当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 糖尿病学杂志 > 2005年 > 第7期 > 正文
编号:11256649
Inhibition of Purinoceptors Amplifies Glucose-Stimulated Insulin Release With Removal of its Pulsatility
     1 Institute of Physiological Sciences, University of Lund, Lund, Sweden

    2 Department of Surgery, University of Lund, Lund, Sweden

    3 Department of Medical Cell Biology, University of Uppsala, Uppsala, Sweden

    ABSTRACT

    External ATP has been proposed to be an autocrine regulator of glucose-stimulated insulin secretion and responsible for the synchronization of the Ca2+ rhythmicity in the -cells required for a pulsatile release of insulin from the pancreas. The importance of external ATP for glucose-stimulated insulin release was evaluated in rats with the aid of 2-deoxy-N-methyladenosine-3,5-bisphosphate (MRS 2179), an inhibitor of the purinoceptors known to affect the Ca2+ signaling in -cells. The concentration of cytoplasmic Ca2+ was measured in single -cells and small aggregates with ratiometric fura-2 technique and the release of insulin recorded from isolated islets and the perfused pancreas. Addition of 1 eol/l ATP induced premature cytoplasmic Ca2+ concentration ([Ca2+]i) oscillations similar to those found in -cells exposed to 20 mmol/l glucose. In most experiments, the presence of 10 eol/l MRS 2179 did not remove the glucose-induced [Ca2+]i rhythmicity in single -cells or the synchronization seen in coupled cells. Nevertheless, the same concentration of MRS 2179 promptly interrupted the pulsatility (frequency 0.22 ± 0.01/min) of insulin secretion, raising the total amounts released from the pancreas. Prolonged exposure of islets to 1 and 10 eol/l MRS 2179 enhanced insulin secretion at 20 mmol/l glucose 33% (P < 0.05) and 63% (P < 0.01), respectively, without affecting the release at 3 mmol/l glucose. The results support the idea that neural ATP signals entrain the islets into a common rhythm resulting in pulsatile release of insulin and that glucose stimulation of the secretory activity is counteracted by accumulation of inhibitory ATP around the -cells.

    Increase of the cytoplasmic concentration of Ca2+ stimulates a number of cellular events, including release of insulin from the pancreatic -cells. Each -cell is a biological oscillator, responding to a glucose stimulus with rhythmic depolarization and subsequent entry of Ca2+ via voltage-activated channels (1). The resulting oscillations of cytoplasmic Ca2+ concentrations ([Ca2+]i) vary considerably among isolated -cells. However, when coupled, the -cells interact with mutual entrainment into a common rhythm, mediated by gap junctions and diffusible messengers (2). Accordingly, it is possible to demonstrate release of insulin in pulses from isolated islets, the frequency of which is fairly constant irrespective of islet size (3). Cyclic variations of circulating insulin result from pulsatile release of the hormone into the portal vein (4,5). A prerequisite for this pulsatility is that the -cell oscillations of [Ca2+]i are coordinated not only within the islets but also appear in the same phase in all islets of the pancreas.

    The purine nucleotide ATP is an external messenger (6,7) released intermittently from nerves and the -cells themselves. ATP has been reported to propagate mechanically induced [Ca2+]i rises in confluent monolayers of rat insulinoma cells (8) and normal mouse -cells (9). Indeed, it was proposed that ATP entrains the secretory activity of the -cells into a common rhythm by inducing synchronous transients superimposed on the oscillations of [Ca2+]i (2,10). Released together with insulin during exocytosis, ATP may also serve as an autocrine regulator of the -cell function (11eC16).

    The role of the purinoceptors in the release of insulin is a matter of controversy, and both stimulatory and inhibitory effects of ATP have been reported (17). It was recently observed in -cells from ob/ob mice that a low concentration of the purinoceptor antagonist 2-deoxy-N-methyladenosine-3,5-bisphosphate (MRS 2179) removes the [Ca2+]i transients supposed to coordinate the secretory activity (2). We now demonstrate in studies of rats that the same inhibitor amplifies glucose-stimulated insulin release with removal of the pulsatility.

    RESEARCH DESIGN AND METHODS

    Female Sprague-Dawley rats, weighing 250eC300 g, were used. The animals were allowed free access to food and were killed by cervical dislocation. Before perfusion of pancreas, the rats were anesthetized with 5% chloral hydrate (1 ml/100 g body wt). The experiments were approved by a local ethical committee.

    Chemicals and solutions.

