Minireviews Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Cases. Oct 6, 2023; 11(28): 6670-6679
Published online Oct 6, 2023. doi: 10.12998/wjcc.v11.i28.6670
Neurotransmitters regulate β cells insulin secretion: A neglected factor
Chu-Chu Kong, Ji-Dong Cheng, Wei Wang, Department of Endocrinology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361100, Fujian Province, China
ORCID number: Wei Wang (0000-0002-7453-7391).
Author contributions: Kong CC wrote and revised the manuscript; Cheng JD searched references; Wang W proposed ideas and gave final approval for the submission.
Supported by National Natural Science Foundation of China, No. 81471081; the Natural Science Foundation of Fujian Province, China, No. 2019J01010; Xiamen Research Foundation for Science and Technology Project No. 3502Z20194037; and Scientific Research Foundation for Advanced Talents, Xiang’an Hospital of Xiamen University, No. PM201809170005.
Conflict-of-interest statement: There is no conflict of interest associated with the senior author or other coauthors who contributed their efforts in this manuscript.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Wei Wang, MD, Doctor, Department of Endocrinology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, No. 2000 Xiang'an East Road, Xiang'an District, Xiamen 361100, Fujian Province, China. wwei19742007@hotmail.com
Received: April 22, 2023
Peer-review started: April 22, 2023
First decision: July 4, 2023
Revised: July 17, 2023
Accepted: August 31, 2023
Article in press: August 31, 2023
Published online: October 6, 2023

Abstract

β cells are the main cells responsible for the hypoglycemic function of pancreatic islets, and the insulin secreted by these cells is the only hormone that lowers blood glucose levels in the human body. β cells are regulated by various factors, among which neurotransmitters make an important contribution. This paper discusses the effects of neurotransmitters secreted by various sympathetic and parasympathetic nerves on β cells and summarizes the mechanisms by which various neurotransmitters regulate insulin secretion. Many neurotransmitters do not have a single source and are not only released from nerve terminals but also synthesized by β cells themselves, allowing them to synergistically regulate insulin secretion. Almost all of these neurotransmitters depend on the presence of glucose to function, and their actions are mostly related to the Ca2+ and cAMP concentrations. Although neurotransmitters have been extensively studied, many of their mechanisms remain unclear and require further exploration by researchers.

Key Words: β cells, Insulin secretion, Neurology, Type 2 diabetes, Islet

Core Tip: β-Cells are the only islet cells in the body that release insulin, and the neurotransmitter is an important factor in regulating insulin secretion. This review systematically describes the release sources of different neurotransmitters and their effects on insulin secretion. There are common mechanisms among different neurotransmitters, which mainly involve neuroanatomy and blood glucose homeostasis in vivo. There are great differences among different species. Although many specific mechanisms remain to be explored, this review provides a certain reference value and direction for subsequent research.



INTRODUCTION

Islets are important endocrine micro-organs for maintaining glucose homeostasis. In animals, intermittent feeding is an important cause of fluctuations in blood glucose levels, which are precisely controlled within a certain range by various hormones secreted by the islets. Islets are scattered in the exocrine parenchyma of the pancreas and are composed of cells, 65%-80% of which are β cells that secrete insulin (the only hypoglycemic hormone in the body)[1]. In rodents, β cells are located in the center of the islets and are surrounded by α-cells (which secrete glucagon), δ-cells (which secrete growth inhibitory hormone), and PP-cells (which secrete pancreatic polypeptide). In humans, islets are formed by three main cell types in close proximity, allowing more local interactions than in rodents[2]. The blood glucose level is undoubtedly an important factor in the regulation of islet function. In addition, islets are innervated by autonomic nerves; thus, a logical hypothesis is that neurotransmitters act as messengers to regulate the autonomic innervation of islets[1]. Recent studies have found that in addition to neurotransmitters, many neuropeptides located in islets also participate in neuroregulation[3]. The nerves that innervate islets mainly include sympathetic, parasympathetic and sensory nerves; acetylcholine (ACh), vasoactive intestinal polypeptide (VIP), gastric releasing peptide (GRP), 5-hydroxytryptamine (5-HT), pituitary adenylate cyclase activating peptide (PACAP) and nitric oxide (NO) are stored in parasympathetic nerve endings; norepinephrine (NE), dopamine (DA), galanin, calcitonin gene-related peptide (CGRP) and neuropeptide Y (NYP) are stored in sympathetic nerve endings; and sensory neuropeptides, cholecystokinin and substance P are stored in sensory nerve endings[1] (Table 1). Neurotransmitters do not only originate from nerve endings; in addition, β cells, as nonneural sources of neurotransmitters, synergize with nerve endings to secrete neurotransmitters and exert autocrine or paracrine effects locally[1]. These neurotransmitters synthesized by β cells are also released into the bloodstream together with insulin (Figure 1).

