diabetes research and clinical practice 106 (2014) 1–10

Contents available at ScienceDirect

Diabetes Research and Clinical Practice journ al h ome pa ge : www .elsevier.co m/lo cate/diabres

Invited review

The forgotten members of the glucagon family Dominique Bataille a,*, Ste´phane Dalle b a

INSERM, 269, Rue Adrien Proby, 34090 Montpellier, France INSERM, Research—Pathophysiology of the Pancreatic b Cell, Institute of Functional Genomic, INSERM U 661, CNRS UMR 5203, Universities Montpellier 1 & 2, Montpellier, France b

article info

abstract

Article history:

From proglucagon, at least six final biologically active peptides are produced by tissue-

Received 13 June 2014

specific post-translational processing. While glucagon and GLP-1 are the subject of perma-

Accepted 26 June 2014

nent studies, the four others are usually left in the shadow, in spite of their large biological

Available online 3 July 2014

interest. The present review is devoted to oxyntomodulin and miniglucagon, not forgetting glicentin, although much less is known about it. Oxyntomodulin (OXM) and glicentin are

Keywords:

regulators of gastric acid and hydromineral intestinal secretions. OXM is also deeply

Proglucagon

involved in the control of food intake and energy expenditure, properties that make this

Glucagon

peptide a credible treatment of obesity if the question of administration is solved, as for any

Oxyntomodulin

peptide. Miniglucagon, the C-terminal undecapeptide of glucagon which results from a

Glicentin

secondary processing of original nature, displays properties antagonistic to that of the

Miniglucagon

mother-hormone glucagon: (a) it inhibits glucose-, glucagon- and GLP-1-stimulated insulin

Post-translational processing

release at sub-picomolar concentrations, (b) it reduces the in vivo insulin response to glucose with no change in glycemia, (c) it displays insulin-like properties at the cellular level using only a part of the pathway used by insulin, making it a good basis for developing a pharmacological workaround of insulin resistance. # 2014 Elsevier Ireland Ltd. All rights reserved.

Contents 1. 2.

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Glucagon-containing C-terminally elongated peptides . . . . . . . . . . . 2.1. Structures of oxyntomodulin and glicentin . . . . . . . . . . . . . . 2.2. Compared activities of OXM and glucagon . . . . . . . . . . . . . . . 2.2.1. Gastric acid secretion . . . . . . . . . . . . . . . . . . . . . . . . . 2.2.2. Other sites of action in the GI tract . . . . . . . . . . . . . . 2.2.3. Sites of action in the CNS. . . . . . . . . . . . . . . . . . . . . . 2.3. Questions about the mode(s) of action of OXM and glicentin

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

* Corresponding author. Tel.: +33 467541697; fax: +33 467541697. E-mail addresses: [email protected] (D. Bataille), [email protected] (S. Dalle). http://dx.doi.org/10.1016/j.diabres.2014.06.010 0168-8227/# 2014 Elsevier Ireland Ltd. All rights reserved.

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

. . . . . . . .

2 3 3 3 3 3 4 4

2

diabetes research and clinical practice 106 (2014) 1–10

3.

4.

1.

Glucagon–insulin relationship . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.1. Glucagon–insulin relationship in the islets of Langerhans . 3.1.1. The observations . . . . . . . . . . . . . . . . . . . . . . . . . . 3.1.2. The molecular mechanisms . . . . . . . . . . . . . . . . . . 3.2. Glucagon and insulin secretion. . . . . . . . . . . . . . . . . . . . . . 3.3. Miniglucagon . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.3.1. Miniglucagon in the islet of Langerhans . . . . . . . . 3.3.2. Miniglucagon in the whole organism . . . . . . . . . . . Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

Introduction

Proglucagon (Fig. 1) is, besides the proopiomelanocortin (POMC), the archetype of a prohormone the post-translational processing of which provides the organism with a series of peptides with different, yet complementary biological activities. All activities of the proglucagon-derived peptides are related to the control of nutrition and metabolism: Glucagon, by its hyperglycaemic effect and GLP-1 via its incretin action, control the carbohydrate metabolism. GLP-2 maintains a healthy morphology of the gut epithelium and its functions, while the glucagon-extended molecules oxyntomodulin and glicentin regulate gastric acid secretion, hydromineral transport across the intestine, gut motility and food intake. Miniglucagon, which derives from a secondary processing of glucagon, plays a strange score in this orchestra, blocking possible unwanted effects of its mother-hormone on insulin secretion, while having insulin-like properties. Since many excellent reviews exist on glucagon itself (see, in particular, the three famous books edited by Lefe`bvre [1–3])

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

4 4 4 5 5 6 6 8 8 9

and on glucagon-like peptide-1 ([4] for a recent review), the present review will focus on the three peptides that contain at least a part of the glucagon sequence, namely glicentin, oxyntomodulin and miniglucagon that have been partially ‘forgotten’ by the researchers in the last years, in spite of their importance in the metabolic regulation. The glucagon-containing peptides derive from tissue specific post-translational processing (Fig. 2). Expression of the prohormone convertase PC-1/3 in intestinal L cells leads to the production of the C-terminally extended peptides glicentin and oxyntomodulin. Expression of the prohormone convertase PC-2 in the a-cells of the islets of Langerhans leads to the production of glucagon. A secondary processing due to the presence of NRD convertase and aminopeptidaseB allows the production of a small, yet biologically significant, proportion of the C-terminal undecapeptide miniglucagon. Every peptide of the family is a true hormone, reaching its remote target(s) through the blood flow, with the exception of miniglucagon, a local regulator acting at the close vicinity of its site of production and quickly degraded.

