throbber
The Multiple Actions of GLP-1 on the Process of
`Glucose-Stimulated Insulin Secretion
`
`Patrick E. MacDonald,1 Wasim El-kholy,1 Michael J. Riedel,2 Anne Marie F. Salapatek,1
`Peter E. Light,2 and Michael B. Wheeler1
`
`The physiological effects of glucagon-like peptide-1
`(GLP-1) are of immense interest because of the poten-
`tial clinical relevance of this peptide. Produced in intes-
`tinal L-cells through posttranslational processing of the
`proglucagon gene, GLP-1 is released from the gut in
`response to nutrient ingestion. Peripherally, GLP-1 is
`known to affect gut motility, inhibit gastric acid secre-
`tion, and inhibit glucagon secretion. In the central
`nervous system, GLP-1 induces satiety, leading to re-
`duced weight gain. In the pancreas, GLP-1 is now known
`to induce expansion of insulin-secreting ␤-cell mass, in
`addition to its most well-characterized effect: the aug-
`mentation of glucose-stimulated insulin secretion.
`GLP-1 is believed to enhance insulin secretion through
`mechanisms involving the regulation of ion channels
`(including ATP-sensitive Kⴙ channels, voltage-depen-
`dent Ca2ⴙ channels, voltage-dependent Kⴙ channels,
`and nonselective cation channels) and by the regulation
`of intracellular energy homeostasis and exocytosis.
`The present article will focus principally on the mecha-
`nisms proposed to underlie the glucose dependence of
`GLP-1’s insulinotropic effect. Diabetes 51 (Suppl. 3):
`S434 –S442, 2002
`
`Glucagon-like peptide 1 (GLP-1) is a potent in-
`
`cretin hormone produced in the L-cells of the
`distal ileum and colon. In the L-cells, GLP-1 is
`generated by tissue-specific posttranslational
`processing of the proglucagon gene (1). Nutrients, includ-
`ing glucose, fatty acids, and dietary fiber, are all known to
`upregulate the transcription of the gene encoding GLP-1,
`
`From the 1Departments of Medicine and Physiology, University of Toronto,
`Toronto, Ontario, Canada; and the 2Department of Pharmacology, University
`of Alberta, Edmonton, Alberta, Canada.
`Address correspondence and reprint requests to Michael B. Wheeler,
`Department of Physiology, University of Toronto, 1 Kings College Circle,
`Toronto, ON, Canada, M5S 1A8. E-mail: michael.wheeler@utoronto.ca.
`Received for publication 18 March 2002 and accepted in revised form 17
`May 2002.
`AKAP, A-kinase anchoring protein; [Ca2⫹]i, intracellular concentration of
`Ca2⫹; CICR, Ca2⫹-induced Ca2⫹ release; CNS, central nervous system; cPKA,
`catalytic subunit of protein kinase A; DP-IV, dipeptidyl-peptidase IV; ERK,
`extracellular signal-related kinase; GEF-II, guanine nucleotide exchange fac-
`tor II; GI, gastrointestinal; GIP, glucose-dependent insulinotropic peptide;
`GLP-1, glucagon-like peptide-1; GSIS, glucose-stimulated insulin secretion;
`HSL, hormone-sensitive lipase; IP3, inositol triphosphate; KATP, ATP-sensitive
`K⫹ channel; KCa, Ca2⫹-sensitive voltage-dependent K⫹ channel; Kv, voltage-
`dependent K⫹ channel; MAPK, mitogen-activated protein kinase; NSCC,
`nonspecific cation channel; PDX-1, pancreatic duodenal homeobox-1; PI3-K,
`phophatidylinositol 3-kinase; PKA, protein kinase A; PKC␨, protein kinase C␨;
`Po, open probability; SU, sulfonylurea; VDCC, voltage-dependent Ca2⫹ chan-
`nel.
`The symposium and the publication of this article have been made possible
`by an unrestricted educational grant from Servier, Paris.
`
`and they can stimulate the release of this hormone (2).
`Although the majority of L-cells are located in the distal
`ileum and colon, the levels of GLP-1 rise rapidly upon food
`ingestion. It is now well accepted that nutrients, princi-
`pally sugars and fats, liberate GLP-1 and GLP-1–releasing
`factors, including glucose-dependent insulinotropic pep-
`tide (GIP), gastrin-releasing peptide, and selective neural
`regulators that also stimulate GLP-1 secretion (rev. in 1–3).
