The pancreas is an exocrine and endocrine organ

The pancreas has key roles in the regulation of macronutrient digestion and hence metabolism/energy homeostasis by releasing various digestive enzymes and pancreatic hormones. It is located behind the stomach within the left upper abdominal cavity and is partitioned into head, body and tail. The majority of this secretory organ consists of acinar—or exocrine—cells that secrete the pancreatic juice containing digestive enzymes, such as amylase, pancreatic lipase and trypsinogen, into the ducts, that is, the main pancreatic and the accessory pancreatic duct. In contrast, pancreatic hormones are released in an endocrine manner, that is, direct secretion into the blood stream. The endocrine cells are clustered together, thereby forming the so-called islets of Langerhans, which are small, island-like structures within the exocrine pancreatic tissue that account for only 1–2% of the entire organ (Figure 1).1 There are five different cell types releasing various hormones from the endocrine system: glucagon-producing α-cells,2 which represent 15–20% of the total islet cells; amylin-, C-peptide- and insulin-producing β-cells,2 which account for 65–80% of the total cells; pancreatic polypeptide (PP)-producing γ-cells,3 which comprise 3–5% of the total islet cells; somatostatin-producing δ-cells,2 which constitute 3–10% of the total cells; and ghrelin-producing ɛ-cells,4 which comprise <1% of the total islet cells. Each of the hormones has distinct functions. Glucagon increases blood glucose levels, whereas insulin decreases them.5 Somatostatin inhibits both, glucagon and insulin release,6 whereas PP regulates the exocrine and endocrine secretion activity of the pancreas.3, 7 Altogether, these hormones regulate glucose homeostasis in vertebrates, as described in more detail below. Although the islets have a similar cellular composition among different species, that is, human, rat and mouse, their cytoarchitecture differs greatly. Although islets in rodents are primarily composed of β-cells located in the center with other cell types in the periphery, human islets exhibit interconnected α- and β-cells.2, 8

Figure 1
figure 1

Anatomical organization of the pancreas. The exocrine function of the pancreas is mediated by acinar cells that secrete digestive enzymes into the upper small intestine via the pancreatic duct. Its endocrine function involves the secretion of various hormones from different cell types within the pancreatic islets of Langerhans. The micrograph shows the pancreatic islets. LM × 760 (Micrograph provided by the Regents of University of Michigan Medical School © 2012). Adapted from Human Anatomy and Physiology, an OpenStax College resource.404

Through its various hormones, particularly glucagon and insulin, the pancreas maintains blood glucose levels within a very narrow range of 4–6 mM. This preservation is accomplished by the opposing and balanced actions of glucagon and insulin, referred to as glucose homeostasis. During sleep or in between meals, when blood glucose levels are low, glucagon is released from α-cells to promote hepatic glycogenolysis. In addition, glucagon drives hepatic and renal gluconeogenesis to increase endogenous blood glucose levels9 during prolonged fasting. In contrast, insulin secretion from β-cells is stimulated by elevated exogenous glucose levels, such as those occurring after a meal.10 After docking to its receptor on muscle and adipose tissue, insulin enables the insulin-dependent uptake of glucose into these tissues and hence lowers blood glucose levels by removing the exogenous glucose from the blood stream (Figure 2).11, 12, 13 Furthermore, insulin promotes glycogenesis,14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26 lipogenesis27, 28 and the incorporation of amino acids into proteins;29 thus, it is an anabolic hormone, in contrast to the catabolic activity of glucagon.

Figure 2
figure 2

Maintenance of blood glucose levels by glucagon and insulin. When blood glucose levels are low, the pancreas secretes glucagon, which increases endogenous blood glucose levels through glycogenolysis. After a meal, when exogenous blood glucose levels are high, insulin is released to trigger glucose uptake into insulin-dependent muscle and adipose tissues as well as to promote glycogenesis.

The insulin secretion signaling pathway

Endocrine cells secrete their respective hormones in response to external signals, such as nutrient intake or stress, via humoral, neural or hormonal signaling pathways. The underlying molecular process that translates the stimulus into the actual hormone release is called stimulus-secretion coupling which is known as the stimulus-dependent exocytosis of a particular substance, such as glucose-stimulated β-cell insulin release.30