Fig. 1 – The glucagon-containing peptides and its derivatives. Besides the four peptides with proved natural existence, are added the two oxyntomodulin fragments with demonstrated biological activities.

diabetes research and clinical practice 106 (2014) 1–10

3

Fig. 2 – Tissue-specific post-translational processing of proglucagon into the glucagon-containing peptides. GRPP: glicentinrelated pancreatic polypeptide; SP-1 and SP-2: spacer peptides 1- & -2; GLP-1 et GLP-2: glucagon-like peptide-1 & -2; PC1/3: prohormone convertase-1/3; PC2: prohormone convertase-2; CPE: carboxypeptidase-E; NDRc: N-arginine dibasic convertase; APB: aminopeptidase-B; KR: lysine–arginine cleavage site: RR: arginine–arginine cleavage site; RK: arginine– lysine cleavage site; R: arginine cleavage site.

2. Glucagon-containing C-terminally elongated peptides 2.1.

Structures of oxyntomodulin and glicentin

Oxyntomodulin (OXM) differs from glucagon (see Fig. 1) by an additional C-terminal octapeptide [5,6], while glicentin is Nterminally elongated, as compared to OXM, by the 32-amino acid (32-AA) glicentin-related polypeptide (GRPP). Although much less is known about glicentin as compared to OXM, both peptides share most, if not all, of their biological activities. The two peptides are produced from proglucagon in cells, such as the intestinal L cells or some specialized neurones in the central nervous system (C.N.S.), which express the prohormone convertase PC1/3 rather than PC2, the expression of which in pancreatic a-cells leads to the glucagon production. During the same process, GLP-1 and GLP-2 are produced and co-secreted with OXM and glicentin from intestinal L cells.

2.2.

Compared activities of OXM and glucagon

Since the single difference between OXM and glucagon is the C-terminal octapeptide, it was of considerable interest to compare the biological activities of the two peptides.

2.2.1.

Gastric acid secretion

The inhibitory effect of OXM on pentagastrin-stimulated gastric acid secretion in the rat was contemporary with determination of its structure [7]. OXM displayed in that model a potency ca 15-fold higher than that of glucagon, showing the

importance of the octapeptide in the biological specificity of OXM. It was shown later that OXM also inhibited histaminestimulated gastric acid secretion [8,9]. Interestingly, the Cterminal (19–37) fragment displayed a roughly two-fold lower potency as compared to the entire molecule. When corrected for comparative clearance rate, both the 37-AA OXM and its Cterminal 19-AA fragment have the same potency, indicating that, on that parameter, the N-terminal fragment of glucagon does not enter into the OXM mode of action on gastric acid secretion. Although much less potent, mostly due to its very short half-life, the C-terminal octapeptide on its own, contains the inhibitor message in both the rat [10] and in the human species [11]. The minimal fully active structure was the Nterminally acetylated hexapeptide (octapeptide lacking the last two aminoacids) [12].

2.2.2.

Other sites of action in the GI tract

Besides its gastric target, OXM is able to modulate the hydromineral transport through the rat small intestine [13]. Its mode of action seems to be the same as, or at least similar to that present in the stomach, since the C-terminal [19–37] fragment mimics the effects of the whole molecule, at least in the jejunum. The same holds true in gastric smooth muscles [14]. The receptor(s) implicated in that model was shown to be linked, via a Gi or Go protein, to phospholipase C (production of inositol triphosphate -IP3-) and inhibition of adenylate cyclase (decreased cyclic AMP production). OXM and glicentin were also shown to inhibit the motility pattern of the small intestine [15], while it was shown that glicentin (OXM was not tested in that study) is able to induce contraction of smooth muscle cells from the human colon and that this effect is abolished by

4

diabetes research and clinical practice 106 (2014) 1–10

the GLP-1 receptor antagonist exendin (9–39) [16]. See the chapter on the mode of action of OXM and glicentin for more details.

2.2.3.

Sites of action in the CNS

The major effect of OXM observed on the central nervous system (CNS) is a negative control of food intake (suppression of hunger -anorexigenic action-). Indeed, OXM inhibits food intake in the rat, either applied centrally [17] or via a peripheral administration [18]. Similar findings were obtained in humans [19], with a reduction of body weight in overweight and obese patients using the subcutaneous route [20]. In addition to its action on energy intake, OXM increases energy expenditure [21]. The effect of peripherally administered OXM on food intake is due to the fact that, like other gastro-intestinal hormones, the peptide is able to cross the blood–brain barrier at the level of the median eminence and reaches the arcuate nucleus of hypothalamus which contains both stimulatory and inhibitory neurons. OXM also inhibits the orexigenic signal carried by ghrelin [23]. Altogether, these features make OXM a good basis for the development of an anti-obesity drug [22]. For recent reviews on appetite regulation see [23,24].

2.3. Questions about the mode(s) of action of OXM and glicentin It is clear that the receptor(s), the precise nature of which is still unknown, which transduce(s) the OXM actions on the GI tract displays a strong preference towards the C-terminal end (including the octapeptide itself) of the molecule, with little or no importance of the N-terminal half common with glucagon and which resembles that of GLP-1. This part of the molecules (glucagon or GLP-1) is crucial for triggering the cyclic AMP pathway associated with classical Gs protein-linked glucagon and GLP-1 receptors. In sharp contrast, the recognition system responsible for its CNS actions seems to be, at least in part, transduced by the GLP-1 receptor. Indeed, OXM has no longer effect on food intake in mice lacking the GLP-1 receptor [25] and the (not entirely specific) GLP-1 antagonist exendin (9–39) blocks the actions of both OXM and GLP-1 [25].