Figure 1
Figure 1 Regulation of insulin secretion by neurotransmitters. With the increase of glucose concentration, the concentration of adenosine triphosphate (ATP) produced by glucose metabolism also increases correspondingly, and the closure of internal rectifying K+-ATP channels leads to membrane depolarization, which opens volt-sensitive Ca2+ channels, and Ca2+ enters intracellularly to mediate insulin secretion in large quantity. Acetylcholine, gastrin releasing peptide, 5-hydroxytryptophan, vasoactive intestinal peptide, pituitary adenylate cyclase activating polypeptide and nitric oxide promote insulin secretion. Dopamine, neuropeptide Y, galanin and norepinephrine inhibit insulin secretion. 5-HTTP: 5-hydroxytryptophan; AADC: Amino acid decarboxylase; ADP: Adenosine diphosphate; cAMP: Cyclic adenosine monophosphate; CCK: Cholecystokinin; cGMP: Cyclic guanosine monophosphate; DAG: Diacylglycerol; GCRP: Calcitoningenerelatedpeptide; GTP: Guanosine triphosphate; IP3: Inositol triphosphate; L-Arg: Levo-arginine; L-DOPA: Levo-dopa; NOS: Nitric oxide synthase; PIP2: Phosphatidylinositol(4,5)bisphosphate; PKA: Protein kinases A; PKC: Protein kinase C; PKG: Protein kinases G; PLC: Phospholipase C; SP: Substance P; TCA: Tricarboxylic acid cycle; TPH: Tryptophan hydroxylase; Trp: Tryptophan; VIP: Vasoactive intestinal peptide.
Table 1 Role of neurotransmitters in the islet.
Name
Source
Receptor
Effect on pancreatic islet secretion
AChVagus nerveM3↑↑↑
NOVagus nerve, beta cellssGCLow concentration ↑, high concentration ↓
GRPVagus nerveGRPR
VIPVagus nerveVIP-2
PACAPVagus nerve, gut-pancreas nervePACAP-3
5-HTVagus nerve, gut-pancreas nerve, beta cellsHtr1↑↑
NESympathetic nervesα2, β2α2 receptors ↑, β2 receptors ↓↓
GalaninSympathetic nervesGALR1
NPYSympathetic nervesY2
DASympathetic nerves, beta cellsD2, D3D2↑, D3↓
CGRPSensory nervesCGRP
SPSensory nervesNK1
CholecystokininSensory nervesCCK

Islets are mainly innervated by sympathetic and parasympathetic nerves. Sympathetic preganglionic neurons are located in the mediolateral spinal cord at the T6-L2 levels, and their axons project from the visceral nerves to the superior mesenteric and celiac ganglia and to the pancreas[4]. The preganglionic fibers of parasympathetic nerves originate from the dorsal nucleus of the vagus nerve[5] and possibly from the ambiguous nucleus[6], both of which are under hypothalamic control. It is traditionally believed that sympathetic nerves play the opposite role as parasympathetic nerves in insulin secretion. In mice and humans, sympathetic nerves mainly stimulate the secretion of glucagon by α cells and reduces the secretion of insulin by β cells by regulating the islet vascular system[7]. However, it has been reported that the level of insulin increases slowly when visceral sympathetic nerves are stimulated in anesthetized dogs[8]. Parasympathetic nerves mainly play a role in regulating insulin secretion in the cephalic stage by integrating taste, smell, and visual signals in the hypothalamus, eventually leading to the stimulation of insulin secretion[9]. Dietary hormones such as cholecystokinin and 5-HT can also activate the vagus nerve. Vagal fiber endings release a variety of neurotransmitters, such as ACh and gut GRP[10]. In a previous study, vagotomy was performed in both fed and fasted animals, and it was found that the vagus nerve had no effect on insulin secretion in fasted animals[11] but was involved in insulin secretion in fed animals. Therefore, it is generally believed that the vagus nerve is involved in insulin secretion under high-glucose feeding conditions but has little effect on basal insulin secretion. In addition, many studies have shown that the mode of glucose administration also affects the control of insulin secretion by the vagus nerve; for example, intravenous glucose administration rarely affects basal insulin secretion, while vagotomy substantially reduces insulin secretion when glucose is administered orally[1]. All these studies suggest that glucose is an important stimulus for the regulation of β cells by the vagus nerve in the gastrointestinal tract. Although the sympathetic nerves in the pancreas originate from the lower thoracic and upper lumbar segments of the spinal cord[12], their myelin sheaths form the paravertebral sympathetic chain[1] or cross the viscera to reach the ventral and mesenteric ganglia[12], and the postganglionic fibers emanating from the paravertebral sympathetic chain, ventral ganglia and mesenteric ganglia ultimately innervate the pancreas. Neurotransmitters may be involved in regulating insulin secretion in healthy people as well as in individuals with type 1, type 2, or other types of diabetes, so we do not distinguish between their specific roles in various types of diabetes.

NEUROTRANSMITTERS ORIGINATING FROM NERVE ENDINGS
ACh

ACh, an important hormone secreted by the vagus nerve, has traditionally been the focus of research by endocrinologists. However, ACh has been shown to be rapidly hydrolyzed by cholinesterase in the cytoplasm, and thus direct measurement of ACh levels is impossible. Therefore, researchers usually choose to measure plasma PP levels instead of Ach levels. PP also plays a role in regulating insulin secretion. PP acts on the Y4 receptor of delta cells, reducing somatostatin secretion and promoting insulin secretion[13].

ACh promotes insulin secretion, but this effect mainly depends on Ca2+ and glucose. After ACh is secreted by the vagus nerve, it acts on M3 receptors in β cells to promote glucose-mediated coupling of phospholipase C (PLC) to G proteins. The coupling of PLC to G proteins results in phosphoinositide (PI) hydrolysis. The hydrolysis of PI leads to the formation of inositol triphosphate (IP3). The binding of IP3 to IP3 receptors promotes the release of Ca2+ from the Ca2+ pool, thereby increasing the [Ca2+]i. It should be emphasized that the increase in Ca2+ release caused by the ACh-induced IP3 pulse peaks every 5-7 min, and this pattern of release is synchronized with the pulse secretion of insulin[14,15]. When a large amount of Ca2+ is released from the intracellular Ca2+ pool, the lack of Ca2+ in the storage pool leads to an inward flow of extracellular Ca2+, leading to volumetric Ca2+ inward flow, a second mechanism by which ACh promotes an increase in Ca2+ concentrations. In addition, voltage-dependent Ca2+ channels are located in the β-cell membrane, and large amounts of Ca2+ enter the cell when they are open.