`Upon its release, GLP-1 affects multiple target tissues
`throughout the body, actions thought to be mediated by a
`single G-protein– coupled receptor isoform. GLP-1 recep-
`tor transcripts and/or protein have been identified in
`several tissues, including pancreatic islets, lung, gastroin-
`testinal (GI) tract, and the central nervous system (CNS)
`(2,3). More questionable is the expression of functional
`GLP-1 receptors in liver and skeletal muscle tissues, where
`gene expression has been detected (4). GLP-1’s ability to
`augment insulin release in a glucose-dependent manner is
`its most well-characterized physiological effect and one of
`its most promising characteristics from a clinical perspec-
`tive.
`
`THE ACTIONS OF GLP-1
`GLP-1 in the pancreas: insulin secretion and ␤-cell
`mass. There are several known and speculated pancreatic
`functions for GLP-1. The GLP-1 receptor is expressed in
`␤-cells, where its activation is proposed to have multiple
`acute and long-term actions (3). With respect to ␤-cell
`function, GLP-1 rapidly and potently stimulates insulin
`secretion, a well-known action that will be discussed
`below. However, GLP-1 also stimulates insulin gene tran-
`scription,
`islet cell growth, and neogenesis, additional
`potentially important functions that may be clinically
`relevant for the treatment of diabetes. In mice, GLP-1
`stimulates ␤-cell proliferation (5), and in rat ␤-cell lines, it
`stimulates DNA synthesis through a phosphatidylinositol
`3-kinase (PI3-K)-dependent pathway (6). Effects on ␤-cell
`insulin gene transcription and proliferation are proposed
`to occur via the upregulation of the transcription factor
`pancreatic duodenal homeobox-1 (PDX-1) (6 – 8). PDX-1
`translocation to the nucleus of RIN 1046-38 cells was also
`shown to be dependent on cAMP/protein kinase A (PKA)
`(9). The actions of GLP-1 on proliferation may involve the
`PI3-K downstream targets extracellular signal-related ki-
`nase (ERK) 1/2 and p38 mitogen-activated protein kinase
`(MAPK) (10). These studies have also demonstrated that a
`GLP-1–induced activation of protein kinase C␨ (PKC␨) is
`implicated in ␤-cell proliferation (10). Thus, at least two
`signaling pathways (PI3-K activation of ERK, MAPK, or
`
`S434
`
`DIABETES, VOL. 51, SUPPLEMENT 3, DECEMBER 2002
`
`MPI EXHIBIT 1090 PAGE 1
`
`

`

`PKC and cAMP/PKA-mediated translocation of PDX-1) can
`be implicated in GLP-1’s effects on ␤-cell proliferation.
`GLP-1 can also increase islet and ␤-cell mass by promoting
`cellular differentiation. Studies have shown that GLP-1 can
`induce the differentiation of rat (ARIP), pancreatic exo-
`crine (AR4J), and human islet progenitor cells and impli-
`cate PDX-1 activation as a potential common mediator
`(3,11). A hint of the clinical relevance of GLP-1 treatment
`to circumvent the loss of ␤-cell capacity can be seen in
`vivo. Prolonged administration of GLP-1 to obese diabetic
`(db/db) mice stimulated insulin secretion, improved blood
`glucose/glycemic excursions, and also enhanced ␤-cell
`neogenesis and islet mass (7). Similar improvements in
`glycemic control and ␤-cell mass could be seen in mice
`with a partial pancreatectomy (12) and in neonatal mice
`treated with streptozotocin (13). Studies examining GLP-1
`loss of function in GLP-1 receptor⫺/⫺ mice demonstrated
`alterations in pancreatic insulin content and insulin gene
`transcription in one study (14) and islet size and compo-
`sition in another (15). It is now thought that the phenotype
`of the GLP-1 receptor⫺/⫺ mouse is likely diminished by
`compensatory upregulation of other signaling peptides,
`including GIP. Together, this work suggests that future
`therapies targeting the GLP-1 signal transduction path-
`ways may be used to improve ␤-cell capacity by improving
`␤-cell mass and secretion.