However, the figure is much more complex, since (a) OXM and GLP-1 are equipotent in their central effects despite a large difference between their respective affinities for the GLP-1 receptor; (b) OXM and GLP-1 behave differently in vivo and seem to act at different places of the CNS, namely primarily arcuate nucleus and brain stem, respectively [25,18]. Thus, whether these confusing observations are related to the existence of a distinct, still unrecognized, receptor or that of sub-classes of GLP-1 receptors with different specificities remains an open question. More recently, however, it appears that the glucagon receptor is also involved in the body-lowering effect of OXM [26]. A likely conclusion is thus that OXM acts on the CNS via two pathways using both GLP-1 and glucagon receptors.

3.

Glucagon–insulin relationship

Although the focus of the present review is not glucagon itself, it is still necessary to briefly recall basic informations on the impact of the relationship of this peptide with insulin, inasmuch as these data are necessary to understand the biological role of miniglucagon. Glucagon represents, in the regulation of metabolism and particularly that of the carbohydrates, the opposite counterpart of insulin. Both peptides are produced in the islets of Langerhans and display complex local relationships. While insulin is secreted after a meal and allows to store fuels for future usage, glucagon is secreted during the interprandial state or when a particular need for energy-producing molecules (mostly glucose and lipids) occurs (e.g. physical exercise). By its effects on the liver and adipose tissue, it liberates the stored fuels and makes them available to the whole body. This catabolic/anabolic hormonal balance is of major importance for the metabolic regulation. If the details on this balance in the whole organism has been the subject of a huge amount of studies since the discovery of glucagon and, accordingly, not much is probably still to be discovered, the relationship between the two antagonistic peptides was still unclear inside the islet of Langerhans, the place of synthesis and secretion of both peptides.

3.1. Glucagon–insulin relationship in the islets of Langerhans 3.1.1.

Fig. 3 – Architecture of the islets of Langerhans. After Orci et al. [27,28].

The observations

In contrast to the clearly opposite nature of glucagon towards insulin recalled above, the relationship between the two hormones inside their production site, the islets of Langerhans, are much more complex. This is possible thanks to the particular architecture of the islets in which many direct contacts between the a- and the b-cells exist. Indeed, 40–50% of the b-cells have direct contacts with a-cells, according to the precise schematic drawings made by Lelio Orci and colleagues (Fig. 3). In the same line, the human islets have particular structure ‘‘allowing all endocrine cells to be adjacent to blood vessels and favoring heterologous contacts between a- and bcells . . .’’ [29]. See also [30] for a pathological approach of paracrinology inside the islets.

diabetes research and clinical practice 106 (2014) 1–10

The close vicinity between the two types of cells allows mutual interactions. If the direct inhibition of glucagon secretion by insulin is well documented [31], many data indicate that glucagon displays important effects on the b-cell. This is possible thanks to the presence of authentic glucagon receptors at the surface of the insulin-secreting cells [32–34]. These receptors are coupled to adenylate cyclase and allow glucagon to control beta cells functions as follows: (a) Glucagon is required for early differentiation of insulinsecreting cells during the mouse development [35]: Addition of a proglucagon antisense to pancreas from 11-week mouse embryo not only blocks differentiation of a-cells but also that of b-cells and that this differentiation may be rescued by addition of glucagon. Also, ablation of the glucagon receptor gene produces alteration in islet development and differentiation [36]. This fits well with the fact that a-cell is the first islet cell to appear during development [37] and thus precedes the b-cell. (b) Rat islets from dorsal glucagon-rich pancreas have a better insulin response than islets taken from the ventral lobe [38]. (c) Purified b-cells separated from a cells have a poor secretory response to glucose [39]. (d) Glucose competence may be restored by adding nM concentrations of glucagon [40]. (e) Addition of a glucagon receptor antagonist in human islets not only blunts the glucagon-induced but also the glucoseinduced insulin release [34]. The conclusion [34] is that ‘‘the exquisite responsiveness of intact islets is not only dependent on the process of glucose recognition, but also on the presence of a certain threshold of intra-islet glucagon concentration which results in activation of glucagon receptors on b cells’’. It is interesting to note that, if GLP-1 regulates insulin release via its incretin effect, it does not seem to share with glucagon its action on glucose competence, since this parameter is preserved in islets with disrupted GLP-1 receptors [41].

3.1.2.