ACh promotes insulin secretion by increasing the Ca2+ concentration in β cells on the one hand and enhances the effect of Ca2+ in promoting insulin secretion on the other hand. According to many studies, the PLC-PKC pathway may be involved in the mechanism by which ACh increases the prosecretory effect of Ca2+[10]. PLC activation results in the production of the diglyceride DAG2, which is a PKC activator that induces the translocation of PKC from the cytoplasm to the cell membrane; however, the mechanism by which PKC increases the effect of Ca2+ in promoting insulin secretion remains to be investigated. Further studies are needed to investigate whether phospholipase A2 is involved in this process. In addition, ACh does not regulate insulin secretion in one direction. ACh stimulates insulin secretion directly but simultaneously inhibits insulin secretion by promoting somatostatin secretion from δ-cells[1].

GRP

Enterostatin is a hormone that promotes insulin secretion, and its mechanism of action is closely related to glucose and Ca2+. However, the mechanism has not been fully elucidated. GRP seems to stimulate insulin secretion through direct and indirect mechanisms, and GRP seems to promote glucagon secretion.

GRP does not increase insulin secretion when the extracellular glucose concentration is less than 11.1 mmol/L in β cells, leading to the conclusion that the effect of GRP in promoting insulin secretion depends on the presence of glucose. Likewise, it was found that GRP did not increase the insulin concentration or insulin secretion when the extracellular culture medium was deficient in Ca2+, suggesting that GRP function also depends on extracellular Ca2+[16]. However, in subsequent experiments, it was found that although GRP acts continuously on β cells, the extracellular secretion of Ca2+ is not continuous; thus, GRP promotes insulin secretion by inducing only transient extracellular secretion of Ca2+. GRP stimulates IP3 production[17] but not IP3 hydrolysis[16]. GRP likely promotes PKC production by stimulating DAG production, and PKC promotes insulin secretion[18], suggesting that GRP also indirectly promotes insulin secretion. Overall, the GRP-mediated promotion of secretion depends on the presence of Ca2+. First, GRP stimulates IP3 production, and although it does not promote its hydrolysis, it still substantially increases the intracellular Ca2+ concentration. In addition, GRP also seems to inhibit K+ channels, which accelerates β-cell depolarization and prolongs its duration, which is more favorable for inward Ca2+ flow. Many studies have also found that in addition to having direct effects, GRP may act indirectly through cholinergic mechanisms. The muscarinic receptor M3 is involved in the action of GRP in promoting insulin secretion[19].

VIP and PACAP

VIP and PACAP have similar mechanisms of action and are colocalized in rat ganglion neurons, so they are often discussed together. PACAP and VIP share the receptors VIP1 and VIP2, while PAC1 is a specific receptor for PACAP[6]. Of course, PACAP can also be secreted by the enteropancreas. PAC1 is expressed in intrapancreatic ganglia, intestinal neurons, and beta cells[3]. Similar to NO and GRP, VIP and PACAP act in a glucose-dependent manner; when glucose is absent in the extracellular matrix, the effects of VIP and PACAP are almost negligible. Most studies suggest that the mechanisms of action of VIP and PACAP are related to their ability to stimulate increased cyclic adenosine monophosphate (cAMP) production[20], which increases the intracellular Ca2+ concentration in β cells. However, when Ca2+ is removed from the extracellular matrix, the intracellular Ca2+ concentration in β cells does not increase significantly, which suggests that instead of intracellular Ca2+ being released from the storage pool, extracellular Ca2+ flows into the cell[20]. VIP and PACAP increase the cAMP concentration, and a peak in the intracellular Ca2+ concentration can indeed be detected in β cells[21]. However, in mice, researchers did not observe an association between VIP-stimulated insulin release and increased cAMP levels[22], suggesting that signaling pathways other than the cAMP pathway might also participate in this process. When the VIP- and PACAP-stimulated increase in cAMP levels was reversed, it was found that VIP and PACAP stimulated a transient increase in cAMP and Ca2+ concentrations but a persistent increase in insulin levels, which supports the existence of other mechanisms[21].

NE

NE is released into the pancreatic vein mainly by sympathetic excitatory postganglionic fibers[23]. In animal experiments, exogenous NE was found to inhibit glucose-mediated insulin secretion; colistin, an α2-adrenergic receptor agonist, was found to inhibit insulin secretion[24], as was the β-blocker, propranolol. Therefore, NE is presumed to inhibit insulin secretion. Phentolamine, however, acts as an α-adrenergic receptor blocker and seems to counteract the inhibition of insulin secretion after neuroelectrical stimulation[25]. NE may likewise promote insulin secretion; one pathway might be the direct stimulation of insulin secretion through the activation of β2-adrenergic receptors in pancreatic β cells, and the other pathway may involve signaling directly through α2 and β2-adrenergic receptors in α cells[26], promotion of glucagon secretion by these cells and indirect stimulation of insulin secretion through glucagon. Therefore, the effect of NE on islets seems to be a vector sum of the actions of two receptors.

However, in general, the main effect of NE on β cells is the inhibition of insulin secretion. The mechanism underlying this effect is mainly related to the promotion of K+-ATP channel opening by NE. The opening of K+-ATP channels directly affects the Ca2+ concentration in β cells[27], and when NE acts on α2-adrenergic receptors, K+-ATP channels remain open, β cells are in a hyperpolarized state, and Ca2+ does not flow inward, which in turn inhibits insulin release[28]. After NE acts on β2-adrenergic receptors, it also inhibits the reduction in cAMP production by β cells[29], which inhibits insulin production through another pathway.

NPY

Galanin and NPY are both released by sympathetic nerve fiber terminals, and their role in inhibiting insulin release is well recognized. Although they exert different inhibitory effects in different species, in general, their directions of action are consistent. Although the results of their actions are well understood, their mechanisms remain unclear. Galanin has been shown to inhibit insulin secretion in a variety of animals[30]. Although the mechanism remains unclear, galanin was found to inhibit the glucose-mediated increase in the Ca2+ concentration in previous studies[31]. Therefore, we speculate that galanin may inhibit K+-ATP channel opening to inhibit β-cell membrane depolarization. When researchers induced β-cell depolarization with K+, galanin did not inhibit insulin secretion[32]; while other mechanisms through which galanin inhibits Ca2+ inward flow must exist, they remain unclear. Many studies suggest that NPY may inhibit insulin secretion by inhibiting G protein coupling and adenylate cyclase activity.