In β-cells, the main stimulus for insulin release are elevated blood glucose levels following a meal.10 The circulating blood glucose is taken up by the facilitative glucose transporter GLUT2 (SLC2A2), which is located on the surface of the β-cells. Once inside the cell, glucose undergoes glycolysis, thereby generating adenosine triphosphate (ATP), resulting in an increased ATP/ADP ratio. This altered ratio then leads to the closure of ATP-sensitive K+-channels (KATP-channels). Under non-stimulated conditions, these channels are open to ensure the maintenance of the resting potential by transporting positively charged K+-ions down their concentration gradient out of the cell. Upon closure, the subsequent decrease in the magnitude of the outwardly directed K+-current elicits the depolarization of the membrane, followed by the opening of voltage-dependent Ca+-channels (VDCCs). The increase in intracellular calcium concentrations eventually triggers the fusion of insulin-containing granules with the membrane and the subsequent release of their content.31 The whole secretory process is biphasic with the first phase peaking around 5 minutes after the glucose stimulus with the majority of insulin being released during this first phase. In the second, somewhat slower, phase, the remaining insulin is secreted.32, 33, 34 Insulin is stored in large dense-core vesicles that are recruited to the proximity of the plasma membrane following stimulation such that insulin is readily available.35, 36 The key molecules that mediate the fusion of the insulin-containing large dense-core vesicles are the synaptosomal-associated protein of 25 kDa (SNAP-25), syntaxin-1 and synaptobrevin 2 (or vesicle-associated membrane protein VAMP2), all of which belong to the superfamily of the soluble N-ethylmaleimide-sensitive factor attachment protein (SNAP) receptor proteins (SNAREs). Together with the Sec1/Munc18-like (SM) proteins they form the so-called SNARE complex.37 To initiate fusion, synaptobrevin 2, a vesicle (v-)SNARE that is integrated into the vesicle’s membrane, fuses with the target (t-)SNAREs syntaxin-1 and SNAP-25, which are located in the target cell membrane,38, 39 with mammalian uncoordinated (munc)-18 playing a key regulatory role (Figure 3).40, 41

Figure 3
figure 3

Glucose-stimulated insulin release from a pancreatic β-cell. Exogenous glucose is taken up by GLUT2 and undergoes glycolysis inside the cell. Elevated adenosine triphosphate (ATP) levels alter the ATP/ADP ratio, which in turn leads to the closure of ATP-sensitive K+-channels. The subsequent membrane depolarization opens voltage-dependent Ca2+-channels in response to increasing intracellular calcium levels, which eventually trigger insulin secretion following vesicle fusion with the membrane.

To date, numerous SNARE isoforms, including syntaxin-1, -3 and -4, SNAP-25 and -23, as well as syntaptobrevins 2 and 3 (VAMP2 and 3), have been shown to be involved in glucose-stimulated insulin secretion,42, 43, 44, 45, 46 whereas VAMP8, a non-essential SNARE protein for glucose-stimulated insulin secretion, has a role in the regulation of the glucagon-like peptide-1-potentiated insulin secretion.47 In addition to SNARE and SM proteins, a calcium sensor is required for the initiation of membrane fusion. Synaptotagmins, which are highly expressed in neurons and endocrine cells, were shown to participate in Ca2+-dependent exocytosis processes. To date, 17 synaptotagmins (Syts 1–17) have been identified and only eight of them, namely Syt-1, -2, -3, -5, -6, -7, -9 and -10, are able to bind calcium.48 Following Ca2+-binding, synaptotagmins form a complex with the SNAREs to facilitate and trigger the vesicle-membrane fusion process. Among the synaptotagmin family, Syt-3, -5, -7, -8 and -9 are implicated in insulin exocytosis.49, 50, 51, 52

External factors affecting pancreatic hormone secretion

Metabolism–cAMP coupling

The glucose-triggered stimulus-secretion coupling is an established paradigm of insulin secretion from β-cells and includes a great variety of modulators that trigger, potentiate or inhibit glucose-stimulated insulin secretion, primarily through G-protein-coupled receptors (GPCRs). The most traditional external factor that initiates insulin secretion is glucose. In addition to its trigger function, glucose also induces pathways that amplify insulin secretion through metabolism-cAMP (cyclic adenosine monophosphate) coupling or the incretin hormones glucagon-like peptide (GLP)-1 and glucose-dependent insulinotropic peptide (GIP).31 Metabolism–cAMP coupling refers to the signaling cascade that occurs after the conversion of ATP, which is generated during intracellular glucose metabolism, into cAMP by adenylate cyclase (AC),53 which in turn activates protein kinase A (PKA)54 and cAMP-regulated guanine nucleotide exchange factors, also referred to as exchange protein directly activated by cAMP (Epac) 2.55, 56 Although Epac2 activation amplifies insulin secretion by mobilizing calcium from internal stores to increase Ca2+ levels57, 58 and by controlling the granule density in proximity to the plasma membrane,59 activated PKA exerts its effects by modulating KATP-channel60, 61 and calcium channel62, 63 activity through phosphorylation, thereby enhancing the number of highly Ca2+-sensitive insulin-containing granules64 and the probability of releasing secretory vesicles from the readily releasable pool,65 respectively.