The molecular mechanisms

The control by glucagon of both b-cell differentiation and the ability of the same cells to respond to glucose (concept of glucose competence) relies on intracellular pathways that have been analyzed in details. A key role in those pathways is played by the extracellularsignal regulated kinases 1 and 2 (ERK1/2) also known as Mitogen-Activated Protein Kinases 1 and 2 (P44/42 MAPKinases). These kinases are activated by double phosphorylation (on a tyrosine and a threonine residues) by the MAPkinase–kinases MEK1/2. The cascade may be triggered either by activation of tyrosine-kinase receptors (such as those for insulin, IGF or EGF) or by activation of G protein-coupled receptors (such as the one for glucagon) via production of cyclic AMP which may act at two different steps of the pathway, namely the small GTPases Rap and P21-Ras or the kinase–kinase–kinases Raf which exists under three different forms (Raf-1, A-Raf, B-raf). A sophistication (which may turn into pathology) of this system is that an excess of PKA

5

activation by cyclic AMP may phosphorylate Raf-1 isoform in an inadequate manner decreasing the activity of the pathway instead of increasing it. The ERK pathway exists in the b-cell as shown by the effect of low glucose in the absence and in the presence of glucagon on ERK activation by double phosphorylation in the MIN6 bcell line [42]. At a low glucose concentration, which corresponds to the physiological situation of glucagon secretion, glucagon stimulates ERK phosphorylation within 10 min by a 10-fold factor. Interestingly, glucagon is able to stimulate ERK even in the absence of added glucose during the experiment [42]. A major target for ERK is the transcription factor CREB (cyclic AMP response element binding protein) which controls expression of genes implicated in many cell functions. Indeed, CREB phosphorylation by glucagon is suppressed by addition of a MEK (MAP kinase–kinase) inhibitor. Unexpectedly, GLP-1, which, similarly to glucagon, stimulates cyclic AMP production, stimulates PKA and phosphorylates ERK, appears to skip the ERK step: Acute GLP-1 stimulation phosphorylates CREB in a non ERK-dependent manner, since the MEK inhibitor has no effect on CREB phosphorylation by GLP-1, while prolonged GLP-1 stimulation phosphorylates CREB in a b-arrestin-1-ERK-dependent manner. The pathways used by glucagon, glucose and GLP-1 in phosphorylating CREB in the b-cell are different. Only glucagon uses a straight pathway (glucagon ! cAMP ! PKA ! ERK ! CREB), while glucose uses increased intracellular calcium concentrations and GLP-1 uses a b-arrestin-1-ERK-dependent manner. This might be related to the different timing of actions of the three regulators, glucagon having plenty of time (the interprandial state) to produce in-depth regulation of CREB-dependent mechanisms, and, thus ‘preparing’ the ß-cell to respond to glucose, as compared to GLP-1 which acts, as an incretin, shortly before glucose reaches the islets. Fig. 4 outlines the central role of ERK in the b-cell biology, particularly in transmission of the glucagon actions: at the level of the nucleus, ERK is directly implicated in activation by phosphorylation of CREB, which controls expression of major genes such as that of proinsulin and that of the anti-apoptotic molecule Bcl-2. Another role of ERK in the b-cell is to phosphorylate proteins participating in insulin secretory granule exocytosis, favouring the secretory action of glucose [43]. In the cytoplasm, glucose-activated ERK phosphorylates proteins which have been proposed to be implicated in insulin granule exocytosis, such as Fak, paxillin, and synapsin-1 [43– 45]. ERK activated by glucose accumulates in the cytoplasm at actin filament tips adjacent to the plasma membrane, indicating that these are the main sites of action for these kinases during insulin secretion [46].

3.2.

Glucagon and insulin secretion

The beneficial effect of glucagon on the b-cell asks a question about its potential direct role in regulating insulin secretion. Indeed, while the maintenance of glucose competence of the insulin cells by glucagon is obviously beneficial, the eventuality of a direct effect of the peptide on insulin exocytosis would ruin the necessary hyperglycaemic action of glucagon. Fortunately, if exogenous glucagon is known for long to

6

diabetes research and clinical practice 106 (2014) 1–10

Fig. 4 – Central role of ERK in b-cell regulation by glucagon and insulin. Synthesis from [42–46].

trigger insulin secretion [47,32], endogenous (interstitial) glucagon is unable by itself to do so [48]. Indeed, acute addition of a glucagon receptor antagonist in the rat perfused pancreas does not modify the ability of glucose to stimulate insulin secretion. Furthermore, under hypoglycaemic conditions where glucagon is physiologically secreted, endogenous secretion of glucagon by isoproterenol does not stimulate insulin secretion [48]. Thus, while glucagon acts on the b-cell via its own receptors to maintain the capacity of those cells to fully respond to glucose, glucagon secreted from the a-cells under hypoglycaemic conditions does not trigger insulin secretion from the neighbouring b-cells. The explanation for this discrepancy is the presence together with glucagon in the secretory granules of a-cells of a powerful inhibitor of insulin secretion.

3.3.

Miniglucagon

3.3.1.

Miniglucagon in the islet of Langerhans

Miniglucagon is the product of a secondary processing of glucagon, due to an original enzymatic system, different from the classical duo [49–51] prohormone-convertase (PC) + carboxypeptidase-E (CPE). This system [52,53], which may be considered as the opposite (cleavage at the N-terminus of basic residues followed by elimination of the remaining basic aminoacids by aminopeptidase) of the PC + CPE system (cleavage at the C-terminus of basic doublet by PC followed by elimination of the remaining basic aminoacids by carboxypeptidase), is made of N-arginine dibasic convertase (NRDc) + aminopeptidase-B. It was shown to catalyze, at least in some tissues, the cut of somatostatin-28 into somatostatin14 and is responsible for the production of miniglucagon from glucagon [54]. About 5% of the glucagon stores present in the a granules are transformed into miniglucagon [55]. This relatively small proportion is more than compensated by the huge (ca 104) difference in the respective potencies of the peptides.