Sensory nerves

In addition to parasympathetic and sympathetic nerves, islets are also innervated by a wide range of sensory nerves, which secrete CGRP and SP. The CGRP receptor complex is expressed in mouse β cells, while the substance SP receptor NK-1R is expressed in mouse α cells. CGRP stimulates the release of gastrointestinal growth inhibitory hormone, which inhibits insulin release, and this pathway might be the mechanism by which CGRP inhibits insulin release. CGRP increases blood flow to the pancreas, which leads to a decrease in the insulin concentration due to an increase in blood flow when an increase in the insulin concentration is detected[33]. Several experiments have shown that CGRP inhibits glucose-induced insulin release, but the exact mechanisms involved need to be further investigated. However, there are some species-specific differences in this phenomenon. For example, CGRP does not affect insulin secretion in cattle[34]. The effect of substance P on insulin shows more obvious species-specific differences. While in vitro experiments on the effect of substance SP on insulin in rats have yielded inconsistent results, it has been shown that substance SP inhibits glucagon secretion; however, in dogs, substance SP stimulates insulin and glucagon secretion in a concentration-dependent manner[35,36]. In vivo, substance SP inhibits insulin secretion in rats but increases insulin and glucagon secretion in pigs[36,37].

CGRP and substance P are also coexpressed with insulin receptors in vagal afferent neurons, but CGRP is predominantly expressed in spinal sensory fibres[6]. In addition, some other neurotransmitters that are secreted by the pancreatic nerve have been identified; for example, cholecystokinin promotes insulin secretion, and its mechanism may be related to the activation of PLC[1].

NEUROTRANSMITTERS ORIGINATING FROM BOTH NERVE ENDINGS AND NONNERVE ENDINGS
5-HT

5-HT is a monoamine derivative that is present in the central nervous system and peripheral tissues[38], and as one of the most intense stimuli of vagal afferent neurons[39], it plays a role not only in neurotransmission in the central nervous system and brain development[40] but also in the physiological control and development of peripheral tissues[41]. In rats, 5-HT is also secreted by enteral neurons in the duodenum and stomach[42]. 5-HT is synthesized by tryptophan hydroxylase (TPH) and aromatic amino acid decarboxylase (AADC) via a two-step reaction[43]. Pancreatic β cells synthesize 5-HT de novo and release it via vesicular monoamine transporter 1/2 (VMAT1/2) as an autocrine and paracrine signal[44]. 5-HT is released from β cells along with insulin and ATP[45], and 5-HT secretion is regulated by the blood glucose concentration[46]. The 5HT3 receptor is a 5-HT receptor that is highly expressed on vagal afferent neurons. Antagonism of 5-HT receptors inhibits the response of vagal sensory neurons to β-cell stimulation[47], although the exact mechanism is unclear. In addition, intracellular 5-HT in β-cell granules acts as a substrate for the production of secretion-related proteins, which enhances insulin granule secretion[43]. However, differences in the effects of exogenous and endogenous or redistributed 5-HT on insulin secretion have been identified. Extracellular 5-HT is usually presumed to attenuate insulin release by stimulating Htr1a receptors in islet tissue, possibly through 5-HT receptor subfamily members (Htr1) that are coupled to Gαi and thus inhibit cAMP formation.

5-HT effectively inhibits insulin and glucagon secretion in islets under nondiabetic conditions. Surprisingly, this inhibition is lost in the islets of individuals with T2D[48], suggesting that functional 5-HT signaling is essential for normal β-cell function. Furthermore, 5-HT has recently been shown to play a role in β-cell expansion and compensatory insulin secretion during pregnancy[45].

DA

DA, an excitatory neurotransmitter secreted by sympathetic nerves, plays an important role in insulin secretion. β cells, which are important sites for nonneuronal DA synthesis and utilization, also secrete DA and coregulate insulin secretion with sympathetic nerves. β cells convert DA precursors (e.g., L-DOPA) to DA mainly through the actions of AADC[49], and newly synthesized DA is subsequently released in response to high glucose stimulation. The increase in DA secretion by β cells in response to glucose stimulation may be due to increased L-DOPA uptake and subsequent DA synthesis, resulting in a larger pool of releasable DA[50]. Synthesized DA is stored in β cells and is consecrated with insulin via VMAT2[38]. This endogenous DA signals in an autocrine/paracrine manner in insulin-secreting cells that express D2-like receptors. When DA is bound, it activates the Gα subunit to negatively regulate cAMP production, thereby decreasing protein kinase A activity[51] and subsequently activating PLC and reducing intracellular Ca2+ levels by affecting intracellular stores and L-type calcium channels[52]. In addition, D2-like receptors regulate G protein-coupled inward rectifying K+ channels[53]. Plasma concentrations of glucose and insulin are usually tightly linked, and alterations in glucose levels may result in rapid alteration in insulin levels in an attempt to bring glucose levels back to normal. Psychotropic drugs such as tiapride have a potent antagonistic effect on D2 receptors. Therefore, in previously reported patients, these counterregulatory mechanisms may not have worked, as they may have resulted in continuous insulin release even in the presence of normal or low glucose levels[54]. Furthermore, a certain rhythmicity in the regulation of insulin secretion by DA was found. When L-DOPA is coadministered with the D2 receptor antagonist sulpiride, the decrease in the circadian rhythm amplitude produced by the L-DOPA treatment was reversed, and the amplitude was restored to the level in carrier-treated cells. Based on these results, the D2 receptor modulates the effect of L-DOPA on circadian rhythm amplitude[55]. In contrast, the D2 receptor agonist bromocriptine significantly reduces the circadian rhythm amplitude in a concentration-dependent manner.