The incretins GLP-1 and GIP

The gut-derived hormones GLP-1 and GIP, which are secreted from enteroendocrine L-cells66 and K-cells,67 respectively, upon glucose,66, 68 fructose,69 amino acid70 and free fatty acid (FFA)71, 72 ingestion, also potentiate insulin release through the so-called incretin effect. This effect describes the observation that orally, but not intravenously, administered glucose enhances insulin secretion by triggering GLP-1 and GIP secretion;73, 74, 75 the resulting potentiation of insulin secretion may account for up to 50% of the total release. The underlying mechanism includes GLP-1 and GIP binding to their GPCRs (GLP-1R and GIPR), both of which are expressed in pancreatic β-cells.76 The binding induces a conformational change in the receptors’ structure, followed by the exchange of guanosine diphosphate for guanosine triphosphate and the subsequent dissociation of the Gsα-subunit from the receptors. This subunit, in turn, activates adenylate cyclase to convert ATP into cAMP, thereby stimulating the cAMP signaling pathway described above.77, 78, 79, 80, 81, 82 Furthermore, GLP-1 increases intracellular calcium concentrations by mobilizing Ca2+ from ryanodine-sensitive stores83, 84 or, similar to GIP, by acting on voltage-dependent Ca2+-channels,85 thereby potentiating insulin release.85, 86, 87 Recent studies have also shown that GLP-1R agonists, such as exendin-488, induce the PKA-mediated phosphorylation of Snapin or Synaptotagmin-7, which in turn enhances GSIS by Snapin interacting with SNAP-2589 or by directly enhancing glucose- and Ca2+-triggered insulin release.90

Free Fatty Acids

FFAs not only stimulate incretin secretion but are also known to modulate insulin release through fatty acid metabolism. Although long-chain FFAs augment insulin secretion, short-chain FFAs inhibit it. The binding and subsequent interaction of long-chain FFAs with the G-protein-coupled free fatty acid receptor (FFAR) 1 in the pancreatic β-cells leads to the activation of phospholipase C. PLC then hydrolyzes phosphatidylinositol-4,5-bisphosphate (PIP2) to diacylglycerol and inositol-1,4,5-triphosphate (IP3), with the latter docking on a calcium channel in the endoplasmic reticulum. The subsequent release of Ca2+ into the cytosol increases the intracellular Ca2+ concentration, which eventually triggers insulin secretion.91, 92, 93, 94 In contrast, short-chain FFAs inhibit glucose-stimulated insulin secretion due to decreased glucose oxidation and the subsequently decreased ATP/ADP ratio.95 Another inhibitor of insulin release is stress, specifically norepinephrine (noradrenaline) produced in response to stress.96 Norepinephrine binds to its α2-adrenergic receptors, which are linked to GPCRs, resulting in the inhibition of AC as well as in hyperpolarization. This prevents an increase in the cytosolic Ca2+ concentration and, subsequently, insulin secretion.97, 98

Interplay between the pancreatic islets and other organs

The brain–islet axis

Just as insulin exerts its effects on other organs and tissues, other organs interact with the pancreas to modulate insulin secretion (Figure 4). One of these interacting organs is the brain, which comprises the mutual brain–islet axis that interacts with the pancreas and vice versa. The pancreas is highly innervated with both, parasympathetic99, 100 and sympathetic100, 101 nerve fibers from the autonomic nervous system. At the same time, insulin receptors are widely distributed within the brain, including the hypothalamus, cerebral cortex, cerebellum102 and hippocampal formation103 in humans, as well as the olfactory and limbic areas,104, 105 hypothalamus106—particularly the periventricular nucleus107 and the arcuate nucleus108, 109—hippocampus and the choroid plexus105 in rat brains. Lesions in various brain regions were shown to affect pancreatic hormone secretion. The destruction of the ventromedial hypothalamus results not only in insulin hypersecretion110, 111, 112 due to loss of the ventromedial hypothalamus-mediated inhibitory impact on pancreatic β-cells113 but also in higher glucagon levels.111, 112 Glucagon secretion may also be modulated by the hypothalamic brain-derived neurotrophic factor114 via efferent nerves,115 whereas the melanocortin system directly reduces basal insulin levels by stimulating sympathetic nerve fibers via α-adrenoceptors.116 Acting via α-adrenoceptors,117 norepinephrine also inhibits insulin secretion,96 which is an important aspect of the fight-or-flight response. The neurotransmitter Neuropeptide Y (NPY), which is mainly expressed in the sympathetic nerve fibers of the autonomic nervous system, also blunts insulin release,118, 119 and the loss of NPY’s inhibitory action results in elevated basal and glucose-stimulated insulin secretion as well as in increased islet mass.120 NPY binding to its GPCR Y1 causes the activated Giα-subunit to block adenylate cyclase activation, which in turn inhibits the cAMP pathway.121 Furthermore, the NPY-mediated inhibition was shown to be Gβγ- and Ca2+-independent.122 In addition to the well-known insulin stimulator acetylcholine, which exerts its effects via M3 muscarinic receptors,123 melanin concentrating hormone, vasoactive intestinal peptide (VIP), its close relative pituitary adenylate cyclase-activating polypeptide (PACAP) and gastrin-releasing peptide also promote insulin and, in the case of VIP124 and PACAP,125 glucagon release. The various neuropeptides exert their effects through various pathways, including the extracellular signal-regulated kinase (ERK)/Akt pathway, and modulation of Ca2+-influx (melanin concentrating hormone),126 cAMP and, to a lesser extent, PI3K signaling (VIP and PACAP),127, 128 muscarinic/β-adrenoceptors signaling, PI3K/PKC signaling and Ca2+-mobilization from intracellular stores (gastrin-releasing peptide).129, 130

Figure 4
figure 4

The interplay of the pancreas with the brain, liver, gut as well as adipose and muscle tissue. The pancreas interacts with the brain, liver, gut and adipose and muscle tissue in a highly sophisticated network via various hormones, neurotransmitters and cytokines. BNDF, brain-derived neurotrophic factor; CCK, cholecystokinin; GIP, glucose-dependent insulinotropic peptide; GLP-1, glucagon-like peptide 1; GRP, gastrin-releasing peptide; IL-6, Interleukin 6; MCH, melanin concentrating hormone; NPY, neuropeptide Y; PACAP, pituitary adenylate cyclase-activating polypeptide; POMC, pro-opiomelanocortin; VIP, vasoactive intestinal peptide.