`GLP-1 in the periphery: gut motility and insulin
`sensitivity. GLP-1 appears to exert a wide range of
`extrapancreatic effects. It participates in an ileal break
`phenomenon by playing an inhibitory role in gastric emp-
`tying and small intestinal transit (16,17). GLP-1 decreases
`gastric motility via direct effects on gastric smooth muscle
`and also inhibits postprandial acid secretion (4). It also
`decreases small intestine movement through inhibition of
`smooth muscle activity, resulting in an overall reduction in
`the absorption of nutrients from the GI tract (4). Reduced
`motility likely causes less severe postprandial glucose
`fluctuations and reduces the need for a large and rapid
`postprandial insulin response. GLP-1 also appears to im-
`prove insulin sensitivity and glucose uptake of both human
`and rat adipose tissue and skeletal muscle (4). Several
`studies suggest that GLP-1 may directly enhance glucose
`disposal in an insulin-independent fashion, although this
`may also result from the overall inhibition of glucagon
`secretion (3). GLP-1 also appears to have some cardiovas-
`cular effects, in part through actions on the CNS, as the
`administration of GLP-1 by intravenous or intracerebro-
`ventricular injection increases heart rate and blood pres-
`sure in rats, an effect that can be reversed by application of
`the antagonist exendin 9-39 or bilateral vagotomy, but not
`when GLP-1 is given by peripheral
`injection (3). The
`physiological relevance of this latter effect is unknown.
`GLP-1 in the CNS: control of appetite and weight.
`GLP-1 has profound effects on feeding behavior. Although
`these actions of GLP-1 could be in part related to its effects
`on intestinal motility, they also appear to involve direct
`effects on hypothalamic feeding centers as GLP-1 recep-
`tors are found in specific nuclei within the hypothalamus
`(18). Acute administration of GLP-1 in humans and ro-
`dents induces satiety and decreases caloric intake (19 –
`21). Administration of the GLP-1 antagonist exendin 9-39
`abrogates the effect of GLP-1 and can itself promote
`
`P.E. MACDONALD AND ASSOCIATES
`
`weight gain (22). Long-term exendin 4 treatment of Zucker
`rats reduces food intake and decelerates weight gain (23).
`Interestingly, GLP-1 receptor⫺/⫺ mice do not become
`obese, possibly because this peptide is not essential for
`weight regulation or because compensatory mechanisms
`are upregulated (24). In humans with type 2 diabetes,
`short-term GLP-1 or exendin 4 administration curbs appe-
`tite and food intake in addition to its insulinotropic actions
`(19,25), suggesting long-term delivery would promote
`weight loss in these patients. The ability of GLP-1 analogs
`to promote weight loss and improve ␤-cell function could
`prove to be ideal for the treatment of type 2 diabetes.
`Type 2 diabetes and GLP-1 receptor agonists. It is well
`known that type 2 diabetes is characterized by defects in
`both insulin secretion and in peripheral insulin sensitivity
`(26). Current treatments for the ␤-cell defect include the
`sulfonylurea (SU) drugs, which were the first therapy
`targeted against insufficient insulin secretion. However,
`these compounds promote insulin secretion independent
`of blood glucose and can therefore cause hypoglycemia
`(27). In addition, as clearly demonstrated in the U.K.
`Prospective Diabetes Study, the ability of SUs to stimulate
`insulin secretion decreases over time, likely reflecting a
`deterioration in ␤-cell function (28). For these reasons,
`there is a need for the development of new drugs to treat
`the ␤-cell defect component of type 2 diabetes. Meglitinide
`analogs belong to a newer family of insulin secretagogues
`with actions similar to the SUs, e.g. ATP-sensitive K⫹
`(KATP) channel inhibition, but have distinct pharmacoki-
`netic and pharmacodynamic properties (rapid onset/short
`duration) (27). The properties of GLP-1 described above,
`including glucose-dependent stimulation of insulin secre-
`tion and the expansion of ␤-cell mass, coupled with the
`inhibition of glucagon secretion and food intake, suggest
`that it would greatly complement current ␤-cell therapies.
`Trials with GLP-1 in diabetic patients have shown it to
`stimulate insulin secretion, inhibit gastric emptying, lower
`circulating glucagon, and improve overall glycemic control
`through both intravenous and subcutaneous injection (1).
`Two major drawbacks in the use of GLP-1 therapy are
`related to its rapid inactivation in circulation and its
`delivery via injection. The issue regarding rapid inactiva-
`tion has been addressed using two promising strategies.