    Reagents of analytical grade and deionized water were used. Roche Diagnostics (Mannheim, Germany) supplied collagenase, BSA, and HEPES. ATP, glucagon, and methoxyverapamil were obtained from Sigma Chemical (St. Louis, MO). The acetoxymethyl ester of fura-2 was purchased from Molecular Probes (Eugene, OR), and MRS 2179 was a product of Tocris Cookson (Bristol, U.K.). The studies were performed at 37°C with a basal medium of Krebs ringer bicarbonate buffer, supplemented with 10 mmol/l HEPES, and gassed with 5% CO2 + 95% O2 to pH 7.4 (18). Medium concentrations of insulin were measured in duplicate with radioimmunoassay (19), and plasma levels of glucose were determined with the glucose oxidase technique.

    Isolation of islets and preparation of -cells.

    Islets were isolated with the aid of collagenase. When not taken for studies of insulin release, the islets were used for preparation of single cells or small aggregates (<10 cells) by shaking in a Ca2+-deficient medium. After suspension in RPMI 1640 medium supplemented with 10% FCS, 100 IU/ml penicillin, 100 e/ml streptomycin, and 30 e/ml gentamicin, the cells were allowed to attach to circular coverslips during 2eC5 days culture at 37°C in an atmosphere of 5% CO2 in humidified air. The selection of -cells for analyses was based on their large size and low nucleareCtoeCcytoplasmic volume ratio compared with the islet cells producing glucagon and somatostatin (20).

    Measurements of cytoplasmic Ca2+ in isolated -cells.

    The cells were loaded with 0.5eC1 eol/l fura-2 acetoxymethyl ester, and [Ca2+]i was measured during superfusion at 37°C using an inverted microscope equipped for epifluorescence fluorometry (10). Fura-2 was excitated with the aid of a monochromator (Cairn Optoscan; Faversham, Kent, U.K.), and images were collected at 510 nm with an intensified CCD camera (Extended Isis-M; Phototonic Science, Robertsbridge, U.K.). Pairs of 340- and 380-nm images, consisting of 10 accumulated video frames, were captured, followed by a delay resulting in measuring cycles of 2 s. The specimens were only illuminated during the image capture, and the excitation light was minimized by means of neutral density filters. Ratio frames were calculated after background subtraction, and [Ca2+]i was estimated as previously described (21,22) using the MetaFluor program (Universal Imaging, Downingtown, PA).

    Measurements of insulin release from isolated islets.

    Freshly isolated islets were preincubated for 30 min in basal medium supplemented with 0.1% BSA and 3 mmol/l glucose. Each vial contained 25 islets in 1.0 ml medium. After preincubation, the medium was replaced and the islets exposed to test agents. Aliquots of medium were taken for analyses both after 12 and 45 min of incubation.

    Measurements of insulin release from the perfused pancreas.

    Pancreas perfusion was performed by modifying the procedure of Grodsky and Fanska (23) to allow contribution of blood from the donor. Unrecycled medium supplemented with 2% BSA (wt/vol) was infused at a rate of 0.4 ml/min. After 5 min of equilibration, effluent from the portal vein was collected as 30-s portions using heparinized vials, centrifuged, and the plasma stored at eC20°C. The contribution of blood to the perfusion medium was 25%, as estimated from a comparison of the glucose concentration in the effluent with that in the circulating blood.

    Analysis of data.

    In the measurements of [Ca2+]i, each experiment refers to analyses of 6eC12 cells attached to a coverslip. In the experiments with pancreas perfusion, the "Cluster Analysis " program (24) was used as a waveform-independent technique to detect significant insulin peaks. Nadir and peak test cluster sizes were set to 2. Minimum t statistics of 4.1 were used as stringency criteria for upstrokes and downstrokes. The settings detected peaks with <1% false-positive errors. Insulin data in the tables are presented as means ± SE and evaluated statistically with Student's t test.

    RESULTS

    Individual -cells responded to 20 mmol/l glucose with oscillations of [Ca2+]i, varying considerably in frequency (0.1eC1/min) and shape (Figs. 1 and 2). However, when situated in aggregates, the -cells were well synchronized (Fig. 3). Addition of 1 eol/l ATP elicited premature [Ca2+]i oscillations similar to the ordinary ones (Fig. 1). The glucose-induced [Ca2+]i rhythmicity usually persisted (30 of 48 cells) in the presence of 10 eol/l MRS 2179. In responding cells, the oscillations tended to be transformed into sustained elevation of [Ca2+]i (Fig. 2). The exposure to MRS 2179 did not affect the synchronization of the [Ca2+]i rhythmicity seen in coupled -cells (Fig. 3).