Likewise, insulin release is stimulated by the so-called cephalic phase, which represents the conditioned reflex of increased hormone secretion, referred to as cephalic phase insulin response,131 even in the absence of nutrients/glucose as a trigger,132, 133, 134 such as when anticipating a meal, to prepare the organism to adequately respond to incoming nutrients.135 Moreover, cephalic phase insulin response is pivotal for ensuring normal postprandial glucose management.136 The neural mechanism underlying cephalic phase insulin response was found to include cholinergic and non-cholinergic processes136 as well as the dorsal vagal complex located in the medulla oblongata.137 Conversely, insulin released in response to a meal enters the brain via the blood–brain–barrier138 to decrease food intake139, 140 by stimulating hypothalamic pro-opiomelanocortin neurons141 and initiating the PI3K signaling pathway142 in these pro-opiomelanocortin neurons.143 In contrast to its pro-opiomelanocortin-stimulating action, insulin inhibits NPY expression144 in Agouti-related peptide (AgRP/NPY) neurons, which are known to secrete the orexigenic neuropeptides NPY145, 146, 147 and AgRP.148, 149 Both, peripheral and central insulin signaling are impaired in obese or diabetic states.150, 151, 152, 153, 154

The liver–islet axis

The second group represents the liver–islet axis. The liver has a key role in glucose homeostasis by storing (glycogenesis) or releasing (glycogenolysis/gluconeogenesis) glucose upon interaction with insulin and glucagon, respectively. The binding of glucagon to its hepatic GPCR evokes the signaling cascade described under ‘External factors affecting pancreatic hormone secretion’, eventually resulting in the activation of PKA, which in turn stimulates two processes; one promotes glycogenolysis/gluconeogenesis and the other inhibits glycolysis/glycogenesis.155, 156 Glycogenolysis is a multistep process that includes the PKA-mediated phosphorylation of phosphorylase kinase,157 cleavage of glucose-1-phosphate (G-1-P) from glycogen by activated glycogen phosphorylase a158 and the conversion of G-1-P into G-6-P,159 eventually resulting in phosphate and free glucose. Hepatic gluconeogenesis is promoted by the PKA-mediated phosphorylation of the cAMP response element-binding protein, which in turn upregulates peroxisome proliferator-activated receptor-γ coactivator (PGC)-1.160 Together with the hepatocyte nuclear factor (HNF)-4, PGC-1 induces the transcription of phosphoenolpyruvate carboxykinase,161 which catalyzes the conversion of oxaloacetate into phosphoenolpyruvate, a rate-limiting step in gluconeogenesis. This is followed by reversed glycolysis, during which stimulation of the bifunctional PFK-2/FBPase-2 leads to both, enhanced gluconeogenesis through the abrogation of disabled fructose-1,6-bisphosphatase (FBPase)-1, which facilitates the successive conversion of substrates into G-6-P, and to suppressed glycolysis.162, 163 Glycolysis is further inhibited by the PKA-mediated inactivation of pyruvate kinase,164, 165, 166 resulting in the production of glucose instead of pyruvate. In addition, glucagon was found to suppress pyruvate kinase gene expression as well as to enhance pyruvate kinase mRNA degradation.167, 168 Finally, the PKA-induced inactivation of hepatic glycogen synthase169, 170, 171 decreases glycogen synthesis and concomitantly increases the hepatic glucose pool.

As glucagon’s opponent, insulin stimulates glycolysis via enhanced expression of the hepatic glucokinase gene,14, 15 a key enzyme that converts glucose into G-6-P. This increase is mediated by the sterol regulatory element binding protein-1c15 and requires the absence of cAMP.14 Furthermore, insulin inactivates glycogen phosphorylase and glycogen synthase kinase (GSK)-3172 through the PI3K pathway, which in turn activates glycogen synthase.18, 19, 20 The second liver-specific effect of insulin is to repress the expression of the phosphoenolpyruvate carboxykinase and G-6-Pase genes; the first by disrupting the association of cAMP response element-binding protein and RNA polymerase II with the phosphoenolpyruvate carboxykinase gene promoter,23 whereas G-6-P suppression requires PKBα/Akt and forkhead transcription factor (FOXO1),24, 25 whose expression was shown to be diminished by the inhibition of GSK-3.26