Miniglucagon was shown to inhibit, at sub-picomolar concentrations, glucose-, sulfonylurea-, glucagon- and GLP1-stimulated insulin secretion from the MIN6 b-cell line [56] and from the rat isolated perfused pancreas [55]. The inhibitory effect of miniglucagon is due to blockage of calcium entry, necessary for insulin secretion stimulated by these agents [56]. Whole cell patch-clamp experiments [54] indicated that the inhibitory action of miniglucagon on calcium entry was linked to repolarization of the membrane (see Fig. 5) which forces the voltage-dependent calcium channels (VDCC) to close, and not to a direct action of the peptide on the VDCC (see Fig. 6), in contrast to another peptide, a-endosulfine [57]. Indeed, 10 mM glucose induces a depolarization from the resting potential (ca 70 mV) up to the potential (ca 40 mV) of activation of the VDCC (Fig. 5 upper panel) and addition of miniglucagon to a depolarized cell induces a repolarization up to the resting potential, suppressing the calcium entry and, consecutively, insulin secretion (Fig. 5 lower panel). As shown

Fig. 5 – Repolarization by miniglucagon of a MIN6 cell depolarized by 10 mM glucose. Data from [56].

diabetes research and clinical practice 106 (2014) 1–10

7

Fig. 6 – Calcium currents at different degrees of depolarization: S40, S30, S20, S10, 0 mV (upper traces), +10, +20, +30, +40 mV, lower traces, in the absence (control) and in the presence of miniglucagon. In contrast to a-endosulfine, miniglucagon does not act directly on the voltage-dependent calcium channels. Data from [56,57] for the inset.

in Fig. 6, when the polarity of the plasma membrane is set to various values from 40 to +40 mV, the calcium currents are not modified by miniglucagon, in contrast to a-endosulfine. Altogether, the data indicate that miniglucagon closes indirectly the VDCC by modifying the membrane polarity, most probably by opening a pertussis toxin-sensitive Gi or Go protein-linked potassium channel (see [58] for a review on those channels), since the miniglucagon effect is suppressed by a treatment with pertussis toxin [56].

The fact that miniglucagon suppresses any insulinosecretagogue effect of glucagon raises the possibility that it also suppresses the beneficial effect of glucagon on the glucose competence of the b-cell. Fortunately, this is not the case [59]. Indeed, since Miniglucagon does not interfere with the ability of glucagon to stimulate cyclic AMP production, it does not interfere either with activation by glucagon of ERK and CREB which rely on the cyclic AMP/PKA pathway. It is interesting to note that somatostatin, which also suppresses the ability of

Fig. 7 – a-Cell–b-cell relationship inside the islet of Langerhans.

8

diabetes research and clinical practice 106 (2014) 1–10

Fig. 8 – Pathways used by insulin and miniglucagon in triggering Glut4 translocation in the adipose tissue. From [64].

glucagon to stimulate insulin secretion, does so by inhibiting adenylate cyclase via a Gi protein. This is an indirect indication that the miniglucagon receptor, the nature of which is unknown, is probably linked to the potassium channel via a Go rather than a Gi protein. In conclusion (Fig. 7), miniglucagon co-released with glucagon suppresses any prejudicial effect of its motherhormone on the b-cell, leaving untouched its beneficial effect: Glucagon, secreted during the interprandial state, maintains the b-cell in a glucose-competent condition via the cyclic AMP/ PKA/ERK/CREB pathway. Co-secreted miniglucagon does not modify this glucagon action, since it is disconnected from that pathway, but, via opening of a G protein-regulated potassium channel, drives the membrane via repolarization to the resting potential, preventing opening of the VDCC and thus insulin secretion. The single situation in which insulin raises in blood together with a significant glucagon (and thus miniglucagon) secretion is shortly after the beginning of a meal. The explanation for such an apparent contradiction is that the early insulin secretion is triggered essentially by vagal stimulation (cephalic phase) [60] which liberates Ca2+ from intracellular stores. Since the miniglucagon inhibitory action is solely mediated by closure of VDCC, it does not interfere with intracellular Ca2+ release and, thus, not with this early phase of insulin secretion.

3.3.2.

Miniglucagon in the whole organism

The data about the relationship between a- and b-cells prompted us to study a possible effect of miniglucagon in vivo, in spite of its extremely short half-life. The data obtained [61–63] indicated that, in the conscious rat, injection of miniglucagon together with a bolus of glucose reduced the insulin by about 40% at 3 min with no change in variations of glycaemia. This suggests either that miniglucagon facilitates the insulin action (potentiation) or that miniglucagon displays,

by itself, an insulin-like effect. Experiments conducted on the 3T3-L1 adipocytes showed that miniglucagon displays effects on Glut4 translocation similarly to insulin and displays insulin-like activity on the same parameter when used in the absence of added insulin. Analysis of its mode of action (Fig. 8) indicates that the miniglucagon receptor forms a scaffold with the insulin receptor and IRS-2 and, when activated, phosphorylates a 50-kDa protein which transmits the message to PI3-kinase. The next steps in the pathway are common with that of insulin. The major interest of such an architecture is that, in case of insulin resistance which implicates mostly the first steps (e.g. Ser/Thr phosphorylation which hampers the Tyr phosphorylation necessary for activation), the use of alternative steps by miniglucagon allows a workaround of insulin resistance. The physiological, pathological and pharmacological implications of these discoveries have still to be analyzed in details.

4.