    Increase of the glucose concentration from 3 to 20 mmol/l resulted in distinct pulses (frequency 0.22 ± 0.01/min; n = 5) of insulin release from the perfused pancreas (Fig. 4). Initial rise of the insulin concentration from 114 ± 22 to 1,087 ± 62 pmol/l (n = 5) was followed by cyclic variations with lower peak values. The effects of MRS 2179 on glucose-stimulated insulin release from the pancreas are shown in Fig. 5. At a concentration of 1 eol/l, MRS 2179 affected neither the amplitude nor the frequency of the oscillations (Fig. 5A). However, exposure to 10 eol/l MRS 2179 was accompanied by decreased variations of the release (Table 1) and loss of significant insulin pulses verified by cluster analysis (Fig. 5B). The latter effect was due to interruption of the downstroke of the oscillations with resulting increase (P < 0.001) of average insulin release (Table 1).

    Further evidence that inhibition of purinoceptors results in an enhanced insulin release from -cells stimulated with glucose was obtained in experiments with isolated islets. As shown in Table 2, neither 1 nor 10 eol/l MRS 2179 modified the release of insulin during an initial 12-min period. Subsequent exposure to MRS 2179 did not affect the insulin secretion at 3 mmol/l glucose but increased the amounts released at 20 mmol/l glucose with 33% (1 eol/l MRS 2179) and 63% (10 eol/l MRS 2179), respectively.

    DISCUSSION

    Divergent opinions have been expressed whether external ATP has stimulatory or inhibitory effects on insulin release. The controversy is not only due to species differences in the types of purinoceptors expressed in the -cells. In the rat, the addition of ATP to a medium containing 8.3 mmol/l glucose has been reported to stimulate insulin release from both isolated islets (11) and the perfused pancreas (25). However, other studies have indicated a dual action of ATP on rat islets exposed to 8.3 mmol/l glucose with inhibition of insulin release when the ATP concentration exceeds 1 eol/l (17). The present data confirm the existence of an inhibitory component in the action of external ATP, demonstrating that the purinoceptor antagonist MRS 2179 augments insulin release at a concentration of glucose (20 mmol/l), which promotes exocytosis.

    It is evident from analyses of isolated mouse -cells that activation of purinoceptors results in prompt depolarization (15), generating premature oscillations based on entry of Ca2+ (10). The effect was mimicked by other stimulators of phospholipase A2 (PLA2) and disappeared when this enzyme was inhibited. It is known from studies of insulin-secreting HIT cells (26) and ordinary mouse -cells (15,27) that the products of PLA2 (arachidonic acid and lyso-phospholipids) have a suppressive effect on ATP-sensitive K+ channels. We now observe that ATP generates premature oscillations of [Ca2+]i also in rat -cells, suggesting that purinoceptor activation of PLA2 is a general mechanism for depolarization of pancreatic -cells.

    Like in other species, the -cells from rats have an intrinsic ability to generate slow oscillations of [Ca2+]i (28eC32). There is no direct proof for a corresponding rhythmicity of circulating insulin, since the studies performed so far with peripheral blood refer to samples taken at intervals as long as 3 min (33). Analyses of 1-min portions of the perifusate from rat pancreas revealed insulin pulses of 6 min (34,35). Increasing the sampling intensity to 30 s, we now demonstrate statistically significant peaks of insulin release with a duration of 4.5 min.

    The isolated -cells differed considerably with regard to shape and frequency of their [Ca2+]i oscillations. To understand how -cells in a rat pancreas overcome these differences and generate pulsatile release of insulin into the portal vein, it is important to distinguish between the coordination within and among the islets (10). Within the islet, the -cells are supposed to interact with mutual entrainment of the [Ca2+]i oscillations into a common rhythm via both gap junctions and diffusible messengers. Evidence has been provided that -cells receive and propagate ATP signals, which act as a coupling force for synchronization by temporarily interrupting the electrical activity (2,10). The efficiency of the intraislet coordination of the rat -cells is illustrated from the observation that pulses of insulin release from different islets have a similar frequency (3). Periodic variations of circulating insulin require that the -cell oscillations of [Ca2+]i appear in the same phase in the numerous islets of a pancreas. Previous studies (36) of insulin release from the perfused rat pancreas support the idea that the islets communicate via neurons. Indeed, there are reasons to believe that intermittent release of ATP from nerves entrains the differently phased islets into a common rhythm (10).