It is not only insulin and glucagon acting on the liver; hepatocyte-derived factors conversely influence the pancreas and/or insulin secretion. Although HNF3β was proposed to be pivotal for the transcription of the pancreatic and duodenal homeobox 1 (pdx1 or insulin promotor factor 1 (IPF-1)) gene, a transcription factor regulating pancreatic development173, 174175, it is the loss of HNF1α resulting in an almost abolished insulin secretion, likely due to a decreased response to intracellular calcium. These findings support the importance of HNF1α in maintaining β-cell function176 and its involvement in maturity-onset diabetes of the young (MODY3).177

The hepatokine betatrophin, also known as TD26, re-feeding induced fat and liver (RIFL), lipasin or angiopoietin-like (ANGPTL) 8, was first identified as a factor that drives β-cell proliferation and thus increases β-cell mass in a murine model of insulin resistance.178 Subsequent studies, however, did not reveal impairments in glucose homeostasis179 or β-cell expansion in Angptl8 knockout mice.180 Moreover, betatrophin does not have an effect on human β-cell replication, challenging its usefulness in diabetes therapy.181 This is substantiated by the fact that betatrophin levels are higher in T2DM patients,182, 183, 184 although they were lower in one study.185 However, this is likely to be due to technical issues.186

The gut–islet axis

Another important axis is the gut–islet axis. The gut releases various hormones upon nutrient ingestion, including GLP-1 and GIP, that bind to their respective receptors on pancreatic β-cells to potentiate insulin secretion, as described under ‘External factors affecting pancreatic hormone secretion’. Furthermore, both hormones exert pancreatic effects, such as GLP-1-stimulated insulin gene expression,77, 187 incretin-induced β-cell neogenesis, proliferation188, 189, 190, 191 and survival,192 the prevention of β-cell apoptosis in general193, 194 and in response to glucolipotoxicity.195 The extrapancreatic actions of GLP-1 include suppression of endogenous glucose production196/glycogenolysis,197 glucagon secretion,197, 198 appetite,199, 200 a delay in gastric emptying198, 199 and improved β-cell insulin sensitivity199, 201, 202 and glucose disposal,203, 204 whereas GIP positively affects lipid205, 206, 207 and bone metabolism.208, 209, 210, 211 Thus, GLP-1 and GIP mediate insulin secretion and concomitantly, insulin modulates GIP212 and GLP-1 release; the latter ocurring through the PI3K/Akt- and mitogen-activated protein kinase kinase (MAPKK or MEK)/ERK1/2 pathway.213 The importance of this interplay is also demonstrated by defective insulin responses and consequent glucose intolerance in GLP-1R−/− and GIPR−/− mice214, 215, 216, 217, 218 as well as in the pathogenesis of T2DM.219, 220, 221, 222, 223

In addition to incretins, there are the so-called decretins, namely limostatin and Neuromedin U (NmU), which are secreted during fasting to suppress insulin release. NmU, a (neuro)peptide that mediates the contraction of smooth muscles in the uterus (hence the ‘U’) among others, was first isolated from the pig spinal cord.224 Further mRNA expression studies, however, revealed NmU to be highly expressed in the gastrointestinal (GI) tract with the highest levels found in the upper GI, that is, duodenum and jejunum.225, 226 Within the GI structure, NmU is mainly located in submucosal and myenteric cells,227, 228 indicating its possible involvement in the neuronal control of GI function.229 In addition to this, NmU is likely to regulate insulin secretion; the G-protein-coupled NmU receptor 1 (NmUR1) is expressed in pancreatic islets and its simulation dose dependently decreased insulin release.230, 231 The underlying mechanism involves the simultaneous release of somatostatin—a known modulator of insulin secretion6—upon NmUR1 activation.232 A very recent study showed231 that the peptide hormone limostatin, which is expressed in Drosophila melanogaster, also reduces insulin secretion and its absence caused hyperinsulinemia, hypoglycemia and obesity. Moreover, knockdown of the fly NmUR orthologue not only reproduced the consequences of limostatin deficiency but also diminished its insulin-suppressing ability. Limostatin release is initiated by food depletion and hence may represent a novel mechanism for modulating insulin secretion during fasting.

Other gastrointestinal hormones that interact with the pancreas are gastrin and cholecystokinin (CCK). Gastrin, which is secreted from G-cells in the stomach and duodenum, acts as an islet growth factor, together with transforming growth factor-α, by promoting differentiation of ductular precursor cells233 and β-cell neogenesis as well as by enhancing the islet mass from transdifferentiated exocrine pancreatic tissue.234 Furthermore, it induces the expression of glucagon genes in α-cells.235 Along the same lines, CCK, which is synthesized and released from duodenal I-cells, potentiates basal, glucose-236, 237 and amino acid-induced insulin secretion,238 and augments glucagon secretion.237, 239 The pivotal role of CCK in modulating glucose homeostasis is reflected in postprandial hyperglycemia, which is due to reduced CCK plasma levels in noninsulin-dependent diabetes mellitus.240