Conclusion

The variety of peptides with different, yet complementary biological activities that derive from a single original molecular motif (glucagon plus its N-terminal and C-terminal extensions present in the gene and consequently in the prohormone), is a particularly rich example of the pivotal role of post-translational processing in the biological regulation. If we add the duplication of this motif during animal evolution (leading to the series of glucagon-like peptides) and the fact that this processing differs from a tissue to another, this is, altogether, not less than six peptides with demonstrated biological activity(ies) that are produced, four of them containing at least a part of glucagon, the original molecular motif. This importance of post-translational processing related to the presence in different tissues of various pro-hormone

diabetes research and clinical practice 106 (2014) 1–10

convertases [65,66] which allows the production of various peptides/proteins with specific roles has also, in some way, been ‘forgotten’, when the focus in biological research was made on genetics, while, although of major importance, the genes are the beginning but not the end of the story.

Conflict of interest The authors declare no conflict of interest.

references

[1] Lefe`bvre PJ, editor. Glucagon I. Handbook experimental pharmacology, 66. 1983. p. 535. I. [2] Lefe`bvre PJ, editor. Glucagon II. Handbook experimental pharmacology, 66. 1983. p. 700. II. [3] Lefebvre PJ, editor. Glucagon III. Handbook experimental pharmacology, 123. 1996. p. 349 p. [4] Cho YM, Fujita Y, Kieffer TJ. Glucagon-like peptide-1: glucose homeostasis and beyond. Annu Rev Physiol 2013;28:262–74. [5] Bataille D, Coudray AM, Carlqvist M, Rosselin G, Mutt V. Isolation of glucagon-37 (bioactive enteroglucagon/ oxyntomodulin) from porcine jejuno-ileum. Isolation of the peptide. FEBS Lett 1982;146:73–8. [6] Bataille D, Tatemoto K, Gespach C, Jo¨rnvall H, Rosselin G, Mutt V. Isolation of glucagon-37 (bioactive enteroglucagon/ oxyntomodulin) from porcine jejuno-ileum. Characterisation of the peptide. FEBS Lett 1982;146:79–86. [7] Dubrasquet M, Bataille D, Gespach C. Oxyntomodulin (glucagon-37 or bioactive enteroglucagon): a potent inhibitor of pentagastrin-stimulated acid secretion in rat. Biosci Rep 1982;2:391–5. [8] Carles-Bonnet C, Jarrousse C, Niel H, Martinez J, Bataille D. Oxyntomodulin and its (19–37) and (30–37) fragments inhibit histamine-stimulated gastric acid secretion in the conscious rat. Eur J Pharmacol 1991;203:245–52. [9] Jarrousse C, Carles-Bonnet C, Niel H, Sabatier R, AudoussetPuech M-P, Blache P, et al. Inhibition of gastric acid secretion by oxyntomodulin and its (19–37) fragment in the conscious rat. Am J Physiol 1993;264:G816–23 (Gastrointest. Liver Physiol. 27). [10] Jarrousse C, Audousset-Puech M-P, Dubrasquet M, Niel H, Martinez J, Bataille D. Oxyntomodulin (glucagon-37) and its C-terminal octapeptide inhibit gastric secretion. FEBS Lett 1985;188:81–4. [11] Veyrac M, Ribard D, Daures JP, Mion H, Le Quellec A, Martinez J, et al. Inhibitory effect of the C-terminal octapeptide of oxyntomodulin on pentagastrin-stimulated gastric acid secretion in man. Scand J Gastroenterol 1989;24:1238–42. [12] Carles-Bonnet C, Martinez J, Jarrousse C, Aumelas A, Niel H, Bataille D. H-Lys-Arg-Asn-Lys-Asn-Asn-OH is the minimal active structure of oxyntomodulin. Peptides 1996;17:557–61. [13] Beauclair F, Eto B, Pansu D, Rodier G, Mochuzoki T, Martinez J, et al. Oxyntomodulin regulates hydromineral transport across the rat small intestine. Dig Dis Sci 1998;43:1814–23. [14] Rodier G, Magous R, Mochizuki T, Le Nguyen D, Martinez J, Bali JP, et al. Glicentin and oxyntomodulin modulate both the phosphoinositide and cyclic adenosine monophosphate signaling pathways in gastric myocytes. Endocrinology 1999;140:22–8.