    The effects of external ATP on rat -cells are mediated by both unspecific cation channels, referred to as P2X receptors, and G-proteineCcoupled P2Y receptors (25). Activation of rapidly desensitizing P2X receptors evokes a small and transient increase of basal insulin release at low glucose concentrations (37). However, the P2Y receptors are important modulators of the release of insulin evoked by stimulatory concentrations of glucose (17,37). So far, receptor genes for P2Y1, P2Y2, P2Y4, and P2Y6 have been detected in rat islets (38), and the P2Y4 subtype was found in -cells with immunocytochemistry (39). Addition of 100 eol/l pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid, an antagonist of the P2Y4 and P2Y6 receptors (7), has been reported to lack effects on the release of insulin from rat islets exposed to 8.3 mmol/l glucose (17). Using the antagonist MRS 2179, which is relatively specific for the P2Y1 receptors (40,41), we now observe a >60% increase of insulin release in the presence of 20 mmol/l glucose. As an adenosine derivative with two phosphate groups, it is unlikely that MRS 2179 penetrates into the -cells. Withdrawal of MRS 2179 during superfusion of -cells from ob/ob mice is known to result in a prompt reversal of its inhibitory effect on [Ca2+]i transients (2).

    Several reports (11eC16) have drawn attention to exocytotic release of ATP as a means for autocrine control of insulin secretion. The present observations make it possible to propose both positive and negative feedback loops for the ATP released from a glucose-stimulated -cell. It is well established that a metabolically derived signal (ATP-to-ADP ratio) accounts for the slow (2eC5 min) periods of depolarization responsible for the Ca2+ entry generating the insulin pulses (1,42). Initially, the discharge of ATP can be expected to have a positive feedback on insulin release by both augmenting the depolarization via activation of PLA2 (see above) and mobilizing the Ca2+ stored in the endoplasmic reticulum. However, during the oscillatory cycles, there will be an accumulation of inhibitory ATP around the -cells so that a negative feedback loop will dominate. The presence of negative feedback is consistent with the report that ATP (>1 eol/l) acts as a G-proteineCcoupled inhibitor of L-type Ca2+ channels in rat -cells (16). The concentration of ATP reached at the surface of glucose-stimulated rat -cells has been reported to be >25 eol/l (14). The concept of accumulation of inhibitory ATP around the -cells explains why the addition of the purinoceptor antagonist MRS 2179 enhanced insulin release from rat islets exposed to 20 mmol/l glucose.

    A prerequisite for pulsatile release of insulin from pancreas is that the -cells generate oscillations of [Ca2+]i and that this secretory signal will be entrained into a common rhythm. Within the islets, the synchronization of the -cell rhythmicity is supposed to occur via both gap junctions (43) and diffusion of ATP (2). Neural activity with intermittent discharge of ATP will then provide the phase shift of the [Ca2+]i oscillations, coordinating the secretory activity of the different islets in the pancreas (10). The present data reinforce the existing arguments that external ATP has a key role for insulin secretion by demonstrating prompt disappearance of insulin pulses during exposure to a purinoceptor antagonist. The observations that MRS 2179 removes the pulsatility of insulin release into the portal vein with maintenance of -cell oscillations of [Ca2+]i and their synchronization in aggregates suggest that the neural coordination of the islets is critically dependent on activation of purinoceptors.

    ACKNOWLEDGMENTS

    This study was supported by grants from the Swedish Research Council (72X-562), the Swedish Diabetes Association, the Albert Phlsson Foundation, the Crafoord Foundation, and the Family Ernfors Foundation.

    The technical assistance of Britt-Marie Nilsson is gratefully acknowledged.