NO

NO is a stable and effective gaseous mediator that has become a hot topic in medical research in recent years, and a consensus on its role in vasodilation has been achieved. However, controversy exists regarding the role of NO in glucose-induced insulin secretion, as both inhibitory and facilitatory effects have been reported; whether glucose inhibits or promotes insulin secretion may be related to the NO concentration.

NO is secreted not only by vagal nerve terminals but also by HIT-T15 in hamster islet β cells stimulated with glucose[56]. L-arginine, a precursor of NO, is present in β cells, and L-arginine was shown to enhance the effect of glucose in promoting insulin secretion as early as 1966. NO synthase (NOS) was also detected in large quantities in β cells. Therefore, it can be inferred that L-arginine is likely to exert its insulin secretion-promoting effect after the conversion of NOS to NO. N-methyl-L-arginine (NMMA) is an NO inhibitor, and in the presence and absence of NMMA, researchers stimulated β cells with glucose and measured the concentration of NO in the extracellular culture medium. Glucose stimulated the release of NO from β cells in a concentration-dependent manner[57], but the mechanism underlying this effect is not clear at present. Based on this information, it can be concluded that glucose stimulates NO production in β cells in a concentration-dependent manner and that L-arginine and NOS are involved in NO production; however, the cytotoxicity of NO should not be ignored. When the glucose concentration exceeds a certain range, excess NO production in β cells reduces insulin release, potentially due to the cytotoxicity of NO. When β cells are stimulated with sodium nitroprusside (SNP, an extracellular NO donor) alone or both sodium nitroprusside and glucose, glucose is still the main factor that induces insulin secretion; NO alone does not induce insulin secretion but only enhances glucose-induced insulin secretion. The mechanism underlying this effect is not clear, but one hypothesis suggests that it may be related to NO-mediated stimulation of transient Ca2+ release[58].

The results of many studies have shown that NO exerts a negative effect on insulin secretion. Sodium nitroprusside inhibits glucose-induced insulin secretion, and hemoglobin, as a scavenger of NO, attenuates this inhibitory effect[59]. Due to the paradoxical effect of NO on glucose-induced insulin secretion, researchers studying islets have shifted their focus to changes in Ca2+ concentrations. Hemoglobin reduces the intracellular Ca2+ concentration in β cells, while high concentrations of NO reduce the intracellular Ca2+ concentration, which may explain the paradoxical effect of NO[60].

CONCLUSION

As an organ that regulates blood glucose levels in the body, the pancreas mainly functions by secreting various hormones. Insulin secreted by β cells is the only hormone that lowers blood glucose levels in the body, and neurotransmitters play an important role in β cell-mediated insulin; thus, the regulatory effect and mechanism of action of β cells are hot research topics. Insulin secretion mainly depends on the Ca2+ concentration in β cells, and when the glucose concentration increases, the concentration of ATP produced by glucose metabolism also increases accordingly. Glucose and other substances that produce ATP after being metabolized induce inwardly rectifying K+-ATP channel closing, subsequently leading to membrane depolarization[61]. Glucose produces a large action potential peak during β-cell depolarization, allowing voltage-sensitive Ca2+ channels to open and Ca2+ to enter the cell in large quantities to mediate insulin secretion[62].

Parasympathetic nerves play a predominant role in promoting insulin secretion, and ACh stimulates IP3 hydrolysis to increase the Ca2+ content in β cells. ACh enhances Ca2+-mediated insulin release via the PLC-PKC pathway, but the mechanism involved is unclear. Some studies also indicate that ACh affects the β-cell membrane potential and promotes β-cell membrane depolarization in the presence of glucose. GRP increases the intracellular Ca2+ concentration to stimulate insulin secretion from β cells. VIP and PACAP have similar mechanisms of action, binding to VIP-2 and PACAP-3 receptors, respectively, to increase cAMP production and stimulate insulin release from β cells. Sympathetic nerves mainly inhibit insulin secretion from β cells. NE promotes K+-ATP channel opening to inhibit cell membrane depolarization and reduce the inward flow of intracellular Ca2+, thus inhibiting insulin release from β cells. Both glycopeptide and NYP inhibit insulin secretion by β cells, and glycopeptide may block inward Ca2+ flow through an as-yet unknown mechanism; moreover, NPY inhibits insulin secretion from β cells by inhibiting cAMP signaling.

While some neurotransmitters are released solely from nerve terminals, many transmitters are released by β cells in conjunction with nerve terminals. 5-HT is synthesized by the conversion of tryptophan to the intermediate product 5-HTP by TPH and AADC followed by the conversion of 5-HTP to 5-HT. The generated 5-HT is released via VMAT1/2, and it functions as an autocrine and paracrine signal to promote insulin secretion. DA precursors (such as L-DOPA) are converted to DA by AADC-dependent enzymes, and as L-DOPA uptake and DA synthesis increase, a releasable DA pool is generated in β cells. DA is then cosecreted with insulin through VMAT2. This endogenous DA acts in an autocrine/paracrine manner on insulin-secreting cells that express D2-like receptors. When DA is bound, it activates the Gα subunit to negatively regulate cAMP production, thereby decreasing protein kinase A activity and inhibiting insulin secretion. In addition, a large amount of NOS exists in β cells, and L-arginine promotes insulin secretion after its conversion to NO. However, the effect of NO on insulin secretion is controversial, as studies reporting its effects in promoting and inhibiting insulin secretion have been published; this suggests that the functions of NO may be related to its concentration. NO does not promote insulin secretion from β cells alone, and NO plays a facilitating role only in the presence of glucose. However, due to the cytotoxicity of NO, high NO concentrations may damage β cells and affect glucose metabolism, leading to a decrease in insulin release.