[15] Pellissier S, Sasaki K, Le-Nguyen D, Bataille D, Jarrousse C. Oxyntomodulin and glicentin are potent inhibitors of the fed motility pattern in small intestine. Neurogastroenterol Motil 2004;16:455–63. [16] Ayachi SE, Borie F, Magous R, Sasaki K, le Nguyen D, Bali JP, et al. Contraction induced by glicentin on smooth muscle cells from the human colon is abolished by exendin (9–39). Neurogastroenterol. Motil 2005;17:302–9. [17] Dakin CL, Gunn I, Small CJ, Edwards CM, Hay DL, Smith DM, et al. Oxyntomodulin inhibits food intake in the rat. Endocrinology 2001;142:4244–50. [18] Dakin CL, Small CJ, Batterham RL, Neary NM, Cohen MA, Patterson M, et al. Peripheral oxyntomodulin reduces food intake and body weight gain in rats. Endocrinology 2004;145:2687–95. [19] Cohen MA, Ellis SM, Le Roux CW, Batterham RL, Park A, Patterson M, et al. Oxyntomodulin suppresses appetite and reduces food intake in humans. J Clin Endocrinol Metab 2003;88:4696–701. [20] Wynne K, Park AJ, Small CJ, Patterson M, Ellis SM, Murphy KG, Wren AM, Frost GS, Meeran K, Ghatei MA, Bloom SR. Subcutaneous oxyntomodulin reduces body weight in overweight and obese subjects: a double-blind, randomized, controlled trial. Diabetes 2005;54:2390–5. [21] Wynne K, Park AJ, Small CJ, Meeran K, Ghatei MA, Frost GS, et al. Oxyntomodulin increases energy expenditure in addition to decreasing energy intake in overweight and obese humans: a randomised controlled trial. Int J Obes 2006;30:1729–36. [22] Druce M, Bloom SR. The regulation of appetite. Arch Dis Child 2006;91:183–7 (Review). [23] Yu JH, Kim MS. Molecular mechanisms of appetite regulation. Diabetes Metab J 2012;36:391–8. [24] Suzuki K, Jayasena CN, Bloom SR. Obesity and appetite control. Exp Diabetes Res 2012;2012:824305. [25] Baggio LL, Huang Q, Brown TJ, Drucker DJ. Oxyntomodulin and glucagon-like peptide-1 differentially regulate murine food intake and energy expenditure. Gastroenterology 2004;127:546–58. [26] Kosinski JR, Hubert J, Carrington PE, Chicchi GG, Mu J, Miller C, et al. The glucagon receptor is involved in mediating the body weight-lowering effects of oxyntomodulin. Obesity 2012;20:1566–71. [27] Orci L, Unger RH. Functional subdivision of islets of Langerhans and possible role of D cells. Lancet 1975;2:1243–4. [28] Orci L, Malaisse-Lagae F, Amherdt M, Ravazzola M, Weisswange A, Dobbs R, et al. Cell contacts in human islets of Langerhans. J Clin Endocrinol Metab 1975;41:841–4. [29] Bosco D, Armanet M, Morel P, Niclauss N, Sgroi A. Unique arrangement of a- and b-cells in human islets of Langerhans. Diabetes 2010;59:1202–10. [30] Lefebvre PJ. Diabetes as a paracrinopathy of the islets of Langerhans. Eur Endocrinol 2011;7:79–83. [31] Lefebvre PJ, Luyckx AS. Glucose and insulin in the regulation of glucagon release from the isolated perfused dog stomach. Endocrinology 1978;103:1579–82. [32] Kawai K, Yokota C, Ohashi S, Watanabe Y, Yamashita K. Evidence that glucagon stimulates insulin secretion through its own receptor in rats. Diabetologia 1995; 38:274–6. [33] Marie JC, Boissard C, Skoglund G, Rosselin G, Breant B. Glucagon acts through its own receptors in the presence of functional glucagon-like peptide-1 receptors on hamster insulinoma. Endocrinology 1996;137:4108–814. [34] Huypens P, Ling Z, Pipeleers D, Schuit F. Glucagon receptors on human islet cells contribute to glucose competence of insulin release. Diabetologia 2000;43:1012–9.

9

10

diabetes research and clinical practice 106 (2014) 1–10

[35] Prasadan K, Daume E, Preuett B, Spilde T, Bhatia A, Kobayashi H, Hembree M, Manna P, Gittes GK. Glucagon is required for early insulin-positive differentiation in the developing mouse pancreas. Diabetes 2002;51:3229–36. [36] Vuguin PM, Kedees MH, Cui L, Guz Y, Gelling RW, Nejathaim M, Charron MJ, Teitelman G. Ablation of the glucagon receptor gene increases fetal lethality and produces alterations in islet development and maturation. Endocrinology 2006;147:3995–4006. [37] Pictet RL, Clark WR, Williams RH, Rutter WJ. An ultrastructural analysis of the developing embryonic pancreas. Dev Biol 1972;29:436–67. [38] Trimble ER, Halban PA, Wollheim CB, Renold AE. Functional differences between rat islets of ventral and dorsal pancreatic origin. J Clin Invest 1982;69:405–13. [39] Pipeleers DG, Schuit FC, in’t Veld PA, Maes E, Hooghe-Peters EL, Van de Winkel M, et al. Interplay of nutrients and hormones in the regulation of insulin release. Endocrinology 1985;117:824–33. [40] Pipeleers DG, in’t Veld PA, Van de Winkel M, Maes E, Schuit FC, Gepts W. A new in vitro model for the study of pancreatic A and B cells. Endocrinology 1985;117: 806–16. [41] Flamez D, Van Breusegem A, Scrocchi LA, Quartier E, Pipeleers D, Drucker DJ, et al. Mouse pancreatic beta-cells exhibit preserved glucose competence after disruption of the glucagon-like peptide-1 receptor gene. Diabetes 1998;47:646–52. [42] Dalle S, Longuet C, Costes S, Broca C, Faruque O, Fonte´s G, et al. Glucagon promotes cAMP-response element-binding protein phosphorylation via activation of ERK1/2 in MIN6 cell line and isolated islets of Langerhans. J Biol Chem 2004;279:20345–5. [43] Longuet C, Broca C, Costes S, Hani EH, Bataille D, Dalle S. Extracellularly regulated kinases 1/2 (p44/42 mitogenactivated protein kinases) phosphorylate synapsin I and regulate insulin secretion in the MIN6 beta-cell line and islets of Langerhans. Endocrinology 2005;146:643–54. [44] Rondas D, Tomas A, Halban PA. Focal adhesion remodeling is crucial for glucose-stimulated insulin secretion and involves activation of focal adhesion kinase and paxillin. Diabetes 2011;60:1146–57. [45] Rondas D, Tomas A, Soto-Ribeiro M, Wehrle-Haller B, Halban PA. Novel mechanistic link between focal adhesion remodeling and glucose-stimulated insulin secretion. J Biol Chem 2012;287:2423–36. [46] Tomas A, Yermen B, Min L, Pessin JE, Halban PA. Regulation of pancreatic beta-cell insulin secretion by actin cytoskeleton remodelling: role of gelsolin and cooperation with the MAPK signalling pathway. J Cell Sci 2006;119: 2156–67. [47] Samols DR. Glucagon and insulin secretion. In: Lefe`bvre PJ, editor. Glucagon I handbook of experimental pharmacology. Berlin Heidelberg, New York: Springer; 1983. p. 485–518. [48] Moens K, Berger V, Ahn JM, Van Schravendijk C, Hruby VJ, Pipeleers D, Schuit F. Assessment of the role of interstitial glucagon in the acute glucose secretory responsiveness of in situ pancreatic beta-cells. Diabetes 2002;51:669–75. [49] Chre´tien M, Gasper L, Benjannet S, Mbikay M, Lazure C, Seidah NG. The Jeremiah Metzger lecture. From POMC to functional diversity of neural peptides: the key importance of convertases. Trans Am Clin Climatol Assoc 1990;102:195– 224 (Review).