    [Ca2+]i, cytoplasmic Ca2+ concentration; MRS 2179, 2-deoxy-N-methyladenosine-3,5-bisphosphate; PLA2, phospholipase A2

    REFERENCES

    Hellman B, Gylfe E, Grapengiesser E, Lund P, Berts A: Cytoplasmic Ca2+ oscillations in pancreatic -cells. Biochim Biophys Acta1113 :295 eC305,1992

    Hellman B, Dansk H, Grapengiesser E: Pancreatic -cells communicate via intermittent release of ATP. Am J Physiol Endocrinol Metab286 :E759 eCE765,2004

    Bergsten P, Hellman B: Glucose-induced amplitude regulation of pulsatile insulin secretion from individual pancreatic islets. Diabetes42 :670 eC674,1993

    Lefeebvre PJ, Paolisso G, Scheen AJ, Henquin JC: Pulsatility of insulin and glucagon release: physiological significance and pharmacological implications. Diabetologia30 :443 eC452,1987

    Prksen N, Munn S, Steers J, Vore J, Veldhuis P, Butler P: Pulsatile insulin secretion accounts for 70% of total insulin secretion during fasting. Am J Physiol269 :E478 eCE488,1995

    Williams M, Jarvis MF: Purinergic and pyrimidinergic receptors as drug targets. Biochem Pharmacol59 :1173 eC1185,2000

    Burnstock G: Purine-mediated signalling in pain and visceral perception. Trends Pharm Sci22 :182 eC188,2001

    Cao D, Lin G, Westphale EM, Beyer EC, Steinberg TH: Mechanisms for the coordination of intercellular calcium signaling in insulin-secreting cells. J Cell Sci110 :497 eC504,1997

    Bertuzzi F, Davalli AM, Nano R, Socci G, Codazzi F, Fesce R, Di Carlo V, Pozza G, Grohovaz F: Mechanisms of coordination of Ca2+ signals in pancreatic islet cells. Diabetes48 :1971 eC1978,1999

    Grapengiesser E, Dansk H, Hellman B: Pulses of external ATP aid to the synchronization of pancreatic -cells by generating premature Ca2+oscillations. Biochem Pharmacol68 :667 eC674,2004

    Blachier F, Malaisse WJ: Effect of exogenous ATP upon inositol phosphate production, cationic fluxes and insulin release in pancreatic islet cells. Biochim Biophys Acta970 :222 eC229,1988

    Hellman B, Gylfe E, Wessleen N, Hallberg A, Grapengiesser E, Marcstrm A: Plasma membrane associated ATP as a regulator of the secretory activity of the pancreatic -cell. Exp Clin Endocrinol93 :125 eC135,1989

    Squires PE, James RFL, London NJM, Dunne MJ: ATP-induced intracellular Ca2+ signals in isolated human insulin secreting cells. Pfle筭ers Arch427 :181 eC183,1994

    Hazama A, Hayashi S, Okada Y: Cell surface measurement of ATP release from single pancreatic -cells using a novel biosensor technique. Pfle筭ers Arch437 :31 eC35,1998

    Poulsen CR, Bokvist K, Olsen HL, Hy M, Capito K, Gilon P, Gromada J: Multiple sites of purinergic control of insulin secretion in mouse pancreatic -cells. Diabetes48 :2171 eC2181,1999

    Gong Q, Kakei M, Koriyama N, Nakazaki M, Morimitsu S, Yaekura K, Tei C: P2Y-purinoceptor mediated inhibition of L-type Ca2+ channels in rat pancreatic -cells. Cell Struct Funct25 :279 eC289,2000

    Verspohl EJ, Johannwille B, Waheed A, Neye H: Effect of purinergic agonists and antagonists on insulin secretion from INS-1 cells (insulinoma cell line) and rat pancreatic islets. Can J Physiol Pharmacol80 :562 eC568,2002

    Henningsson R, Alm P, Lindstrm E, Lundquist I: Chronic blockade of NO synthase paradoxically increases islet NO production and modulates islet hormone release. Am J Physiol Endocrinol Metab279 :E95 eCE107,2000

    Heding LH: Determination of free and antibody-bound insulin in insulin treated diabetic patients. Horm Metab Res1 :145 eC146,1969

    Hellman B: The effect of ageing on the total volumes of the A and B cells in the islets of Langerhans in the rat. Acta Endocrinol (Copenh)33 :92 eC112,1959

    Grynkiewicz G, Poenie M, Tsien RY: A new generation of Ca2+ indicators with greatly improved fluorescence properties. J Biol Chem260 :3440 eC3450,1985

    Gylfe E, Grapengiesser E, Hellman B: Propagation of cytoplasmic Ca2+ oscillations in clusters of pancreatic -cells exposed to glucose. Cell Calcium12 :229 eC240,1991

    Grodsky GM, Fanska RE: The in vitro perfused pancreas. Methods Enzymol39 :364 eC372,1975

    Veldhuis JD, Johnson ML: Cluster analysis: a simple, versatile, and robust algorithm for endocrine pulse detection. Am J Physiol250 :E486 eCE493,1986