In previous studies, the effects of neurotransmitters on insulin secretion were initially identified, and it was found that neurotransmitters mainly act during the last step of insulin release, i.e. on cytosolic insulin. The general direction of the sympathetic and parasympathetic effects on insulin secretion has been determined, but many unresolved questions remain. The current research can't answer these questions, but we hope that future researchers can investigate these directions: (1) Do differences in the release of peri-islet neurotransmitters exist under normoglycemic and hyperglycemic conditions? (2) Does glucose increase neurotransmitter secretion by stimulating nerve endings under hyperglycemic conditions? (3) Do any stimulatory conditions excite the peri-islet vagus nerve and promote insulin secretion by increasing neurotransmitter secretion? (4) Why does ACh promote insulin secretion for a short period but inhibit insulin secretion upon prolonged stimulation? (5) Do neurotransmitters originating from dual sources play a more important role in regulating insulin secretion than those from a single source (i.e. nerves)? (6) Are neurotransmitters involved in insulin synthesis and transport? (7) Are the concentrations of neurotransmitters in human blood altered by drugs used to regulate blood glucose levels? Do neurotransmitters affect the first and second phases of insulin secretion? (8) Does neurotransmitter release decrease in response to peri-islet nerve ending lesions during the development of diabetes mellitus? (9) Is the effect of sympathetic nerve radiofrequency ablation in alleviating type 2 diabetes related to neurotransmitters? And (10) Whether the brain itself affects insulin secretion through neurotransmitters?

ACKNOWLEDGEMENTS

We acknowledge all of the participants in our study and core facility of biomedical sciences, Xiamen University.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Endocrinology and metabolism

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B, B

Grade C (Good): 0

Grade D (Fair): D

Grade E (Poor): 0

P-Reviewer: Alkhatib AJ, Jordan; Mittal M, India; Nagamine T, Japan S-Editor: Yan JP L-Editor: Filipodia P-Editor: Yan JP