Another important factor that is related to metabolic disorders such as obesity, T2DM and type 1 DM (T1DM) is the gut microbiota. Obesity, T2DM and T1DM patients display alterations in the composition of their microbiota that may initiate and/or promote the respective disorder. Recent findings linked an aberrant microbiome, which is generally represented by diminished diversity, including fewer butyrate-producing (butyrate was shown to trigger mucin production and hence gut integrity) and mucin-degrading bacteria,241 to the development of autoimmunity in T1DM.242 An altered microbiota composition may also contribute to obesity243, 244 as well as to T2DM245, 246, 247 and ‘correction’ by antibiotics,248 probiotics249 or prebiotics, the last of which causing a short-chain FFA-stimulated increase in GLP-1,250 may improve the disease condition.251

The adipocytes/myocytes–islet axis

On one hand, insulin’s interplay with adipose and muscle tissues is broadly based on facilitating insulin-dependent glucose uptake through the glucose transporter 4 (GLUT4).11, 12, 13 On the other hand, adipokines and myokines secreted from the adipose and muscle tissue, respectively, modulate insulin release. As part of the so-called adipoinsular axis,252 leptin, the most famous adipokine, mainly acts on its receptors in the hypothalamic arcuate nucleus to inhibit food intake and control whole body homeostasis.253 However, leptin receptor (Ob-R) mRNA expression was also observed in pancreatic islets254 and its stimulation caused a reduction in insulin secretion255, 256, 257 due to the activation of KATP-channels, which in turn prevented Ca2+-influx258 and the subsequent signaling pathway. Furthermore, leptin was shown to suppress insulin gene expression,259, 260 representing a negative feedback loop. Conversely, insulin enhances ob gene expression and leptin secretion.261, 262, 263, 264 Likewise, insulin modulates the expression of adiponectin, another well-known adipokine, the abundance of its receptor in adipose and muscle tissue265, 266 as well as its secretion.267, 268 Adiponectin is not only involved in glucose and fatty acid metabolism269 but it also forestalls β-cell apoptosis and induces insulin gene expression and release;270 the latter was mediated by the ERK/Akt pathway in one study270 and by the AMPK pathway in another study.271 Other adipokines, such as apelin,272, 273 chemerin,274, 275, 276 omentin,277, 278 resistin279 and visfatin,280, 281 were also shown to directly interact with insulin, whereas retinol-binding protein 4, tumor necrosis factor-α and vaspin are related to insulin in an indirect manner.282 In addition to adipokine secretion by adipocytes, myocytes release cytokines, which are referred to as myokines. Fibroblast growth factor-21 is a widely expressed protein with a broad mode of action, including the regulation of carbohydrate and fatty acid metabolism283 and may be considered as a myokine due to its secretion from muscle cells.284 Fibroblast growth factor-21 is regulated by insulin285 through the PI3K/Akt1 signaling pathway.286 Interleukin (IL)-6, which is both an adipokine and myokine,287 was shown to influence the pancreas by controlling the expression of pro-glucagon mRNA as well as glucagon secretion. It also increases α-cell proliferation and islet mass while protecting the pancreas from metabolic stress-induced apoptosis.288 Furthermore, IL-6 increased GLP-1 production from proglucagon in pancreatic α-cells and its secretion from α-cells and intestinal L-cells, eventually resulting in a GLP-1-mediated increase in insulin secretion.289

Modulating insulin secretion as a means of diabetes therapy

Due to the worldwide, still spreading epidemic of T2DM, there is an urgent need for (new) anti-diabetic drugs and therapies that are more effective and have fewer side effects. Currently, the most commonly used drugs can be classified into agents that enhance insulin secretion (secretagogues such as sulfonylureas (SUs) and incretin mimetics), sensitize the target organs of insulin (for example, metformin from the class of biguanides or thiazolidinediones), or reduce glucose absorption from the gastrointestinal tract (inhibitors of gastrointestinal α-glucosidase). Different therapies address different problems and stages of T2DM and may be prescribed in combination to exert synergistic effects.

Sulfonylureas

A-glucosidase inhibitors and sensitizers do not target the pancreas or insulin secretion itself but instead target the upstream (slowed intestinal glucose absorption) or downstream (improved insulin sensitivity) processes. In contrast, insulin secretagogues directly modulate insulin release. The SUs are the first broadly applied oral anti-hyperglycemic drugs. To date, there are two generations of agents: acetohexamide, chlorpropamide, tolazamide and tolbutamide, which constitute the first generation and glibenclamide/glyburide, gliclazide, glimepiride, glipizide and gliquidone, which comprise the second generation. First-generation SUs are rarely used these days since tolbutamide intake was associated with an increase in lethal cardiac events.290, 291 More importantly, the second-generation SUs are more potent due to modifications in their side chains’ structure, resulting in improved SUR-affinity, accompanied by lower effective plasma levels, which in turn may reduce undesirable drug-protein interactions.