`The first involves the development and use of GLP-1
`analogs resistant to proteolytic cleavage by the enzyme
`dipeptidyl-peptidase IV (DP-IV) (1). Currently, several
`pharmaceutical companies are testing their GLP-1 analogs
`in a variety of models, including humans. The second
`strategy involves the neutralization of the DP-IV enzyme
`itself. Thus far, DP-IV inhibition has been shown to pro-
`long the action of GLP-1, improve glycemia in diabetic
`models, and more recently to delay the onset of diabetes in
`Zucker diabetic fatty rats (29 –32). Although both strate-
`gies are promising, several obstacles still remain. With
`respect to DP-IV inhibition, it is not clear what long-term
`effects such a therapy would have, given that this enzyme
`cleaves a number of biologically active peptides with
`various biological activities, including glucagon, vasoac-
`tive intestinal peptide, glucose-dependent insulinotropic
`polypeptide, neuropeptide Y, and substance P, for exam-
`ple (33). With respect to GLP-1 therapy, it is unlikely that
`it will gain widespread acceptance in the diabetes commu-
`
`DIABETES, VOL. 51, SUPPLEMENT 3, DECEMBER 2002
`
`S435
`
`MPI EXHIBIT 1090 PAGE 2
`
`

`

`GLP-1 AND THE ␤-CELL
`
`FIG. 1. The ionic mechanism (KATP channel– dependent) of
`stimulus-secretion coupling in the pancreatic ␤-cell. 1: Glucose
`enters the ␤-cell via GLUT2 transporters. 2: It is metabolized to
`produce an increase in the intracellular ATP-to-ADP ratio. 3:
`This results in closure of KATP channels. 4: This also results in
`membrane depolarization. 5: Finally, it results in the opening of
`VDCCs. 6: Entry of Ca2ⴙ is the main trigger for exocytosis of
`insulin-containing granules. 7: Also triggered by membrane
`depolarization, Kv and KCa channels open. 8: This repolarizes
`the membrane. 9: Repolarization leads to closure of VDCCs,
`limiting Ca2ⴙ entry and insulin secretion.
`
`coexpressed with the GLP-1 receptor in a mammalian cell
`line. The physiological consequences of GLP-1–facilitated
`KATP channel closure would be to 1) augment the excit-
`ability of cells already above the threshold for insulin
`
`FIG. 2. Effects of GLP-1 on ␤-cell excitability and native and recombi-
`nant KATP channels. A: Membrane potential was recorded from an
`INS-1 cell using the patch-clamp technique in current-clamp mode.
`Application of GLP-1 (20 nmol/l) causes membrane depolarization and
`an increase in the action potential firing rate. A family of ionic currents
`elicited from an individual INS-1 cell using the patch-clamp technique
`in voltage-clamp mode is shown in B, upper panel. Cells were held at
`ⴚ80 mV and stepped at 10-mV intervals from ⴚ120 to 40 mV for 200 ms.
`Recordings were made with 5 mmol/l Kⴙ in the bath solution. In the
`lower panel of B, GLP-1 is shown to inhibit KATP channels in INS-1 cells.
`To measure inward KATP current, cells were held at 0 mV and stepped
`to ⴚ100 mV every 10 s. GLP-1 (20 nmol/l) was superfused over cells for
`3–5 min until a steady-state current was obtained. Tolbutamide sensi-
`tivity was confirmed as a marker of KATP current (traces not shown). C:
`GLP-1 inhibits KATP channels in a recombinant system. tsA201 cells
`were transiently transfected with GLP-1R, Kir6.2, and SUR1 clones
`48 –72 h before recording. Current recordings were performed under
`symmetrical Kⴙ conditions (140 mmol/l Kⴙ in the bath solution). GLP-1
`(20 nmol/l) was applied to the cells in a similar manner to B, upper
`panel. Barium chloride (BaCl2, 2 mmol/l) was used in the recombinant
`system as a fully washable potassium channel blocker to assess the
`amount of recombinant KATP current present. The amphotericin-perfo-
`rated patch-clamp technique mode was used to measure whole-cell
`KATP currents and membrane potentials while maintaining the integrity
`of the intracellular environment.
`
`nity when effective oral agents are available. However, one
`can anticipate that a small molecule that holds some or all
`of the properties of native GLP-1 will eventually be devel-
`oped. To facilitate this process, it is important to under-
`stand how GLP-1 controls insulin secretion.