[50] Seidah NG, Day R, Marcinkiewicz, Chre´tien M. Precursor convertases: an evolutionary ancient, cell-specific, combinatorial mechanism yielding diverse bioactive peptides and proteins. Ann NY Acad Sci 1998;839:9–24. [51] Steiner DF. The proprotein convertases. Curr Opin Chem Biol 1998;2:31–9. [52] Chesneau V, Pierotti AR, Prat A, Gaudoux F, Foulon T, Cohen P. N-arginine dibasic convertase (NRD convertase): a newcomer to the family of processing endopeptidases. An overview. Biochimie 1994;76:234–40 (Review). [53] Cadel S, Foulon T, Viron A, Balogh A, Midol-Monet S, Noel N, Cohen P. Aminopeptidase B from the rat testis is a bifunctional enzyme structurally related to leukotriene-A4 hydrolase. Proc Nat Acad Sci USA 1997;94:2963–8. [54] Fonte´s G, Lajoix A-D, Bergeron F, Cadel S, Prat A, Foulon T, et al. Miniglucagon-generating endopeptidase, which processes glucagon into miniglucagon, is composed of NRD-convertase and aminopeptidase-B. Endocrinology 2005;146:702–12. [55] Dalle S, Fonte´s G, Lajoix A-D, Le Brigand L, Gross R, Ribes G, Dufour M, Barry L, Le-Nguyen D, Bataille D. Miniglucagon (glucagon 19–29): a novel regulator of the pancreatic islet physiology. Diabetes 2002;51:406–12. [56] Dalle S, Smith P, Blache P, Le-Nguyen D, Le Brigand L, Bergeron F, Ashcroft FM, Bataille D. Miniglucagon (glucagon 19–29): a potent and efficient inhibitor of secretagogueinduced insulin release through a Ca2+ pathway. J Biol Chem 1999;274:10869–76. [57] Virsolvy A, Smith P, Bertrand G, Gros L, He´ron L, Salazar G, Puech R, Bataille D. Block of pancreatic ß-cells Ca2+channels by a-endosulfine inhibits insulin release. Br J Pharmacol 2002;135:1810–8. [58] Yamada M, Inanobe A, Kurachi Y. G protein regulation of potassium ion channels. Pharmacol Rev 1998;50:723–60 (Review). [59] Dalle S, Longuet C, Costes S, Broca C, Faruque O, Fonte´s G, Hani E-H, Bataille D. Glucagon promotes CREB phosphorylation via activation of ERK1/2 (extracellular signal-related kinase 1/2) in MIN6 cell line and isolated islets of Langerhans. J Biol Chem 2004;279:20345–5. [60] Power ML, Schulkin J. Anticipatory physiological regulation in feeding biology: cephalic phase responses 2008;50:194–206. [61] Fonte´s G, Imamura T, Ilic C, Puech C, Ktorza A, Bataille D, et al. Miniglucagon: an unexpected insulin’s partner in the peripheral effects of the hypoglycemic hormone.In: The 40th meeting of the European Association for the Study of Diabetes (EASD); Diabetologia 2004;47(S1):A91. [62] Bataille D, Fonte´s G, Costes S, Longuet C, Imamura T, Ilic C.. et al. The glucagon/miniglucagon interplay: a new level in the metabolic regulation. In: The VIIth international symposium on VIP, PACAP and related peptides; 2005. [63] Bataille D, Fonte´s G, Costes S, Longuet C, Dalle S. The glucagon-miniglucagon interplay: a new level in the metabolic regulation. Ann NY Acad Sci 2006;1070:161–6. [64] Bataille D. Proglucagon-derived peptides, metabolic regulation and diabetes: the yin, the yang and beyond. In: The 13th diabetes and endocrine conference; 2007. [65] Mbikay M, Seidah NG, editors. Proprotein convertases. Methods in molecular biology, vol 768. New York Dordrecht Heidelberg London: Humana Press Springer; 2011. [66] Seidah NG, Sadr MS, Chre´tien M, Mbikay M. The multifaceted proprotein convertases: their unique, redundant, complementary, and opposite functions. J Biol Chem 2013;288:21473–81.

The forgotten members of the glucagon family.

From proglucagon, at least six final biologically active peptides are produced by tissue-specific post-translational processing. While glucagon and GL...
2MB Sizes 0 Downloads 5 Views