liew CW, Assmann A, et al. Total insulin and IGF-I resistance in pancreatic [beta] cells causes overt diabetes. Nat Genet. 2006;38:583–588. 10.1038/ng1787.
    1. Zhao J, Zhang N, He M, Yang Z, Tong W, et al. Increased Beta-Cell Apoptosis and Impaired Insulin Signaling Pathway Contributes to the Onset of Diabetes in OLETF Rats. Cellular Physiology and Biochemistry. 2008;21:445–454.

Source: PubMed

Подписаться