`
`GLP-1 AND INSULIN SECRETION
`Overview of the ATP-sensitive pathway. Glucose-stim-
`ulated insulin secretion (GSIS) is regulated by a number of
`ionic and nonionic signaling pathways, also known as the
`KATP-dependent and -independent pathways (34,35). The
`KATP-dependent mechanism of stimulus-secretion cou-
`pling is reviewed in Fig. 1. In general, the ␤-cell adapts
`insulin secretion to prevailing blood glucose levels
`through glucose metabolism. When glucose levels rise, the
`rate of glycolysis increases, which generates substrates
`(mainly pyruvate) for mitochondrial oxidative metabo-
`lism, the result of which is the generation of ATP, or more
`correctly an increase in the ATP-to-ADP ratio (36). This
`event provides the functional
`link between a glucose
`stimulus and insulin secretion. The increase in this ratio
`causes the closure of ␤-cell KATP channels, leading to
`plasma membrane depolarization, activation of voltage-
`dependent Ca2⫹ channels (VDCCs), and an increase in the
`intracellular concentration of Ca2⫹ ([Ca2⫹]i), the main
`trigger for insulin secretion. Repolarization of ␤-cells is
`likely mediated by voltage-dependent K⫹ (Kv) channels
`and Ca2⫹-sensitive voltage-dependent K⫹ (KCa) channels
`(37), which open in response to glucose-induced mem-
`brane depolarization to restore the outward flux of K⫹.
`GLP-1 is proposed to modulate GSIS by regulating the
`activity of several ion channels involved in KATP-depen-
`dent insulin secretion as well as steps distal to channel
`modulation.
`GLP-1 and ␤-cell KATP channels. One of the many
`observed cellular effects of GLP-1 is the inhibition of ␤-cell
`KATP channels (38 – 40). The resulting membrane depolar-
`ization induced by KATP channel closure initiates Ca2⫹
`influx through VDCCs and triggers the exocytotic release
`of insulin. Figure 2 shows the excitatory effect of GLP-1 on
`membrane potential and its inhibitory effect on both native
`KATP channel currents from INS-1 cells and currents
`mediated by recombinant KATP channels (SUR1/Kir6.2)
`
`S436
`
`DIABETES, VOL. 51, SUPPLEMENT 3, DECEMBER 2002
`
`MPI EXHIBIT 1090 PAGE 3
`
`

`

`release and 2) increase the percentage of ␤-cells actively
`secreting insulin at glucose concentrations normally sub-
`threshold for the release of insulin.
`The consensus view is that the inhibitory effect of GLP-1
`on KATP channels is cAMP/PKA-dependent (38 – 41), al-
`though one study using rat ␤-cells disagrees (42). This
`assertion by Suga et al. (42) is based on their finding that
`the specific PKA inhibitor Rp-cAMPS (100 ␮mol/l) was
`unable to prevent the cellular depolarization and reduction
`in the whole-cell KATP current elicited by GLP-1. The
`completeness of PKA inhibition by Rp-cAMPS should be
`questioned because in the same study, forskolin induced
`significant insulin secretion even in the presence of Rp-
`cAMPS. Secondly, Suga et al. suggest that GLP-1 causes a
`slight increase in the ATP sensitivity of the KATP channel
`such that at low micromolar ATP concentrations, the KATP
`channel will be more susceptible to closure. However, in
`the normal rat pancreatic ␤-cell, millimolar levels of ATP
`are present (43), and at these physiological ATP levels,
`very similar KATP channel open probability (Po) in the
`absence and presence of GLP-1 are predicted as follows.
`Our calculations indicate that with an intracellular [ATP]
`of 2 mmol/l, the KATP channel Po is reduced from 0.005 to
`0.003 in the presence of GLP-1. It is plausible that a
`leftward shift of ATP sensitivity does occur in the pres-
`ence of GLP-1. However, the observed magnitude of
`GLP-1–induced KATP current reduction seen in whole-cell
`patch-clamp recordings (Fig. 2) is likely too large to be
`accounted for solely by the small decreases in Po calcu-
`lated from the data of Suga et al. (42). Recent work from
`our laboratory has shown that the membrane-permeant
`specific PKA inhibitor H-89 (44) is capable of completely
`inhibiting KATP current reduction by GLP-1 (45). Moreover,
`others have shown similar results using Rp-8-Br-cAMPS, a
`more membrane-permeable analog of Rp-cAMPS (38,41).
`The actions of GLP-1 on KATP channels may also involve
`other signaling pathways because KATP channel inhibition
`by GLP-1 in mouse ␤-cells was demonstrated to be cal-
`modulin dependent, using the calmodulin inhibitors W-7
`and calmidazolium (46).
`The glucose dependency of GLP-1 actions has been well
`established, although the precise mechanisms for this
`dependence are unclear (41,47). However, the cellular
`actions of PKA on the KATP channel may provide a link
`between this kinase and the glucose sensitivity of GLP-1.