    Hillaire-Buys D, Chapal J, Bertrand G, Petit P, Loubatieeres-Mariani MM: Purinergic recptors on insulin-secreting cells. Fundam Clin Pharmacol8 :117 eC127,1994

    Eddlestone GT: ATP-sensitive K+ channel modulation by products of PLA2 action in the insulin-secreting HIT cell line. Am J Physiol268 :C181 eCC190,1995

    Thams P, Capito K: Inhibition of glucose-induced insulin secretion by the diacylglycerol lipase inhibitor RHC 80267 and the phospholipase A2 inhibitor ACA through stimulation of K+ permeability without diminuition by exogenous arachidonic acid. Biochem Pharmacol53 :1077 eC1086,1997

    Wang JL, McDaniel ML: Secretagogue-induced oscillations of cytoplasmic Ca2+ in single beta and alpha-cells obtained from pancreatic islets by fluorescence-activated cell sorting. Biochem Biophys Res Commun166 :813 eC818,1990

    Herchuelz A, Pochet R, Pastiels C, Van Praet A: Heterogeneous changes in [Ca2+]i induced by glucose, tolbutamide and K+ in single rat pancreatic B cells. Cell Calcium12 :577 eC586,1991

    Theler JM, Mollard P, Gueerineau N, Vacher P, Pralong WF, Schlegel W, Wollheim CB: Video imaging of cytosolic Ca2+ in pancreatic -cells stimulated by glucose, carbachol, and ATP. J Biol Chem267 :18110 eC18117,1992

    Yada T, Nakata M, Shiraishi T, Kakei M: Inhibition by simvastatin, but not pravastatin, of glucose-induced cytosolic Ca2+ signalling and insulin secretion due to blockade of the L-type Ca2+ channels in rat islet -cells. Br J Pharmacol126 :1205 eC1213,1999

    Ahmed M, Grapengiesser E: Ca2+ handling of rat pancreatic -cells exposed to ryanodine, caffeine, and glucagon. Endocrine17 :103 eC108,2002

    Chou HF, McGivern R, Berman N, Ipp E: Oscillations of circulating plasma insulin concentrations in the rat. Life Sci48 :1463 eC1469,1991

    Stagner JI, Samols E: Comparison of insulin and glucagon pulsatile secretion between the rat and dog pancreas in-vitro. Life Sci43 :929 eC934,1988

    Safarik RH, Joy RM, Curry DL: Episodic release of insulin by rat pancreas: effects of CNS and state of satiety. Am J Physiol254 :E384 eCE388,1988

    Stagner JI, Samols E: Perturbation of insulin oscillations by nerve blockade in the in vitro canine pancreas. Am J Physiol248 :E516 eCE521,1985

    Petit P, Hillaire-Buys D, Manteghetti M, Debrus S, Chapal J, Loubatieeres-Mariani MM: Evidence for two different types of P2 receptors stimulating insulin secretion from pancreatic B cell. Br J Pharmacol125 :1368 eC1374,1998

    Bokvist K, Efanov A, Sandusky G, Sewing S, Treinies I, Gromada J: The P2Y4 pyrimidinergic receptor is important for nucleotide stimulation of insulin secretion in rat pancreatic -cells (Abstract). Diabetes Metab29 :4S 77eC4S78,2003

    Coutinho-Silva R, Parsons M, Robson T, Lincoln J, Burnstock G: P2X and P2Y purinoceptor expression in pancreas from streptozotocin-diabetic rats. Mol Cell Endocrinol204 :141 eC154,2003

    Boyer JL, Mohanram A, Camaioni E, Jacobson KA, Harden TK: Competitive and selective antagonism of P2Y1 receptors by N6-methyl 2'-deoxyadenosine 3',5'-bisphosphate. Br J Pharmacol124 :1 eC3,1998

    Brown SG, King BF, Kim YC, Burnstock G, Jacobson KA: Activity of novel adenine nucleotide derivatives as agonists and antagonists at recombinant P2X receptors. Drug Dev Res49 :253 eC259,2000

    Ainscow EK, Rutter GA: Glucose-stimulated oscillations in free cytosolic ATP concentration imaged in single islet -cells: evidence for a Ca2+-dependent mechanism. Diabetes51 (Suppl. 1) :S162 eCS170,2002

    Meda P: Cx36 involvement in insulin secretion: characteristics and mechanism. Cell Commun Adhes10 :431 eC435,2003(Albert Salehi, Saleem S. )