References
1.  Gilon P, Henquin JC. Mechanisms and physiological significance of the cholinergic control of pancreatic beta-cell function. Endocr Rev. 2001;22:565-604.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 181]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
2.  Henquin JC. Paracrine and autocrine control of insulin secretion in human islets: evidence and pending questions. Am J Physiol Endocrinol Metab. 2021;320:E78-E86.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 24]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
3.  Ahrén B, Taborsky GJ Jr, Porte D Jr. Neuropeptidergic versus cholinergic and adrenergic regulation of islet hormone secretion. Diabetologia. 1986;29:827-836.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 201]  [Cited by in F6Publishing: 216]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
4.  Buijs RM, Chun SJ, Niijima A, Romijn HJ, Nagai K. Parasympathetic and sympathetic control of the pancreas: a role for the suprachiasmatic nucleus and other hypothalamic centers that are involved in the regulation of food intake. J Comp Neurol. 2001;431:405-423.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Makhmutova M, Caicedo A. Optical Imaging of Pancreatic Innervation. Front Endocrinol (Lausanne). 2021;12:663022.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
6.  Hampton RF, Jimenez-Gonzalez M, Stanley SA. Unravelling innervation of pancreatic islets. Diabetologia. 2022;65:1069-1084.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 23]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
7.  Mateus Gonçalves L, Almaça J. Functional Characterization of the Human Islet Microvasculature Using Living Pancreas Slices. Front Endocrinol (Lausanne). 2020;11:602519.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
8.  Kaneto A, Kajinuma H, Kosaka K. Effect of splanchnic nerve stimulation on glucagon and insulin output in the dog. Endocrinology. 1975;96:143-150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 50]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
9.  Katschinski M. Nutritional implications of cephalic phase gastrointestinal responses. Appetite. 2000;34:189-196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 63]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
10.  Grise F, Taib N, Monterrat C, Lagrée V, Lang J. Distinct roles of the C2A and the C2B domain of the vesicular Ca2+ sensor synaptotagmin 9 in endocrine beta-cells. Biochem J. 2007;403:483-492.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 17]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
11.  Matthews DR, Clark A. Neural control of the endocrine pancreas. Proc Nutr Soc. 1987;46:89-95.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
12.  Wakiya T, Ishido K, Yoshizawa T, Kanda T, Hakamada K. Roles of the nervous system in pancreatic cancer. Ann Gastroenterol Surg. 2021;5:623-633.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
13.  Kim W, Fiori JL, Shin YK, Okun E, Kim JS, Rapp PR, Egan JM. Pancreatic polypeptide inhibits somatostatin secretion. FEBS Lett. 2014;588:3233-3239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 20]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
14.  MacDonald PE, Rorsman P. The ins and outs of secretion from pancreatic beta-cells: control of single-vesicle exo- and endocytosis. Physiology (Bethesda). 2007;22:113-121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 40]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
15.  Fendler B, Zhang M, Satin L, Bertram R. Synchronization of pancreatic islet oscillations by intrapancreatic ganglia: a modeling study. Biophys J. 2009;97:722-729.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 32]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
16.  Monstein HJ, Grahn N, Truedsson M, Ohlsson B. Progastrin-releasing peptide and gastrin-releasing peptide receptor mRNA expression in non-tumor tissues of the human gastrointestinal tract. World J Gastroenterol. 2006;12:2574-2578.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 4]  [Cited by in F6Publishing: 9]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
17.  Wahl MA, Landsbeck EA, Ammon HP, Verspohl EJ. Gastrin-releasing peptide: binding and functional studies in mouse pancreatic islets. Pancreas. 1992;7:345-351.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Pendharkar SA, Drury M, Walia M, Korc M, Petrov MS. Gastrin-Releasing Peptide and Glucose Metabolism Following Pancreatitis. Gastroenterology Res. 2017;10:224-234.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
19.  Pettersson M, Ahrén B. Gastrin releasing peptide (GRP): effects on basal and stimulated insulin and glucagon secretion in the mouse. Peptides. 1987;8:55-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 28]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
20.  Karpiesiuk A, Palus K. Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) in Physiological and Pathological Processes within the Gastrointestinal Tract: A Review. Int J Mol Sci. 2021;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
21.  Merech F, Soczewski E, Hauk V, Paparini D, Ramhorst R, Vota D, Pérez Leirós C. Vasoactive Intestinal Peptide induces glucose and neutral amino acid uptake through mTOR signalling in human cytotrophoblast cells. Sci Rep. 2019;9:17152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
22.  Papazoglou I, Lee JH, Cui Z, Li C, Fulgenzi G, Bahn YJ, Staniszewska-Goraczniak HM, Piñol RA, Hogue IB, Enquist LW, Krashes MJ, Rane SG. A distinct hypothalamus-to-β cell circuit modulates insulin secretion. Cell Metab. 2022;34:285-298.e7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 12]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
23.  Gyires K, Zádori ZS, Török T, Mátyus P. alpha(2)-Adrenoceptor subtypes-mediated physiological, pharmacological actions. Neurochem Int. 2009;55:447-453.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 87]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
24.  Moullé VS, Tremblay C, Castell AL, Vivot K, Ethier M, Fergusson G, Alquier T, Ghislain J, Poitout V. The autonomic nervous system regulates pancreatic β-cell proliferation in adult male rats. Am J Physiol Endocrinol Metab. 2019;317:E234-E243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 16]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
25.  Kurose T, Seino Y, Nishi S, Tsuji K, Taminato T, Tsuda K, Imura H. Mechanism of sympathetic neural regulation of insulin, somatostatin, and glucagon secretion. Am J Physiol. 1990;258:E220-E227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 22]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
26.  Pan M, Yang G, Cui X, Yang SN. Subthreshold α₂-adrenergic activation counteracts glucagon-like peptide-1 potentiation of glucose-stimulated insulin secretion. Exp Diabetes Res. 2011;2011:604989.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
27.  Kitaguchi T, Oya M, Wada Y, Tsuboi T, Miyawaki A. Extracellular calcium influx activates adenylate cyclase 1 and potentiates insulin secretion in MIN6 cells. Biochem J. 2013;450:365-373.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 60]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
28.  Guo LL, Shun XL, He B, Fang PH, Bo P, Zhu Y, Zhang ZW. Cooperation between galanin and insulin in facilitating glucose transporter 4 translocation in adipose cells of diabetic rats. J Biol Regul Homeost Agents. 2019;33:1327-1335.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Kuo WN, Hodgins DS, Kuo JF. Adenylate cyclase in islets of Langerhans. Isolation of islets and regulation of adenylate cyclase activity by various hormones and agents. J Biol Chem. 1973;248:2705-2711.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Ahrén B, Lindskog S. Galanin and the regulation of islet hormone secretion. Int J Pancreatol. 1992;11:147-160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 19]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
31.  Hohmann JG, Teklemichael DN, Weinshenker D, Wynick D, Clifton DK, Steiner RA. Obesity and endocrine dysfunction in mice with deletions of both neuropeptide Y and galanin. Mol Cell Biol. 2004;24:2978-2985.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 21]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
32.  Zorrilla EP, Brennan M, Sabino V, Lu X, Bartfai T. Galanin type 1 receptor knockout mice show altered responses to high-fat diet and glucose challenge. Physiol Behav. 2007;91:479-485.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 51]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
33.  Nishi S, Seino Y, Ishida H, Seno M, Taminato T, Sakurai H, Imura H. Vagal regulation of insulin, glucagon, and somatostatin secretion in vitro in the rat. J Clin Invest. 1987;79:1191-1196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 43]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