All SUs share a central SU backbone but differ in their side chains. Despite having different pharmacokinetics, they work in the same way, namely by triggering endogenous insulin release by blocking KATP-channels and hence activating the insulin signaling pathway. More precisely, SUs bind to the sulfonylurea receptor (SUR) subunit of the KATP-channel with high affinity.292, 293 SUR, together with the pore-forming subunit Kir6.x, forms a hetero–octameric complex consisting of four inner Kir6.x subunits surrounded by four SUR subunits (4:4 stoichiometry).294, 295 Moreover, different isoforms of the two subunits are expressed, depending on the tissue-specific expression of the KATP-channels: SUR1 and Kir6.2 are expressed in the pancreas and brain,296 Kir6.2 and SUR2A are expressed in the heart and skeletal muscle,297 while SUR2B is expressed in the brain and smooth muscle,298 and Kir6.1 and SUR2B are expressed in vascular smooth muscle.299 Although SUs bind to both, SURs and Kir6.2, the interactions with the latter are of low affinity300, 301 and hence only SUR-interacting agents are used for diabetes treatment. In addition to their mode of action as inhibitors of KATP-channels, SUs were shown to improve glucose uptake into insulin-dependent tissues and glucose disposal as well as to reduce hepatic glycogenolysis/gluconeogenesis.302, 303, 304

In contrast to SUs inactivating the KATP-channels by binding to the SUR1 subunit, ATP closes them by interacting with Kir6.2.305 Moreover, while the binding of only one ATP molecule is sufficient to completely close the channel,306 inhibition by SUs is incomplete as the channel might still open even when SUs are bound to SUR1.299 Nonetheless, second-generation SUs reduce the glycated hemoglobin or HbA1c, which represent the average plasma glucose concentrations over time and thus serve as a diagnostic measure for diabetes mellitus, by 1.0–2.0%. In addition to the weight gain attributed to the anabolic effects of increased insulin secretion, the main side effect of SUs is hypoglycemia307, 308 due to excess circulating insulin levels and due to the fact that SUs evoke insulin secretion in a glucose-independent manner.309

Although they are not SUs per se, meglitinides, that is, repaglinide and nateglinide, share their mode of action of inhibiting KATP-channels.310 However, meglitinides and some of the second-generation SUs, for example, glibenclamide, interact with both, the SUR1 and the SUR2A or B isoforms.311 Despite the possible disadvantage of this generalized binding that may cause undesirable effects on other KATP-channel types, for example, those in the heart,312 meglitinides, namely nateglinide, have an earlier onset of action and a faster dissociation rate from the sulfonylurea receptor,313, 314, 315 resulting in a diminished risk of hypoglycemia.316 Like SUs, meglitinides also cause weight gain.317, 318

Incretin mimetics

Another group of insulin secretagogues is comprised of the incretins GLP-1 and GIP. As both incretins are rapidly inactivated by the enzyme dipeptidyl peptidase IV (DPP-IV),319 their application in T2DM treatment focuses on modified analogues320, 321, 322, 323, 324, 325 or receptor agonists, including the well-known, short-acting exenatide.326, 327, 328 The long-lasting agonists exenatide LAR,329, 330 liraglutide331, 332 and lixisenatide333, 334, 335 are currently under investigation. However, based on the lipogenetic properties205, 206, 207 of GIP, insufficient insulin-potentiating effects in T2DM patients220, 336 and a possible worsening effect by GIP,337, 338 the focus is on GLP-1 analogues/receptor agonists for T2DM treatment. By acting on its receptor, GLP-1 induces the signaling cascade described under ‘External factors affecting pancreatic hormone secretion’, resulting in its main effect: potentiating insulin secretion. In addition to reducing the HbA1C levels, GLP-1 analogues/receptor agonists promote weight loss and, more importantly, do not evoke hypoglycemia, as do SUs,326, 327, 328, 329, 330, 331, 332, 333, 334 due to the glucose-dependent mode of action and the self-regulating mechanism of GLP-1.68, 336, 339 When blood glucose levels are lowered to physiological levels, GLP-1 is incapable of enhancing insulin secretion, thereby preventing hypoglycemia.79, 340 In addition, GLP-1 (analogues/receptor agonists) exerts further pancreatic and extrapancreatic actions, as mentioned under ‘Interplay between the pancreatic islets and other organs’. Although GLP-1 (analogues/receptor agonists) exhibits some minor side effects, including nausea, vomiting or gastrointestinal impairments,326, 327, 328, 329, 330, 331, 332, 333, 334, 335 the beneficial properties outweigh the negative effects, and thus, GLP-1 is a promising anti-diabetic agent.

Insulin sensitizers

Metformin, which is generally the most widely used first-line anti-diabetic medication,341 is a so-called (insulin) sensitizer. It not only diminishes hepatic glucose output due to glycogenolysis/gluconeogenesis342 but it also enhances glucose uptake into peripheral tissues, such as skeletal muscle, by activating 5′-adenosine monophosphate-activated protein kinase (AMPK-α2).343 Furthermore, it supports weight loss344 by reducing food consumption.345 With respect to its effects on β-cell function, metformin was shown to increase insulin gene expression,346 possibly by nuclear accumulation of pdx1 and its subsequently improved DNA-binding activity.347 Interestingly, metformin exerts opposing effects on β-cell proliferation and/or apoptosis; on the one hand, it suppresses β-cell proliferation and enhances apoptosis through an AMPK-dependent and autophagy-mediated mechanism348 following the metformin-induced activation of c-Jun-N-terminal kinase and caspase-3.349 On the other hand, metformin reduces caspase-3- and -8-mediated apoptosis in isolated islets from T2DM patients350 and protects against lipotoxicity-induced β-cell defects.348, 351