`Other groups have shown that addition of the catalytic
`subunit of PKA (cPKA) to excised patches containing KATP
`channels results in an augmentation of KATP current
`(39,48). Our laboratory has recently shown that the effect
`of cPKA on KATP current is dependent on ADP (45). When
`ADP levels are elevated, cPKA increases KATP channel
`current in a recombinant system, consistent with the
`results of Lin et al. (39). Conversely, as ADP levels are
`decreased, cPKA reduces KATP current (45). Physiologi-
`cally, this may result in a negligible enhancement of ␤-cell
`excitability when glucose levels are low (high [ADP]),
`whereas when glucose levels rise (low [ADP]), GLP-1–
`mediated closure of KATP channels, via a PKA-dependent
`pathway, leads to membrane depolarization and subse-
`quent increases in ␤-cell excitability. Special attention
`must be given to the cellular ATP-to-ADP ratio when
`considering KATP channel activity because it is changes in
`
`DIABETES, VOL. 51, SUPPLEMENT 3, DECEMBER 2002
`
`P.E. MACDONALD AND ASSOCIATES
`
`this ratio, more than simply changes in intracellular [ATP]
`per se, that govern the activity of KATP channels in the
`intact ␤-cell. Free ATP is highly buffered within the cell by
`membrane and cytosolic ATPases (43) and is predicted not
`to change significantly with increased glucose metabolism
`(36). In contrast, the reciprocal change in ADP, which is
`not buffered to the same extent, is more significant and
`results in a change in the ATP-to-ADP ratio (36). Indeed,
`the importance of ADP in controlling ␤-cell KATP channel
`activity has been demonstrated because mutations in the
`ADP-sensing region of the human KATP channel lead to
`uncontrolled insulin secretion and hypoglycemia (49). The
`molecular identity of the PKA phosphorylation site(s) is of
`significant importance and is still under investigation.
`Both the Kir6.2 and SUR1 subunits of the KATP channel
`contain putative target sequences for PKA-mediated phos-
`phorylation (39,48), and systematic mutation of these
`residues should clarify the relative contributions of these
`sites to the action of PKA on the KATP channel.
`GLP-1, VDCCs, and intracellular Ca2ⴙ stores. GLP-1
`has been shown to enhance currents through VDCCs in
`mouse, rat, and human ␤-cells (38,50 –52), although the
`magnitude of this effect varies and often does not reach
`statistical significance. In single human ␤-cells, GLP-1
`was shown to increase L-type VDCC activity and the
`amplitude of depolarization-evoked intracellular calcium
`transients, an effect which accounted for 40% of the
`increase in GLP-1–potentiated exocytosis (52). Although
`L-type VDCCs are classically regarded as the major regu-
`lators of Ca2⫹ influx leading to insulin secretion, ␤-cells
`are known to express multiple Ca2⫹ channel isoforms
`(53). Pereverzev et al. (54) reported recently that mice
`lacking the ␣1E-isoform of Cav2.3 have reduced glucose
`tolerance and diminished insulin responses to glucose.
`They speculate that G-protein regulation of this channel
`may modulate insulin secretion based on muscarinic ace-
`tylcholine receptor regulation of VDCCs in vitro (55).