34.  Edwards AV, Bloom SR. Pancreatic endocrine responses to substance P and calcitonin gene-related peptide in conscious calves. Am J Physiol. 1994;267:E847-E852.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 4]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
35.  Hermansen K. Effects of substance P and other peptides on the release of somatostatin, insulin, and glucagon in vitro. Endocrinology. 1980;107:256-261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 50]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
36.  Chiba Y, Kawai K, Okuda Y, Munekata E, Yamashita K. Effects of substance P and substance P-(6-11) on hormone release from isolated perfused pancreas: their opposite actions on rat and canine islets. Endocrinology. 1985;117:1996-2000.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
37.  Schmidt PT, Tornøe K, Poulsen SS, Rasmussen TN, Holst JJ. Tachykinins in the porcine pancreas: potent exocrine and endocrine effects via NK-1 receptors. Pancreas. 2000;20:241-247.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
38.  Kim YG, Moon JH, Kim K, Kim H, Kim J, Jeong JS, Lee J, Kang S, Park JS. β-cell serotonin production is associated with female sex, old age, and diabetes-free condition. Biochem Biophys Res Commun. 2017;493:1197-1203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 5]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
39.  Bellono NW, Bayrer JR, Leitch DB, Castro J, Zhang C, O'Donnell TA, Brierley SM, Ingraham HA, Julius D. Enterochromaffin Cells Are Gut Chemosensors that Couple to Sensory Neural Pathways. Cell. 2017;170:185-198.e16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 453]  [Cited by in F6Publishing: 478]  [Article Influence: 68.3]  [Reference Citation Analysis (0)]
40.  Bonnin A, Goeden N, Chen K, Wilson ML, King J, Shih JC, Blakely RD, Deneris ES, Levitt P. A transient placental source of serotonin for the fetal forebrain. Nature. 2011;472:347-350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 368]  [Cited by in F6Publishing: 396]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
41.  Namkung J, Kim H, Park S. Peripheral Serotonin: a New Player in Systemic Energy Homeostasis. Mol Cells. 2015;38:1023-1028.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 74]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
42.  Kirchgessner AL, Gershon MD. Innervation of the pancreas by neurons in the gut. J Neurosci. 1990;10:1626-1642.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 134]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
43.  Cataldo Bascuñan LR, Lyons C, Bennet H, Artner I, Fex M. Serotonergic regulation of insulin secretion. Acta Physiol (Oxf). 2019;225:e13101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 31]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
44.  Kim K, Oh CM, Ohara-Imaizumi M, Park S, Namkung J, Yadav VK, Tamarina NA, Roe MW, Philipson LH, Karsenty G, Nagamatsu S, German MS, Kim H. Functional role of serotonin in insulin secretion in a diet-induced insulin-resistant state. Endocrinology. 2015;156:444-452.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 86]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
45.  Schraenen A, Lemaire K, de Faudeur G, Hendrickx N, Granvik M, Van Lommel L, Mallet J, Vodjdani G, Gilon P, Binart N, in't Veld P, Schuit F. Placental lactogens induce serotonin biosynthesis in a subset of mouse beta cells during pregnancy. Diabetologia. 2010;53:2589-2599.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 102]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
46.  Cataldo LR, Cortés VA, Mizgier ML, Aranda E, Mezzano D, Olmos P, Galgani JE, Suazo J, Santos JL. Fluoxetine impairs insulin secretion without modifying extracellular serotonin levels in MIN6 β-cells. Exp Clin Endocrinol Diabetes. 2015;123:473-478.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 13]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
47.  Makhmutova M, Weitz J, Tamayo A, Pereira E, Boulina M, Almaça J, Rodriguez-Diaz R, Caicedo A. Pancreatic β-Cells Communicate With Vagal Sensory Neurons. Gastroenterology. 2021;160:875-888.e11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 39]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
48.  Bennet H, Balhuizen A, Medina A, Dekker Nitert M, Ottosson Laakso E, Essén S, Spégel P, Storm P, Krus U, Wierup N, Fex M. Altered serotonin (5-HT) 1D and 2A receptor expression may contribute to defective insulin and glucagon secretion in human type 2 diabetes. Peptides. 2015;71:113-120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 66]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
49.  Fabrazzo M, Monteleone P, Prisco V, Perris F, Catapano F, Tortorella A, Monteleone AM, Steardo L, Maj M. Olanzapine Is Faster than Haloperidol in Inducing Metabolic Abnormalities in Schizophrenic and Bipolar Patients. Neuropsychobiology. 2015;72:29-36.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
50.  Farino ZJ, Morgenstern TJ, Maffei A, Quick M, De Solis AJ, Wiriyasermkul P, Freyberg RJ, Aslanoglou D, Sorisio D, Inbar BP, Free RB, Donthamsetti P, Mosharov EV, Kellendonk C, Schwartz GJ, Sibley DR, Schmauss C, Zeltser LM, Moore H, Harris PE, Javitch JA, Freyberg Z. New roles for dopamine D(2) and D(3) receptors in pancreatic beta cell insulin secretion. Mol Psychiatry. 2020;25:2070-2085.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 43]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
51.  Enjalbert A, Bockaert J. Pharmacological characterization of the D2 dopamine receptor negatively coupled with adenylate cyclase in rat anterior pituitary. Mol Pharmacol. 1983;23:576-584.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Hernandez-Lopez S, Tkatch T, Perez-Garci E, Galarraga E, Bargas J, Hamm H, Surmeier DJ. D2 dopamine receptors in striatal medium spiny neurons reduce L-type Ca2+ currents and excitability via a novel PLC[beta]1-IP3-calcineurin-signaling cascade. J Neurosci. 2000;20:8987-8995.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 357]  [Cited by in F6Publishing: 347]  [Article Influence: 15.1]  [Reference Citation Analysis (0)]
53.  Klein MO, Battagello DS, Cardoso AR, Hauser DN, Bittencourt JC, Correa RG. Dopamine: Functions, Signaling, and Association with Neurological Diseases. Cell Mol Neurobiol. 2019;39:31-59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 273]  [Cited by in F6Publishing: 420]  [Article Influence: 70.0]  [Reference Citation Analysis (0)]
54.   Nagamine T. Severe Hypoglycemia Associated with Tiapride in an Elderly Patient with Diabetes and Psychosis. Innov Clin Neurosci 2017; 14: 12–13. .  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Wei H, Zapata RC, Lopez-Valencia M, Aslanoglou D, Farino ZJ, Benner V, Osborn O, Freyberg Z, McCarthy MJ. Dopamine D(2) receptor signaling modulates pancreatic beta cell circadian rhythms. Psychoneuroendocrinology. 2020;113:104551.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
56.  Gheibi S, Ghasemi A. Insulin secretion: The nitric oxide controversy. EXCLI J. 2020;19:1227-1245.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
57.  Thompson B, Satin LS. Beta-Cell Ion Channels and Their Role in Regulating Insulin Secretion. Compr Physiol. 2021;11:1-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 8]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
58.  Dugbartey GJ. Carbon Monoxide in Pancreatic Islet Transplantation: A New Therapeutic Alternative to Patients With Severe Type 1 Diabetes Mellitus. Front Pharmacol. 2021;12:750816.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
59.  Tsuura Y, Ishida H, Hayashi S, Sakamoto K, Horie M, Seino Y. Nitric oxide opens ATP-sensitive K+ channels through suppression of phosphofructokinase activity and inhibits glucose-induced insulin release in pancreatic beta cells. J Gen Physiol. 1994;104:1079-1098.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 60]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
60.  Grapengiesser E, Gylfe E, Dansk H, Hellman B. Nitric oxide induces synchronous Ca2+ transients in pancreatic beta cells lacking contact. Pancreas. 2001;23:387-392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
61.  Wuttke A, Idevall-Hagren O, Tengholm A. P2Y₁ receptor-dependent diacylglycerol signaling microdomains in β cells promote insulin secretion. FASEB J. 2013;27:1610-1620.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 41]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
62.  Cook DL, Hales CN. Intracellular ATP directly blocks K+ channels in pancreatic B-cells. Nature. 1984;311:271-273.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 855]  [Cited by in F6Publishing: 865]  [Article Influence: 21.6]  [Reference Citation Analysis (0)]