The other members of the sensitizer group include the thiazolidinediones (or glitazones). Currently, only pioglitazone is available; troglitazone was withdrawn from the market in 2000 and rosiglitazone was withdrawn in 2010 due to liver toxicity, drug-induced hepatitis352, 353, 354 and the increased risk of cardiovascular events, respectively.355 Their mode of action involves activation of the peroxisome proliferator-activated receptor (PPARγ), a nuclear transcription factor that is highly expressed in adipose tissue, and the subsequent regulation of genes that are involved in glucose and fat metabolism.356, 357, 358 By promoting lipogenesis, FFAs are removed from the blood stream, whereupon cells become dependent on glucose as an energy substrate. However, enhanced lipogenesis also leads to the weight gain observed in thiazolidinedione-treated T2DM patients.359 In contrast to metformin, pioglitazone prevents (oxidative stress-induced) apoptosis360, 361 by decreasing the expression of apoptosis-promoting genes, while increasing anti-apoptotic and anti-oxidative gene expression. However, this may depend on the disease state.362, 363 Furthermore, pioglitazone increases β-cell mass by upregulating cell differentiation/proliferation genes.364 Although they have partially different modes of action, both groups of sensitizers cause a reduction in the HbA1c level by 1.5–2.0%.

A-glucosidase inhibitors

A-glucosidase inhibitors, such as acarbose, miglitol and voglibose, not only decelerate the breakdown of starch into glucose in the small intestine but also decrease its bioavailability, resulting in reduced levels of glucose entering the blood stream and hence attenuated postprandial glucose excursions.365, 366, 367, 368, 369, 370 In addition, they support weight loss371, 372 and ameliorate blood pressure,373 insulin sensitivity367, 368 and triglyceride levels.369, 370 Similar to pioglitazone, α-glucosidase inhibitors attenuate reductions in β-cell mass, which may delay the onset of diabetes.374, 375, 376 As α-glucosidase inhibitors only mildly reduce HbA1c levels (0.5–1.0%), they are usually only used in the early stage of T2DM, that is, impaired glucose tolerance or in combination with other drugs.377

Conclusions and outlook

The pancreas has key roles in maintaining normal blood glucose levels by producing and releasing insulin and glucagon. These opponents interact not only with each other through the intra-islet insulin axis378, 379, 380, 381 but also with other organs/tissues, that is, the brain, liver, gut as well as insulin-dependent adipose and muscle tissues. Altogether, the islet–organ/tissues axes described here form a highly sophisticated network that includes, but is not limited to, various signaling molecules, that is, neuropeptides (brain-derived neurotrophic factor, NPY, melanin concentrating hormone, gastrin-releasing peptide, VIP and PACAP), hepatokines (betatrophin and HNFs), enteroendocrine hormones (the incretins GLP-1 and GIP, the decretins NmU and limostatin, gastrin and CCK) as well as adipokines (leptin and adiponectin) and myokines (fibroblast growth factor-21 and IL-6) that mainly interact through GPCR signaling pathways, such as the cAMP cascade. In good health, the well-functioning interactions between all of the organs and tissues involved ensure glucose homeostasis. However, impairments in the secretion of and/or sensitivity to insulin may result in metabolic diseases, such as T2DM. Referring to the American Diabetes Association, T2DM and noninsulin-dependent diabetes mellitus are characterized by insulin resistance, hyperglycemia and a relative insulin deficiency. Furthermore, T2DM is associated with low-grade inflammation,382, 383 cardiovascular disease,384, 385 nephropathy386, 387 and alterations in the secretion of various hormones, including IL-6, IL-18, tumor necrosis factor-α,388 adiponectin and leptin,389 neuropeptides,390 ghrelin391, 392 and the incretins GLP-1 and GIP.219, 220, 221, 222, 223 Although lifestyle interventions393 and weight loss394 reverse T2DM in early stages, when insulin is still secreted, T2DM patients may become dependent on anti-diabetic drugs in later stages. Currently, there are three classes of agents: insulin secretatogues, insulin sensitizers and α-glucosidase inhibitors, all of which have different modes of action and hence target different stages and symptoms of T2DM. Treatments that modulate insulin release—on condition of an appropriate insulin sensitivity of the target organs—appear to be promising approaches. Current research is unveiling new molecules, enzymes and interactions that are involved in the signaling pathways underlying insulin secretion, among others, and is likely to introduce new therapeutic approaches. Strategies that target these mediating molecules may include, but are not limited to, the calcium sensor Syt-7,90, 395 the SNARE-associated protein Snapin,89 the t-SNARE SNAP-25,396 cyclin-dependent kinase (Cdk) 5,397 ryanodine receptor (RyR) 2,398 the nucleotide exchange factor and intracellular cAMP sensor Epac2,57, 58, 59, 399 mammalian uncoordinated proteins (munc)13400, 401 and munc1840, 41 as well as the Ras-related proteins (Rab) 3A402 and 27A.403