`We have found in HIT-T15 insulinoma cells transfected
`with the GLP-1 receptor that GLP-1 causes an increase in
`voltage-dependent Ca2⫹ currents (see 56 and Fig. 3A). This
`is due, at least partly, to a leftward shift in the voltage
`dependence of activation reminiscent of the effect of
`VDCC phosphorylation by PKA (57,58). We also observed
`a rightward shift in the voltage dependence of steady-state
`inactivation such that in the presence of GLP-1,
`less
`channels were effectively inactivated at a given holding
`potential (50). This is supported by Britsch et al. (50), who
`suggest that GLP-1 treatment of mouse ␤-cells slows the
`inactivation of voltage-dependent Ca2⫹ currents. Addition-
`ally, GLP-1 led to an increase in intracellular calcium only
`after glucose addition, an effect that was blocked in part
`by VDCC antagonists (Fig. 3C). The ability of GLP-1 to
`enhance Ca2⫹ currents is, like the effect on KATP channels,
`cAMP dependent (51,52), based on the ability of Rp-
`cAMPS to prevent an increase in currents. Indeed, treat-
`rat ␤-cells with dibutyryl cyclic-AMP, a
`ment of
`membrane-permeable cAMP analog, replicated the effect
`of GLP-1 on Ca2⫹ currents (51). Additionally,
`in our
`studies (56), the VDCC response to GLP-1 was lost in
`HIT-T15 cells expressing a mutant GLP-1 receptor lacking
`critical residues required for coupling to adenylyl cyclase,
`whereas VDCC activity could still be enhanced by the
`
`S437
`
`MPI EXHIBIT 1090 PAGE 4
`
`

`

`GLP-1 AND THE ␤-CELL
`
`FIG. 3. GLP-1 enhances VDCC activity and [Ca2ⴙ]i responses to glucose
`in HIT-T15 cells overexpressing the wild-type GLP-1 receptor. HIT-T15
`cells overexpressing the wild-type GLP-1 receptor were voltage-
`clamped in the whole-cell configuration under conditions to measure
`voltage-dependent Ca2ⴙ currents, held at ⴚ70 mV and stepped from
`ⴚ60 to 60 mV in 10-mV steps for 250 ms. A: Representative traces for
`basal control and after sequential cumulative addition of GLP-1 (10ⴚ8
`mol/l) and nifedipine (10 ␮mol/l). Current-voltage relationships are
`shown in B. C: A representative trace of [Ca2ⴙ]i in wild-type GLP-1
`receptor–transfected HIT-T15 cell measured with Fura 2-AM after
`sequential cumulative addition of glucose (10 mmol/l), GLP-1, and
`nifedipine as in A and B.
`
`cAMP independent agonist BAYK8644. Recent evidence
`suggests that an A-kinase anchoring protein (AKAP),
`AKAP18, targets PKA to VDCCs and that this kinase may
`be involved in GLP-1 modulation of these channels (59).
`In addition to effects on VDCCs, GLP-1 can mobilize
`intracellular calcium stores in a cAMP-dependent manner
`(60,61), possibly contributing to the oscillatory [Ca2⫹]i
`response to GLP-1 seen in HIT-T15 cells (Fig. 3C) (62). Our
`studies suggest that GLP-1 application results in oscilla-
`tions in [Ca2⫹]i in HIT-T15 cells and that these oscillations
`are not abolished (although they are diminished in ampli-
`tude) by removal of extracellular Ca2⫹ or by VDCC block-
`ade (Fig. 3C). Indeed, in a number of cell types, Ca2⫹
`oscillations induced by an agonist are primarily caused by
`Ca2⫹ release through inositol trisphosphate (IP3) and/or
`ryanodine-sensitive stores (63– 65). In ␤-cells, GLP-1 mo-
`bilizes Ca2⫹ stores in large part by sensitizing the ryano-
`dine receptor (likely the type 2 isoform, RYR-2) to the
`process of Ca2⫹-induced Ca2⫹ release (CICR) (61,66).
`Several studies have demonstrated that GLP-1 can in-
`crease [Ca2⫹]i in a PKA-independent manner (67– 69). This
`mechanism has recently been ascribed to CICR from
`ryanodine-sensitive stores via cAMP-regulated guanine
`nucleotide exchange factor II (GEF-II or Epac2) and its
`interaction with either the Ras-related small G-protein
`Rap1 or with Rab3 small G-protein effector Rim2 (68). The
`importance of cAMP-GEF-II–Rim2 has been demon-
`strated, because inactivation of this complex (by antisense
`
`oligonucleotides or mutant constructs) attenuated the
`secretory response of mouse islets or MIN6 insulinoma
`cells to GLP-1 (67). Because the affinity of cAMP for PKA
`is much higher (⬃100 nmol/l) compared with cAMP–
`GEF-II (⬃10 mmol/l), it is interesting to speculate that the
`GLP-1–stimulated cAMP–GEF-II pathway might operate
`on a rise in local cAMP rather than global changes.
`Although GLP-1 receptor signaling stimulates IP3 produc-
`tion in GLP-1 receptor– expressing COS cells (70), a role
`for IP3-sensitive Ca2⫹ stores in global [Ca2⫹]i is in doubt
`because the GLP-1–stimulated IP3 production in primary
`␤-cells is reportedly minimal (71,72) and the IP3 receptor
`antagonist xestospongin C failed to block release of intra-
`cellular Ca2⫹ stores by forskolin treatment (68). However,
`IP3-regulated Ca2⫹ release from insulin granules has been
`suggested by the studies of Nakagaki et al. (62), who
`suggest that GLP-1 may uniquely regulate temporal and
`spatial release of intracellular calcium through local IP3
`signaling. Thus, GLP-1–mediated release of intracellular
`stores, along with potentiation of Ca2⫹ entry through
`VDCCs, likely contribute to the insulinotropic effect of
`GLP-1.
`GLP-1 and ␤-cell Kv channels. Voltage-dependent K⫹
`currents, such as those mediated by Kv or KCa channels,
`mediate repolarization of ␤-cells after a depolarizing stim-
`ulus, such as glucose (37). Recently, we reported that Kv1
`and Kv2 family channels regulate insulin secretion, be-
`cause dominant-negative functional knockout of either of
`these channel families enhanced GSIS (73). Kv2.1 channels
`mediate the majority of this effect (⬎60%), the mechanism
`of which involves enhanced glucose-stimulated membrane
`depolarization and Ca2⫹ entry (unpublished observations).
`Because ␤-cell Kv currents are potent glucose-dependent
`regulators of insulin secretion, we hypothesized that phys-
`iological secretagogues, such as GLP-1, may regulate Kv
`channel function. Indeed, we report elsewhere in this
`supplement that the GLP-1 receptor agonist exendin 4
`inhibits voltage-dependent outward K⫹ currents in rat
`␤-cells voltage-clamped in the whole-cell configuration by
`40% and significantly prolongs the time course of ␤-cell
`repolarization after transient depolarization by current
`injection. This compares to an 86% reduction in outward
`K⫹ currents achieved with the general Kv channel antag-
`onist tetraethylammonium. GLP-1 antagonized voltage-
`dependent outward K⫹ currents in rat ␤-cells in the
`absence of glucose. However, this effect may still contrib-
`ute to the glucose dependence of GLP-1’s insulinotropic
`effect, because Kv channels are not normally expected to
`be active until after a glucose-induced depolarization of
`the cell membrane (37). Additionally, and similar to the
`effect of GLP-1 on the other ion channels mentioned
`above, exendin 4 –mediated inhibition of ␤-cell Kv chan-
`nels is dependent on cAMP signaling. One recent study,
`however, suggested that cAMP signaling was not sufficient
`in itself to antagonize voltage-dependent K⫹ currents in an
`insulin-secreting cell line (INS-1) (74).
`Numerous studies have described effects of hormone-
`mediated alterations in voltage-dependent K⫹ currents,
`both excitatory and inhibitory. The best-characterized of
`these effects is the voltage-dependent K⫹ current down-
`regulation in lymphocytes and upregulation in cardiac
`myocytes (75,76). In both of these tissues, the cAMP/PKA
`
`S438
`
`DIABETES, VOL. 51, SUPPLEMENT 3, DECEMBER 2002
`
`MPI EXHIBIT 1090 PAGE 5
`
`

`

`signaling pathway has been implicated in the regulation of
`these channels (76,77). Reports suggest that cAMP can
`reduce voltage-dependent K⫹ currents in murine lympho-
`cytes (76) and a pituitary cell
`line (78) but enhance
`voltage-dependent K⫹ currents in cardiac myocytes (77), a
`finding that has been confirmed at the single-channel level
`in frog atrial myocytes (79) and the giant squid axon (80).
`Phosphorylation may occur directly on the channel, be-
`cause PKA phosphorylation of an atrial Kv channel near
`the NH2-terminus enhanced channel activity (81), and
`phosphorylation of Kv1 channel ␣-subunits regulates the
`extent of inhibition of these channels conferred by a
`regulatory ␤-subunit (82). Phosphorylation of ␤-subunits
`themselves may also modulate the regulatory interaction
`with pore-forming ␣-subunits (83). It has recently been
`demonstrated that regulation of a cardiac Kv channel
`(KvLQT) by cAMP requires the expression of AKAP15/18
`or AKAP79 (84). Additionally, an increase in voltage-
`dependent K⫹ current is implicated in epinephrine-in-
`duced inhibition of the glucose-dependent increase in
`[Ca2⫹]i in ob/ob and ⫹/⫹ mouse ␤-cells (85) since the
`effect was reversed by tetraethylammonium. Interestingly,
`the inhibitory effect of epinephrine on [Ca2⫹]i was also
`reversed by the adenylyl cyclase activator forskolin (85).
`Therefore, we believe there is mounting evidence to
`suggest that hormonal modulation of Kv currents is phys-
`iologically important. Specifically, GLP-1 inhibition of
`these currents is expected to lead to enhanced ␤-cell
`excitability.
`GLP-1 and other ␤-cell ion channels. Increases in
`intracellular cAMP have long been known to enhance Na⫹
`currents (86), an effe

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket