Open Access

Cellular signaling pathways regulating β‑cell proliferation as a promising therapeutic target in the treatment of diabetes (Review)

  • Authors:
    • Wen‑Juan Jiang
    • Yun‑Chuan Peng
    • Kai‑Ming Yang
  • View Affiliations

  • Published online on: August 13, 2018     https://doi.org/10.3892/etm.2018.6603
  • Pages: 3275-3285
  • Copyright: © Jiang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

It is established that a decrease in β‑cell number and deficiency in the function of existing β‑cells contribute to type 1 and type 2 diabetes mellitus. Therefore, a major focus of current research is to identify novel methods of improving the number and function of β‑cells, so as to prevent and/or postpone the development of diabetes mellitus and potentially reverse diabetes mellitus. Based on prior knowledge of the above‑mentioned causes, promising therapeutic approaches may include direct transplantation of islets, implantation and subsequent induced differentiation of progenitors/stem cells to β‑cells, replication of pre‑existing β‑cells, or activation of endogenous β‑cell progenitors. More recently, with regards to cell replacement and regenerative treatment for diabetes patients, the identification of cellular signaling pathways with related genes or corresponding proteins involved in diabetes has become a topic of interest. However, the majority of pathways and molecules associated with β‑cells remain unresolved, and the specialized functions of known pathways remain unclear, particularly in humans. The current article has evaluated the progress of research on pivotal cellular signaling pathways involved with β‑cell proliferation and survival, and their validity for therapeutic adult β‑cell regeneration in diabetes. More efforts are required to elucidate the cellular events involved in human β‑cell proliferation in terms of the underlying mechanisms and functions.

Introduction

Due to an increasing incidence of diabetes and its complications, there is an urgent need to identify therapeutic approaches for diabetes. Both type 1 and type 2 diabetes patients eventually require lifelong insulin replacement therapy. However, long-term exogenous insulin replacement therapy is not only associated with numerous side effects, but can also not delay and/or prevent the development of diabetes and the emergence of its complications. In addition, islet transplantation has been studied but progress in this field is slow due to a lack of islet donors, organ transplant rejection and low survival rates following transplantation in humans (1). Therefore, the realization of endogenous islet cells regeneration has become the focus of all researchers. At present, a fundamental method of treating diabetes is regenerative therapy, which generally involves the development of feasible therapeutic methods that induce an expansion of adult human β-cells (2). In previous studies, a number of mechanisms had been proposed to explain the proliferation of β-cells, including the proliferation of pancreatic duct stem cells (3), the replication of β-cell progenitor cells (4) and the endogenesis of residual β-cells (5). Insight into the network of β-cell signaling pathways that promote β-cell proliferation may help to overcome key difficulties in diabetes therapy. As research methods for the evaluation of cellular signaling pathways have improved, an increasing number of pathways have been identified that are associated with β-cell proliferation, including insulin, growth factors and hormones. However, difficulties remain in various aspects of diabetes research, including specific molecular mechanisms remaining unclear and differences between animal and human trials. Despite these limitations, more research into the related signal pathways may aid to develop therapeutic strategies for the treatment of diabetes.

Insulin signaling pathways

Insulin signal transduction pathways within pancreatic β-cells are a complex system, and are mainly composed of insulin receptors (IR), including the insulin receptor (IR)-related receptor and insulin-like growth factor 1 (IGF-1) receptor, and substrates belonging to the insulin receptor substrates (IRS) family, including IRS, IR tyrosine kinase, phosphoinositide linositol-3-kinase (PI3K) and its downstream effectors such as protein kinase B [PKB; also known as serine/threonine kinase (AKT)], mitogen activated protein kinase (MAPK), MAPK kinase (MAPKK/MEK), raf protein and rat sarcomas (Ras) proteins (small G proteins) (68). Activated IR stimulates IRS, which is present in two major forms in β-cells; IRS-1 and IRS-2 (9). IRS-1 is generally distributed in islet β-cells, and closely regulates glucose-dependent insulin secretion (9). By contrast, IRS-2 is mainly located in the peripheral region of the pancreatic islets and at low levels in β-cells, and positively moderates the compensation mechanism of β-cells (9). In terms of regulatory mechanisms, IRS-1 is activated through the activation of PI3K and calcium channels, among others, while IRS-2 is activated by PI3K, MAPK, Ras and other pathways, and they ultimately modulate the growth, proliferation and mitosis of β-cells (10,11).

PI3K-AKT/PKB signaling pathway

The PI3K signaling pathway serves a dominant role in the regulation of β-cell function (12), which is orchestrated by an intricate network of mediators. AKT/PKB, acting as a downstream target of PI3K, regulates the proliferation of β-cells through modulation of its multiple downstream genes, which include Forkhead box protein O1 (FOXO1) (13), glycogen synthase kinase-3 (GSK3) (14) and mammalian target of rapamycin (mTOR) (8), among others.

FOXO1 is a member of the phosphorylase Fox gene family and its transcription activity is modulated through phosphorylation of PI3K/AKT signaling proteins (15). In β-cells, FOXO1 has been identified as a weak inhibitor of pancreatic and duodenal homeobox 1 (Pdx1) (15), which participated in the maintenance of common features of β-cells, including the regulation of target gene expression for genes such as glucose transporter 2 (GLUT2), insulin secretion and maintenance of appropriate cell function (16). This indicates the pivotal role of FOXO1 in the maintenance of β-cell function through modulation of Pdx1.

GSK3 has been demonstrated to be a downstream target of multiple signaling pathways and is associated with cell proliferation and cell cycle regulation (17). It has been reported that stimulation of GSK3 negatively regulates β-cell function by maintaining the sub-cellular localization and stability of Pdx1 (18). Furthermore, upon activation of mTOR, GSK3 is suppressed by FOXO1, which is activated through phosphorylated PI3K-AKT (Fig. 1) (19). mTOR, as a member of the serine/threonine protein kinase family, is a fundamental component within two mTOR complexes, mTORC1 and mTORC2 (2022). The related signaling proteins of mTOR include PI3K-AKT, AMP activated protein kinase and growth factors, which influence multiple metabolic indices such as cellular metabolism, stem cell differentiation and dedifferentiation, pancreatic β-cell function, insulin secretion and resistance and cellular survival or death (2123). Maiese (24) reported that downregulation of mTOR and mTORC1 activated PI3K-AKT through a feedback mechanism, which blocked all anticipated clinical benefits. PI3K-AKT suppressed activation of tuberous sclerosis complex 1/2 (TSC1/2), which was an upstream negative regulator of mTORC1, and then S6K1 and 4E-BPs promoted mRNA translation and synthesis of ribosomes, and eventually increased β-cell proliferation (Fig. 1) (7,8). Furthermore, P21cip1 and p27kip1 were inhibitors of the cell cycle (Fig. 1) (7). PI3K-AKT suppressed P21cip1 by activating MDM2/P53 gene, and increased β-cell proliferation, while it enhanced P27kip1 by activating FOXO1 gene, and hindered β-cell proliferation (Fig. 1) (7). A recent study demonstrated that the immortalized mouse β-cell line (exclusively expressing IR A isoform) was sensitive to the mitogenic response induced by IGF-1, which further activated β-cell proliferation, primarily through mTORC1 (Fig. 1) (25). Furthermore, in partial pancreatectomy (60%), mTORC1 exerted a compensatory effect on β-cell mass mainly through the cyclin D2 pathway in early stages (26).

Figure 1.

Binding of PDGF, GLP-1, IGF-1 and BTC to their respective receptors serves an important role in β-cell proliferation. The Wnt pathway regulates GSK3β to inhibit the phosphorylation of β-catenin and thus regulate the expression of Tcf7L2 and Cyclin D1-2, cMyc and cdk4, which ultimately modulates proliferation. PDGF binds to PDGFR and then activates the ERK1/2 pathway, increasing the expression of EZH2. IGF-1 binds to IGF-1R, and BTC binding to ErbB1 and ErbB2 also activates the IRS/PI3K pathway. GLP-1 binds to GLP-1R to activate cAMP-PKA, and it also acts via ADAM proteins to trigger the secretion of BTC that acts upon ErbB1/2. Circles of the same color represent signals within the same pathway. Red and black lines indicate inhibition and promotion, respectively. PDGF, platelet-derived growth factor; PDGFR, platelet-derived growth factor receptor; GLP-1, glucagon like peptide 1; GLP-1R, glucagon like peptide 1 receptor; IGF-1, insulin-like growth factor 1; IGF-1R, insulin-like growth factor 1 receptor; BTC, β-catenin; Dsh, disheveled; GSK3β, glycogen synthase kinase-3β; APC, adenomatous polyposis colis; cdk, cyclin-dependent kinase; PK, protein kinase; MEK, mitogen-activated protein kinase kinase; EZH2, enhancer of zeste homolog 2; EGFR, epidermal growth factor receptor; IRS, insulin receptor substrate; PI3K, phosphoinositide 3-kinase; PDK1, phosphoinositide-dependent kinase 1; TSC1/2, tuberous sclerosis complex 1/2; Rheb, Ras homolog enriched in brain; mTORC, mechanistic target of rapamycin complex; 4E-BP, eukaryotic translation initiation factor 4E-binding protein 1; S6K1, ribosomal S6 kinase 1; MDM2, mouse double minute 2 homolog; FOXO1, forkhead box protein O1; Pdx1, pancreatic and duodenal homeobox 1; P, phosphorylated.

Recently, studies have focused on the mechanism behind inulin-mediated fat and glucose metabolism, though few have investigated protein metabolism (13). For instance, it has been reported that hyperglycemia promoted compensatory β-cell proliferation to meet the insulin demand induced by hyperglycemia (27). Lipid metabolism has previously been investigated in pregnant female mice (10), whereby mice were randomly assigned to receive either a high-fat diet (MO-HF) or standard chow (MO-SC) without advance preparation. It was observed that MO-HF induced a failure in β-cell proliferation and survival by downregulating IRS1, PI3K and GLUT2 protein, while upregulating insulin protein and FOXO1 when compared with MO-SC mice (10,27). Furthermore, a high-fat diet has been demonstrated to negatively impact on β-cell proliferation, leading to the development of insulin resistance and D2M characteristics (28,29). Rhee et al (30) also suggested that preadipocyte factor 1 (Pref-1) may independently stimulate insulin secretion via AKT-Rab43 (Fig. 2). Finally, there has been dispute regarding the interplay between protein metabolism and the related signaling pathways of β-cell proliferation. Chen et al (13) argued that FOXO1 may inhibit the expression of albumin, while other proteins such as phycocyanin have been demonstrated to promote β-cell proliferation by regulating PI3K-AKT signaling and downstream FOXO1, and ameliorate diabetes mice by stimulating glucokinase expression and insulin signaling in the pancreas (11,31). It has also been reported that Exendin-4 may enhance β-cell proliferation in some instances by stimulating PI3K-AKT signaling, potentially via an intermediate ligand-binding activation step involving corresponding receptors (14).

Calcium-mediated signaling pathway

As an important second messenger, calcium participates in apoptosis and regulates the synthesis of enzymes and hormones, including that of insulin. The maintenance of normal Ca2+ concentration in the body is termed calcium homeostasis, and is critical to cell survival (32). Previous results have indicated that disruption of calcium homeostasis may accelerate β-cell death and lead to type 1 diabetes mellitus (D1M) (32). Furthermore, PI3K signaling stimulated the autocrine function of pancreatic β-cells to moderate β-cell proliferation by inhibiting endoplasmic reticulum Ca2+ ATPase, which increased the intracellular Ca2+ concentration and subsequently activated calcium-mediated signaling pathways (33,34). In calcium-mediated signaling, increases in intracellular calcium activate calmodulin (CaM), which has two substrates, namely carbohydrate response element-binding (CREB)-regulated transcription coactivator-2 (CRTC2) and transcription factors of the nuclear factor of activated T cells (NFAT) family (3538). Notably, dephosphorylated NFAT translocated to the nucleus and combined with the promoters of cell-cycle activating agents, including cyclin A and D, cMyc, cyclin-dependent kinase (cdk) 2 and 4, FOX protein M1 (FOXM1; Fig. 2) and the promoter of IRS-2, which participated in PI3K-AKT and Ras-Raf-MAPK signaling to activate β-cell proliferation (3537). By contrast, NFAT function may be improved by interaction with transcription factors, including CREB, cAMP-response element modulator and activator protein 1 (38). Changes in cytosolic Ca2+ concentration have been documented to exert a significant effect on insulin secretion by modulating gene expression from electrically excitable pancreatic β-cells (39). In pancreatic β-cells, intracellular high calcium concentration triggers exocytosis and leads to insulin secretion (40). In addition, previous data suggest that Ca2+ may be drawn into pancreatic islets either by the opening L-type voltage-dependent Ca2+ channels or crosstalk between intracellular Ca2+ concentration and protein kinase (PK) A/L/C to stimulate insulin secretion (40,41). Indeed, Ca2+/CaM-dependent protein kinase 2 (CaMKK2) is considered to be a molecular regulator of insulin function, and Ca2+/CaM/CaMKK2 has been documented to serve a crucial role in the modulation of holistic body metabolism (42,43). Collectively these observations indicate that calcium-mediated signaling pathways may be therapeutic targets for the modulation of β-cell proliferation in diabetes.

RAS-extracellular signal-regulated kinase (ERK)/MAPK signaling pathway

Three types of Ras protein are expressed in humans, namely H-Ras, K-Ras, and N-Ras. K-Ras is considered to be the most important form, acting as a downstream target of multiple cellular signaling pathways that regulate cell growth, proliferation and differentiation (44). Chamberlain et al (45) demonstrated that K-Ras stimulated the anti-proliferative ras association domain family 1A (Rasf1a) pathway, which diminished cell proliferation (Fig. 2). Therefore, K-Ras may serve a negative regulatory role in the proliferation of β-cells, and downregulation of the K-Ras gene may enhance proliferation by reducing Rassf1a activity.

ERKs (also known as classical MAPKs), are diffusely expressed cellular signaling molecules that participate in the modulation of cell proliferation (46). In pancreatic β-cells, ERK1 (MAPK3) and ERK2 (MAPK1) have been documented to be the major expressed forms of ERK (5). In general, the binding of a ligand to its cognate tyrosine kinase or G protein coupled receptor stimulates the small GTPase Ras, resulting in the formation of activated Ras-GTP, which phosphorylates c-Raf and subsequently activates MEK, and ultimately activates ERK1/2 (5). Furthermore, a number of growth factors and hormones have been reported to induce β-cell proliferation via the activation of ERK1/2. For instance, it was observed that stimulation of platelet derived growth factor receptor required activated ERK1/2, leading to upregulation of the polycomb group protein EZH2 (enhancer of zeste homolog 2, EZH2), inhibition of P16LINK4 tumor suppressor protein and ultimately adult β-cell expansion (Fig. 1) (47). ERK signaling was also demonstrated to serve a pivotal role in 3,5,3′-Triidothyronine (T3)-induced β-cell proliferation (Fig. 2) (46). In addition, reactive oxygen species-facilitated β-cell differentiation by regulating the ERK1/2 pathway (48), and the anti-apoptotic function of visfatin was mediated by stimulation of the ERK1/2 signaling pathway (49). In a study by Ozaki et al (50), a mouse model of diabetes was established in high-fat diet KKAy mice and leptin receptor-deficient (db/db) mice. In these mice it was observed that obesity-associated insulin resistance could be treated with an inhibitor of MEK (50), indicating that regulation of ERK signaling may be an effective treatment for type 2 diabetes (D2M).

Growth factor signaling pathways

Several growth factors have been implicated in the modulation of Β-cell proliferation, including IGF-1, members of the epidermal growth factor (EGF) family, nerve growth factor (NGF), members of the transforming growth factor (TGF)-β superfamily, placental growth factor (PLGF) and their corresponding receptors (IGF-1R, EGFR, NGFR, TGFβR and PLGFR, respectively) (51). These mitogen signals have been associated with multiple downstream pathways, including PI3K-AKT-mTOR, ERK (Figs. 1 and 2) and janus kinase-signal transducers and activators of transcription (JAK-STAT), in the modulation of β-cell proliferation and survival (51).

EGF family signaling pathway

Multiple downstream proteins of the EGF family, including EGF, β-cellulin (BTC), TGF-α, heparin-binding (HB)-EGF and epiregnin, have been implicated in β-cell proliferation (52). For instance, BTC-dependent mitogenic signals were involved in the upregulation of IRS and activation of both EGF/ErbB1 and ErbB2 receptors in β-cells (Fig. 1) (53). BTC has also been linked to the IRS-PI3K signaling pathway, which signals through 3′-phosphoinositide-dependent protein kinase 1 (PDK1) to regulate AKT and FOXO1 (Fig. 1) (53). As members of the EGF superfamily, EGF and TGF-α have been reported to transiently activate ERK1/2, though the interaction was not strong enough to induce β-cell proliferation (53). Furthermore, constitutively active EGFR protected β-cells against damage and even death (54). It has also been reported that excessive nutrients may promote β-cell expansion via crosstalk between EGFR signaling, FOXM1-mediated cell proliferation and mTOR activation (55). Further study into the mechanism of β-cell survival has demonstrated that β-cell proliferation may be regulated via EGF and Raf-1/MEK/ERK signaling, which primarily increased the half-life of β-cells (51).

NGF signaling pathway

NGF regulates the development of neuronal cells and may control the function of non-neuronal cells (56,57). In islet β-cells, NGF is among the peptides stored by β-cells within secretary vesicles, and it may interact with β-cell receptors to trigger autocrine and paracrine effects in response to appropriate stimulation (57). NGF primarily acts via two membrane receptors, p75 neurotrophin receptor (P75 NTR) and tropomyosin receptor kinase A (trkA). trkA functions upon its phosphorylation, and mainly modulates the activation of signals involved in cell survival, differentiation and proliferation (58), while P75 NTR recruits specific intracellular effectors depending on its NGF-bound and unbound forms (59). A recent study suggested that islet NGF served a pivotal role in fine-tuning insulin secretion, to maintain both basal insulin secretion and biphasic secretion of glucose (60).

TGFβ superfamily signaling pathway

Various members of the TGFβ superfamily, including TGFβ, inhibins, activins, follistains, mullerian inhibitor substance and bone morphogenetic proteins, have been demonstrated to function in β-cells (5), though the exact underlying mechanisms remain unclear. The TGFβ superfamily, which has been implicated in numerous developmental processes, including proliferation, differentiation and growth arrest, served a crucial role in modulating pancreatic endocrine development and maturation (6164). In particular, interplay between the inhibitor TGFβ signaling Smad7 and intracellular mediators of TGFβ signaling were involved in coordinating this process (61). A recent investigation into TGFβ signaling demonstrated that macrophages released TGFβ1 to trigger Smad7 expression, which then facilitated β-cell proliferation (65). Smad7 was also increased in β-cells after partial pancreatic duct ligation (PDL) (65). In general, the functions of TGFβ superfamily members affects downstream Smad proteins in β-cells, which typically suppresses proliferation (64). Therefore, suppression of TGFβ signaling may promote β-cell proliferation (6570). In cultured human pancreatic duct cells, TGFβ1 induced the epithelial-mesenchymal transition and inhibited differentiation into β-cells, and bound with the downstream targets ERK1/2, AKT and Ras (71). The underlying mechanisms of pancreatic islet cell expansion may involve an upregulation in TGFβ signaling (68). Blockade of TGFβ signaling with short hairpin RNA (shRNA) against TGFβ1R, such as Alk5 shRNA, suppressed the differentiation and proliferation of β-cell-derived (BCD) cells, which may be associated with the AKT-FOXO1 pathway (Fig. 2) (72).

PLGF and Pref-1 signaling pathway

Finally, gestational PLGF may stimulate endothelial cells within the pancreatic islets to release growth factors that activate the PI3K-AKT pathway (Fig. 2), thus enhancing β-cell proliferation (73). A previous study demonstrated that Pref-1 independently facilitated the phosphorylation of AKT and ERK1/2, and triggered alterations in the expression of FOXO1 and Pdx1 (Fig. 2) (29).

Hormone signaling pathways

Hormones including thyroid hormone (TH), growth hormone (GH), glucagon like peptide 1 (GLP-1), IGF-1, and prolactin (PRL) and their cognate receptors (TR, GHR, GLP-1R, IGF-1R and PRLR, respectively) have been critically associated with β-cell survival, growth, proliferation, differentiation and insulin secretion, and may be involved in PI3K-AKT, JAK-STAT and ERK crosstalk (74). A stimulatory effect of T3 on cellular growth was observed on TRs in several cell lines. TR expression in the pancreas suggested that pancreatic β-cell proliferation might be induced by T3 (46). The study demonstrated that T3 induced pancreatic β-cell proliferation via the ERK signaling pathway (46). The current review has mainly focused on the incretin hormone GLP-1, which is primarily released from intestinal L-cells in response to hormones, nutrients and neurons, and may serve a significant role in β-cell proliferation and insulin secretion (75,76). In general, GLP-1 combined with its receptor (GLP-1R) may enhance insulin secretion. However, it has been reported that activation of GLP-1R requires a host of complex proteins interactions (77). Recent studies have identified multiple novel GLP-1R interactors, including the Ras superfamily proteins Rab5b and Rab5c, ATPase H+-transporting lysosomal accessory protein 2 (ATO6Ap2) and progesterone receptor membrane component 1, which served dual roles by regulating both GLP-1R signaling and insulin secretion through EGF receptor and cAMP signaling (77,78). In β-cells, GLP-1 might activate various signaling pathways, including cAMP-PKA, PI3K-AKT, ERK1/2 and Wnt (Fig. 1) (79,80). In a study of the hyperglycemic response, GLP-1 reinforced glucose-stimulated insulin secretion principally through activation of cAMP-PKA signaling in the β-cell line INS-1E (81). GLP-1 binding to GLP-1R has been reported to activate cAMP-PKA, and via ADAM proteins GLP-1 triggered the secretion of BTC, which acted upon ErbB1/2 (Fig. 1) (53). Furthermore, a recent report suggested that geniposide may enhance insulin secretion by activating GLP-1R in tandem with adenylyl cyclase (AC)/cAMP signaling (82). Yang et al (83) demonstrated that the peroxisome proliferator-activated receptor β/δ agonist, GW501516 (GW), in tandem with its transcriptional modulation of GLP-1R, may protect against lipotoxic apopotosis in β-cells. In addition, conditional chicken ovalbumin upstream promoter transcription factor II, as a novel mediator of GLP-1 signaling required for GLP-1 activation, depended on β-catenin signaling, and its expression was regulated by TCF7L2, which increased β-cell mass during the neonatal stage (84).

The present review has described a number of key hormone pathways implicated in β-cell proliferation, though further research is required to elucidate the full involvement of hormones in β-cell proliferation.

Wnt signaling pathways

The wnt signaling pathway is generally composed of extracellular wnt ligands, disheveled protein, Frizzled receptor, β-catenin, axin, GSK3β and adenomatous polyposis colis protein (Fig. 1) (85,86). The wnt pathway may be divided into three types: A canonical wnt/β-catenin pathway, a planar cell polarity (PCP) pathway and a non-canonical pathway, which includes wnt/Ca2+, activated phospholipase C, PKC and small GTPase (87,88).

In terms of function, wnt signaling molecules have been demonstrated to influence pancreatic endocrine development and regulate the postnatal β-cell functions of insulin secretion, proliferation, survival and differentiation (89). Bader et al (90) concluded that wnt/PCP genes were responsible for functional β-cell heterogeneity and induced β-cell maturation. Fltp, as a wnt/PCP effector and firefly luciferase, is considered to be a marker gene that may aid to elucidate the molecular mechanisms of islet cell plasticity and heterogeneity, and may be a target in endocrine cells for functional β-cell regeneration in diabetes (91). Wnt4, a regulator of β-cell proliferation, has been documented to primarily regulate canonical wnt signaling (85,86). Krutzfeldt et al (91) demonstrated that the expression and upregulation of wnt4 was common in the β-cells of obese mice. Furthermore, stimulation with exendin-4 enhanced wnt4 expression in β-cells (87). It has also been reported that wnt3a modulated the proliferation of β-cells and enhanced β-cell function by activating the wnt/β-catenin pathway, indicating crosstalk with PI3K signaling (92). Rulifson et al (93) observed that wnt3a induced the expression of Cdk4 and cyclin D-1 and −2, and stimulated enhanced β-cell proliferation in vitro. Regarding stem cells, canonical wnt signaling facilitated stem cell proliferation and survival via the expression of β-catenin-related downstream targets, such as cMyc, which was conducive to the transplantation of pluripotent stem cells in D1M (9496). Afelik et al (97) also documented that wnt7b was a modulator of pancreatic progenitor cell development. T cell factor 7-like 2 (TCF7L2) has been identified as a significant component of wnt/β-catenin signaling (Fig. 1), and a potent factor in the pathogenesis and progression of D2M (98). Notably, the dominant-negative form or ‘at-risk’ alleles of TCF7L2 served a deleterious role in β-cell proliferation and insulin secretion (99). Furthermore, the functional consequences of reduced high-mobility group protein B1 in BCD (INS1) and human colon cancer (HCT116) cells were reductions in TCF7L2 mRNA expression, TCF7L2 transcriptional regulatory activity and glucose triggered insulin secretion (100). A recent study also documented that geniposide promoted β-cell proliferation and survival by modulating the β-catenin/TCF7L2 signaling pathway (101).

JAK-STAT signaling pathways

JAK-STAT is a relatively novel signaling pathway, which has been associated with multiple cytokines, such as interleukin (IL)-1, −2 and −6, and hormones such as leptin, GH, placental lactogens, erythropoietin (EPO) and PRL (Fig. 3) (102105). JAK-STAT participates in the regulation of key biological processes, including cell proliferation, differentiation and apoptosis (102). Furthermore, JAKs may serve as a fundamental switch that must be activated through a cytokine to trigger STAT proteins, which enables downstream target activation and subsequent regulation of target gene transcription (106). It has been reported that activated STAT proteins may also trigger suppressors of cytokine signaling (SOCS), protein repressors of activated STAT and transcription of cytokine-inducible SH2 domain-containing proteins (CISH; Fig. 3) (107,108). SOCS proteins are considered to be pivotal negative modulators of JAK-STAT signaling by terminating its upstream signals, of which four-SOC1, SOC2, SOC3 and CISH have been reported to serve as primary factors in the JAK-STAT pathway (106,109).

The progressive loss of pancreatic β-cells leads to D1M, which is closely associated with autoimmune assault (110). During the inflammatory progressive stage, overexpression of JAK-STAT molecules in pancreatic islets has been reported to contribute to β-cell dysfunction (111). A recent study documented that BRD0476, a novel inhibitor of β-cell apoptosis, impeded inteferon-γ-induced JAK2 and STAT1 signaling to facilitate β-cell survival (112). Unlike general JAK-STAT pathway suppressors, BRD0476 blocked JAK-STAT signaling pathways via ubiquitin-specific peptidase 9X (USP9X) without inhibiting the kinase function of any JAK (112). With regard to PRL, it has been observed that human β-cells failed to expand in response to PRL, potentially due to a deficiency of PRL receptors, among other explanations (5). Notably, a failure of β-cell expansion could be overcome by overexpression of murine STAT5a, due to the resulting upregulation and nuclear translocation of cdk4 and cyclins D1-3 (Fig. 3). This STAT5 signaling has been linked with β-cell cycle activity (113). Furthermore, EPO, acting via the EPO receptor, has been reported to stimulate β-cell proliferation through JAK2-STAT5 (Fig. 3) (114). De Groef et al (115) demonstrated that PDL stimulated the expression of multiple cytokines that served as positive ligands of STAT3 signaling in β-cells. In addition, β-cell cycling was enhanced by activating STAT3 with IL-6 (Fig. 3). STAT3 played an important role in maintaining the stability of β-cells by regulating the cell cycle and protecting against DNA damage (115).

TLR4 signaling pathway

Toll like receptor 4 (TLR4) is principally expressed in the innate immune system, and is considered to be involved in DM (116). In D2M, the expression of TLR4 may increase due to activation of the nuclear factor-κB (NF-κB) signaling pathway. While NF-κB has not been associated with the inflammatory reaction in diabetes, it may have an important influence on glucose metabolism and insulin reaction. NF-kB binds to the promoters of innate immune genes, particularly those of pro-inflammatory cytokines, including tumor necrosis factor-α (TNF-α) and IL-1β, which is considered to exert positive feedback on TLR4 expression (117119). Sepehri et al (119) reported that TLR4 was upregulated in the peripheral blood mononuclear cells of patients with D2M patients, which was independent of sex, age, body mass index and blood sugar content. Furthermore, TLR4 is considered to be a significant candidate in the related complications of D2M, including nephropathy (120) and atherosclerosis (121).

Conclusions and perspectives

Cellular signaling pathways are substantial targets, and are capable of stimulating β-cell proliferation, as demonstrated in various islets and β-cell systems. As such, they may be useful in future treatment strategies for diabetes. Cellular signaling pathways involving interactions with ligands, receptors and the proliferative machinery of β-cells are complex systems. The present review evaluated the potential underlying mechanisms of insulin, growth factor, Wnt, JAK-STAT and TLR4 signaling. While various cellular signaling mechanisms were discussed, not all signaling pathways implicated in β-cell proliferation were included, such as those involving islet cell autoantigen-512, inhibin, activin, muscarinic and octeocalicn.

Cellular signaling molecules and their downstream targets form an intricate network, which may directly or indirectly regulate β-cell proliferation and survival. While an increased number of studies have investigated cellular signaling, difficulties remain in their applications. For instance, the expression of cellular signals in β-cells varies among different species, ages and tissues. Furthermore, various cellular signals have not been investigated, and their specific functions and mechanisms within β-cells remain unclear and disputed by researchers. Though notably, excessive activation of signaling pathways may have negative impacts on β-cells.

In conclusion, multiple other therapeutic strategies may be used for the treatment of diabetes, including stem cell differentiation induction and islet transplantation. Cellular signaling pathways may have untapped potential in the induction of β-cell proliferation. Identification of appropriate molecular targets within these pathways may aid to develop novel strategies in the treatment of diabetes and improve the outcome for patients.

Acknowledgements

Not applicable.

Funding

The present work was supported by the Yunnan Province Project Education Fund (grant no. 2015J117).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

WJJ wrote the manuscript, collected images and analyzed the data; YCP processed the images; KMY analyzed the data, processed the images and revised the manuscript. All authors have read and approved the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Farney AC, Sutherland DE and Opara EC: Evolution of islet transplantation for the last 30 years. Pancreas. 1:8–20. 2016. View Article : Google Scholar

2 

Tiwari S, Roel C, Wills R, Casinelli G, Tanwir M, Takane KK and Fiaschi-Taesch NM: Early and late G1/S cyclins and Cdks act complementarily to enhance authentic human β-cell proliferation and expansion. Diabetes. 10:3485–3498. 2015. View Article : Google Scholar

3 

Wang GS, Rosenberg L and Scott FW: Tubular complexes as a source for islet neogenesis in the pancreas of diabetes-prone BB rats. Lab Invest. 5:675–688. 2005. View Article : Google Scholar

4 

Dor Y, Brown J, Martinez OI and Melton DA: Adult pancreatic beta-cells are formed by self-duplication rather than stem-cell differentiation. Nautre. 429:41–46. 2004. View Article : Google Scholar

5 

Stewart AF, Hussain MA, Garcia-Ocana A, Vasavada RC, Bhunshan A, Bernal-Mizrachi E and Kulkarni RN: Human β-cell proliferation and intracellular signaling: Part 3. Diabetes. 4:1872–1885. 2015. View Article : Google Scholar

6 

Saltiel AR and Kahn CR: Insulin signaling and the regulation of glucose and lipid metabolism. Nature. 414:799–806. 2001. View Article : Google Scholar : PubMed/NCBI

7 

Kulkarni RN, Mizrachi EB, Ocana AG and Stewart AF: Human β-cell proliferation and intracellular signaling: Driving in the dark without a road map. Diabetes. 9:2205–2213. 2012. View Article : Google Scholar

8 

Pende M, Kozma SC, Jaquet M, Oorschot V, Burcelin R, Le Marchand-Brustel Y, Kluperman J, Thorens B and Thomas G: Hypoinsulinaemia, glucose intolerance and diminished beta-cell size in S6K1-deficient mice. Nature. 408:994–997. 2000. View Article : Google Scholar : PubMed/NCBI

9 

Arajuo EP, Amaral ME, Souza CT, Bordin S, Ferreira F, Saad MJ, Boschero AC, Maqalhaes EC and Velloso LA: Blockade of IRS1 in isolated rat pancreatic islets improves glucose-induced insulin secretion. FEBS Lett. 531:437–442. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Bringhenti I, Ornellas F, Mandarim-de-Lacerda CA and Aguila MB: The insulin-signaling pathway of the pancreatic islet is impaired in adult mice offspring of mothers fed a high-fat diet. Nutrition. 32:1138–1143. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Ou Y, Ren Z, Wang J and Yang X: Phycocyanin ameliorates alloxan-induced diabetes mellitus in mice: Involved in insulin signaling pathway and GK expression. Chem Biol Interact. 247:49–54. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Kaneko K, Ueki K, Takahashi N, Hashimoto S, Okamoto M, Awazawa M, Okazaki Y, Ohsugi M, Inabe K, Umehara T, et al: Class IA phosphatidylinositol 3-kinase in pancreatic β cells controls insulin secretion by multiple mechanisms. Cell Metab. 12:619–632. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Chen Q, Lu M, Monks BR and Birnabaum MJ: Insulin is required to maintain albumin expression by inhibiting forkhead box o1 protein. J Biol Chem. 291:2371–2378. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Wang C, Chen X, Ding X, He Y, Gu C and Zhou L: Exendin-4 promotes beta cell proliferation via PI3K/Akt signaling pathway. Cell Physiol Biochem. 35:2223–2232. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Katoh M and Katoh H: Human FOX gene family (Review). Int J Oncol. 25:1495–1500. 2004.PubMed/NCBI

16 

Seyer P, Vallois D, Poitry-Yamate C, Schutz F, Metref S, Tarussio D, Maechler P, Staels B, Lanz B, Grueter R, et al: Hepatic glucose sensing is required to preserve β cell glucose competence. J Clin Invest. 123:1662–1676. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Huang L, Jiang X, Gong L and Xing D: Photoactivation of Akt1/GSK3β Isoform-Specific signaling axis promotes pancreatic β-cell regeneration. J Cell Biochem. 116:1741–1754. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Boucher MJ, Selander L, Carlsson L and Edlund H: Phosphorylation marks IPF1/PDX1 protein for degradation by glycogen synthase kinase 3-dependent mechanisms. J Biol Chem. 281:6395–6403. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Morral N: Novel targets and therapeutic strategies for type 2 diabetes. Trends Endocrinol Metab. 14:169–175. 2003. View Article : Google Scholar : PubMed/NCBI

20 

Foster KG and Fingar DC: Mammalian target of rapamycin (mTOR): Conducting the cellular signaling symphony. J Biol Chem. 285:14071–14077. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Duzgn Z, Eroglu Z and Biray-Avci C: Role of mTOR in glioblastoma. Gene. 575:187–190. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Maiese K: Novel nervous and multi-system regenerative therapeutic strategies for diabetes mellitus with mTOR. Neural Regen Res. 11:372–385. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Wang J, Yang X and Zhang J: Bridges between mitochondrial oxidative stress, ER stress and mTOR signaling in pancreatic β cells. Cell Signal. 28:1099–1104. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Maiese K: Targeting molecules to medicine with mTOR, autophagy and neurodegenerative disorders. Br J Clin Pharmacol. 82:1245–1266. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Escribano O, Gomez-Hernandez A, Diaz-Castroverde S, Nevado C, Garcia G, Otoro YF, Perdomo L, Beneit N and Benito M: Insulin receptor isoform A confers a higher proliferative capability to pancreatic β cells enabling glucose availability and IGF-1 signaling. Mol Cell Endocrinol. 409:82–91. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Li W, Zhang H, Nie A, Ni Q, Li F, Ning G, Li X, Gu Y and Wang Q: mTORC1 pathway mediates beta cell compensatory proliferation in 60% partial-pancreatectomy mice. Endocrine. 53:117–128. 2016. View Article : Google Scholar : PubMed/NCBI

27 

McCurdy CE and Klemm DJ: Adipose tissue insulin sensitivity and macrophage recruitment: Does PI3K pick the pathway. Adipocyte. 2:135–142. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Bringhenti I, Moraes-Teixeira JA, Cunha MR, Ornellas F, Mandarim-de-lacerda CA and Aguila MB: Maternal obesity during the preconception and early life periods alters pancreatic development in early and adult life in male mouse offspring. PLoS One. 8:e557112013. View Article : Google Scholar : PubMed/NCBI

29 

Cerf ME, Williams K, Chapman CS and Louw J: Compromised beta-cell development and beta-cell dysfunction in weanling offspring from dams maintained on a high-fat diet during gestation. Pancreas. 34:347–353. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Rhee M, Lee H, Kim JW, Ham DS, Park HS, Yang HK, Shin JY, Cho JH, Kim YB, Youn BS, et al: Preadipocyte factor 1 induces pancreatic ductal cell differentiation into insulin-producing cells. Sci Rep. 6:239602016. View Article : Google Scholar : PubMed/NCBI

31 

Gao Y, Liao G, Xiang C, Yang X, Cheng X and Ou Y: Effects of phycocyanin on INS-1 pancreatic β-cell mediated by PI3K/Akt/FoxO1 signaling pathway. Int J Biol Macromol. 83:185–194. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Ahn C, An BS and Jeung EB: Streptozotocin induces endoplasmic reticulum stress and apoptosis via disruption of calcium homeostasis in mouse pancreas. Mol Cell Endocrinol. 412:302–308. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Khan S, Yan-Do R, Duong E, Wu X, Bautista A, Cheley S, MacDonald PE and Braun M: Autocrine activation of P2Y1 receptors couples Ca (2+) release in human pancreatic beta cells. Diabetologia. 57:2235–2245. 2014. View Article : Google Scholar

34 

Roper MG, Qian WJ, Zhang BB, Kulkarni RN, Kahn CR and Kennedy RT: Effect of the insulin mimetic L-783,281 on intracellular Ca2 and insulin secretion from pancreatic beta-cells. Diabetes. 51 Suppl:S43–S49. 2002. View Article : Google Scholar : PubMed/NCBI

35 

Demozay D, Tsunekawa S, Briaud I, Shah R and Rhodes CJ: Specific glucose-induced control of insulin receptor substrate-2 expression is mediated via Ca2+-dependent calcineurin/NFAT signaling in primary pancreatic islet β-cells. Diabetes. 60:2892–2902. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Goodyer WR, Gu X, Liu Y, Bottino R, Crabtree GR and Kim SK: Neonatal β cell development in mice and humans is regulated by calcineurin/NFAT. Dev Cell. 23:21–34. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Nguidjoe E, Sokolow S, Bigabwa S, Pachera N, D'Amico E, Allagnat F, Vanderwinden JM, Sener A, Manto M, Depreter M, et al: Heterozygous inactivation of the Na/Ca exchanger increases glucose-induced insulin release, β-cell proliferation, and mass. Diabetes. 60:2076–2085. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Lawrence MC, Naziruddin B, Levy MF, Jackson A and McGlynn K: Calcineurin/nuclear factor of activated T cells and MAPK signaling induce TNF-{alpha} gene expression in pancreatic islet endocrine cells. J Biol Chem. 286:1025–1036. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Gilon P, Chae HY, Rutter GA and Ravier MA: Calcium signaling in pancreatic β-cells in health and in type 2 diabetes. Cell Calcium. 5:340–361. 2014. View Article : Google Scholar

40 

Kappel VD, Frederico MJ, Postal BG, Mendes CP, Cazarolli LH and Silva FR: The role of calcium in intracellular pathways of rutin in rat pancreatic islets: Potential insulin secretagogue effect. Eur J Pharmacol. 702:264–268. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Castro AJ, Cazarolli LH, de-Carvalho FK, da Luz G, Altenhofen D, dos Santos AR, Pizzolatti MG and Silva FR: Acute effect of 3β-hidroxihop-22(29)ene on insulin secretion is mediated by GLP-1, potassium and calcium channels for the glucose homeostasis. J Sterois Biochem Mol Biol. 150:112–122. 2015. View Article : Google Scholar

42 

Marcelo KL, Ribar T, Means CR, Tsimelzon A, Stevens RD, Llkayeva O, Bain JR, Hilsenbeck SG, Newgard CB, Means AR and York B: Research resource: Roles for calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2) in systems metabolism. Mol Endocrinol. 30:557–572. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Markwardt ML, Seckinger KM and Rizzo MA: Regulation of glucokinase by intracellular calcium levels in pancreatic β cells. J Biol Chem. 291:3000–3009. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Nussinov R, Tsai CJ, Muratcioglu S, Jang H, Gursoy A and Keskin O: Principles of K-Ras effector organization and the role of oncogenic K-Ras in cancer initiation through G1 cell cycle. Expert Rev Proteomics. 12:669–682. 2015. View Article : Google Scholar : PubMed/NCBI

45 

Chamberlain CE, Scheel DW, Mcglynn K, Kim H, Miyatsuka T, Wang J, Nguyen V, Zhao S, Mavropoulos A, Abraham AG, et al: Menin determines K-RAS proliferative outputs in endocrine cells. J Clin Invest. 124:4093–4101. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Kim TK, Lee JS, Jung HS, Ha TK, Kim SM, Han N, Lee EJ, Kim TN, Kwon MJ, Lee SH, et al: Triiodothyronine induces proliferation of pancreatic β-cells through the MAPK/ERK pathway. Exp Clin Endocrinol Diabetes. 122:240–245. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Chen H, Gu X, Liu Y, Wang J, Wirt SE, Bottino R, Schorle H, Sage J and Kim SK: PDGF signaling controls age-dependent proliferation in pancreatic β-cells. Nature. 478:349–355. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Hoarau E, Chandra V, Rustin P, Scharfmann R and Duvillie B: Pro-oxidant/antioxidant balance controls pancreatic β-cell differentiation through the ERK1/2 pathway. Cell Death Dis. 5:e14872014. View Article : Google Scholar : PubMed/NCBI

49 

Xiang RL, Mei M, Su YC, Li L, Wang JY and Wu LL: Visfatin protects rat pancreatic β-cells against IFN-γ-induced apoptosis through AMPK and ERK1/2 signaling pathways. Biomed Environ Sci. 28:169–177. 2015.PubMed/NCBI

50 

Ozaki KI, Awazu M, Tamiya M, Lwasaki Y, Harada A, Kugisaki S, Tanimura S and Kohno M: Targeting the ERK signaling pathway as a potential treatment for insulin resistance and type 2 diabetes. Am J Physiol Endocrino Metab. 310:E643–E651. 2016. View Article : Google Scholar

51 

Wang H, Gambosova K, Cooper ZA, Holloway MP, Kassai A, Lzquierdo D, Cleveland K, Boney CM and Altura RA: EGF regulates surviving stability through the Raf-1/ERK pathway in insulin-secreting pancreatic β-cells. BMC Mol Biol. 11:662010. View Article : Google Scholar : PubMed/NCBI

52 

Ernesto BM, Kulkarni RN, Scott DK, Mauvais-Jarvis F, Stewart AF and Garcia-Ocana A: Human β-cell proliferation and intracellular signaling part 2: Still driving in the dark without a road map. Diabetes. 63:819–831. 2014. View Article : Google Scholar : PubMed/NCBI

53 

Oh YS, Shin S, Lee YJ, Kim EH and Jun HS: Betacellulin-induced beta cell proliferation and regeneration is mediated by activation of ErbB-1 and ErbB-2 receptors. PLoS One. 6:e238942011. View Article : Google Scholar : PubMed/NCBI

54 

Hakonen E, Ustinov J, Eizirik DL, Sariola H, Miettinen PJ and Otonkoski T: In vivo activation of the PI3K-Akt pathway in mouse beta cells by the EGFR mutation L858R protects against diabetes. Diabetologia. 57:970–979. 2014. View Article : Google Scholar : PubMed/NCBI

55 

Zarrouki B, Benterki I, Fontes G, Peyot ML, Seda O, Prentki M and Poitout V: Epidermal growth factor receptor signaling promotes pancreatic β-cell proliferation in response to nutrient excess in rats through mTOR and FOXM1. Diabetes. 63:982–993. 2014. View Article : Google Scholar : PubMed/NCBI

56 

Zhang CC, Yin X, Cao CY, Wei J, Zhang Q and Gao JM: Chemical constituents from hericium erinaceus and their ability to stimulate NGF-mediated neurite outgrowth on PC12 cells. Bioorg Med Chem Lett. 25:5078–5082. 2015. View Article : Google Scholar : PubMed/NCBI

57 

Rosenbaum T, Vidaltamayo R, Sanchez-Soto MC, Zentella A and Hiriart M: Pancreatic beta cells synthesize and secrete nerve growth factor. Proc Natl Acad Sci USA. 95:7784–7788. 1998. View Article : Google Scholar : PubMed/NCBI

58 

Freund V and Frossard N: Expression of nerve growth factor in the airways and its possible role in asthma. Prog Brain Res. 146:335–346. 2004. View Article : Google Scholar : PubMed/NCBI

59 

Roux PP and Barker PA: Neurotrophin signaling through the P75 neurotrophin receptor. Prog Neurobiol. 67:203–233. 2002. View Article : Google Scholar : PubMed/NCBI

60 

Pingitore A, Caroleo MC, Cione E, Castanera-Gonzalez R, Huang GC and Perasud SJ: Fine tuning of insulin secretion by release of nerve growth factor from mouse and human islet β-cells. Mol Cell Endocrinol. 436:23–32. 2016. View Article : Google Scholar : PubMed/NCBI

61 

EI-Gohary Y, Tulachan S, Guo P, Welsh C, Wiersch J, Prasadan K, Paredes J, Shiota C, Xiao X, Wada Y, et al: Smad signaling pathways regulate pancreatic endocrine development. Dev Biol. 378:83–93. 2013. View Article : Google Scholar : PubMed/NCBI

62 

Feng Z, Zi Z and Liu X: Measuring TGF-β ligand dynamics in culture medium. Methods Mol Biol. 1344:379–389. 2016. View Article : Google Scholar : PubMed/NCBI

63 

Richardson CC, To K, Foot VL, Hauge-Evans AC, Carmignac D and Christie MR: Increased perinated remodeling of the pancreas in somatostatin-deficient mice: Potential role of transforming growth factor-beta signaling in regulating beta cell growth in early life. Horm Metab Res. 47:56–63. 2015.PubMed/NCBI

64 

Xiao X, Wiersch J, EI-Gohary Y, Guo P, Prasadan K, Paredes J, Welsh C, Shiota C and Gittes GK: TGFβ receptor signaling is essential for inflammation-induced but not β-cell workload-induced β-cell proliferation. Diabetes. 62:1217–1226. 2013. View Article : Google Scholar : PubMed/NCBI

65 

Blum B, Roose AN, Barrrandon O, Maehr R, Arvanites AC, Davidow LS, Davis JC, Peterson QP, Rubin LL and Melton DA: Reversal of β cell de-differentiation by a small molecule inhibitor of the TGFβ pathway. Elife. 3:e028092014. View Article : Google Scholar : PubMed/NCBI

66 

Bruun C, Christensen GL, Jacobsen ML, Kanstrup MB, Jensen PR, Fjordvang H, Mandrup-Poulsen T and Billestrup N: Inhibition of beta cell growth and function by bone morphogenetic proteins. Diabetologia. 57:2546–2554. 2014. View Article : Google Scholar : PubMed/NCBI

67 

EI-Gohary Y, Tulachan S, Wiersch J, Guo P, Welsh C, Prasadan K, Paredes J, Shiota C, Xiao X, Wada Y, et al: A smad signaling network regulates islet cell proliferation. Diabetes. 63:224–236. 2014. View Article : Google Scholar : PubMed/NCBI

68 

Lei C, Zhou X, Pang Y, Mao Y, Lu X, Li M and Zhang J: TGF-β signaling prevents pancreatic beta cell death after proliferation. Cell Prolif. 48:356–362. 2015. View Article : Google Scholar : PubMed/NCBI

69 

Wu H, Mezghenna K, Marmol P, Guo T, Moliner A, Yang SN, Berggren PO and Lbanez CF: Differential regulation of mouse pancreatic islet insulin secretion and Smad proteins by activin ligands. Diabetologia. 57:148–156. 2014. View Article : Google Scholar : PubMed/NCBI

70 

Xiao X, Gaffar I, Guo P, Wiersch J, Fischbach S, Peirish L, Song Z, El-Gohary Y, Prasadan K, Shiota G and Gittes GK: M2 macrophages promote beta-cell proliferation by up-regulation of SMAD7. Prov Natl Acad Sci USA. 111:E1211–E1220. 2014. View Article : Google Scholar

71 

Shin JA, Hong OK, Lee HJ, Jeon SY, Kim JW, Lee SH, Cho JH, Lee JM, Choi YH, Chang SA, et al: Transforming growth factor-β induces epithelial to mesenchymal transition and suppresses the proliferation and transdifferentiation of cultured human pancreatic duct cells. J Cell Biochem. 112:179–188. 2011. View Article : Google Scholar : PubMed/NCBI

72 

Toren-Haritan G and Efrat S: TGFβ pathway inhibition redifferentiates human pancreatic islet β cells expanded in vitro. PLoS One. 9:e01391682015. View Article : Google Scholar

73 

Li J, Ying H, Cai G, Guo Q and Chen L: Pre-Eclampsia-associated reduction in placental growth factor impaired beta cell proliferation through PI3K signaling. Cell Physiol Biochem. 36:34–43. 2015. View Article : Google Scholar : PubMed/NCBI

74 

Huang Y and Chang Y: Regulation of pancreatic islet beta-cell mass by growth factor and hormone signaling. Prog Mol Biol Transl Sci. 121:321–349. 2014. View Article : Google Scholar : PubMed/NCBI

75 

Lee YS and Jun HS: Anti-diabetic actions of glucagon-like peptide-1 on pancreatic beta-cells. Metabolism. 63:9–19. 2014. View Article : Google Scholar : PubMed/NCBI

76 

Tian L and Jin T: The incretin hormone GLP-1 and mechanisms underlying its secretion. J Diabetes. 8:753–765. 2016. View Article : Google Scholar : PubMed/NCBI

77 

Dai FF, Bhattacharjee A, Liu Y, Batchuluun B, Zhang M, Wang XS, Huang X, Luu L, Zhu D, Gaisano H and Wheeler MB: A novel GLP1 receptor interacting protein ATP6ap2 regulates insulin secretion in pancreatic beta cells. J Biol Chem. 290:25045–25061. 2015. View Article : Google Scholar : PubMed/NCBI

78 

Zhang M, Robitaille M, Showalter AD, Huang X, Liu Y, Bhattacharjee A, Willard FS, Han J, Froese S, Wei L, et al: Progesterone receptor membrane component 1 is a functional part of the glucagon-like peptide-1 (GLP-1) receptor complex in pancreatic β cells. Mol Cell Proteomics. 13:3049–3062. 2014. View Article : Google Scholar : PubMed/NCBI

79 

Campbell JE and Drucker DJ: Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab. 17:819–837. 2013. View Article : Google Scholar : PubMed/NCBI

80 

Lavine JA and Attie AD: Gastrointerstinal hormones and the regulation of β-cell mass. Ann NY Acad Sci. 1212:41–58. 2010. View Article : Google Scholar : PubMed/NCBI

81 

Kong X, Yan D, Wu X, Guan Y and Ma X: Glucotoxicity inhibits cAMP-protein kinase A-potentiated glucose-stimulated insulin secretion in pancreatic β-cells. J Diabetes. 7:378–385. 2015. View Article : Google Scholar : PubMed/NCBI

82 

Zhang Y, Ding Y, Zhong X, Guo Q, Wang H, Gao J, Bai T, Ren L, Guo Y, Jiao X and Liu Y: Geniposide acutely stimulates insulin secretion in pancreation β-cells by regulating GLP-1 receptor/cAMP signaling and ion channels. Mol Cell Endocrinol. 430:89–96. 2016. View Article : Google Scholar : PubMed/NCBI

83 

Yang Y, Tong Y, Gong M, Lu Y, Wang C, Zhou M, Yang Q, Mao T and Tong N: Activation of PPARβ/δ protects pancreatic β cells from palmitate-induced apoptosis by upregulating the expression of GLP-1 receptor. Cell Signal. 26:268–278. 2014. View Article : Google Scholar : PubMed/NCBI

84 

Boutant M, Ramos OH, Tourrel-Cuzin C, Movassat J, Llias A, Vallois D, Planchais J, Pegorier JP, Schuit F, Petit PX, et al: COUP-TFII controls mouse pancreatic β-cell mass through GLP-1-β-catenin signaling pathways. PLoS One. 7:e308472012. View Article : Google Scholar : PubMed/NCBI

85 

Nelson WJ and Nusse R: Convergence of Wnt, beta-catenin, and cadherin pathways. Science. 303:1483–1487. 2004. View Article : Google Scholar : PubMed/NCBI

86 

Nusse R: Wnt signaling in disease and in development. Cell Res. 15:28–32. 2005. View Article : Google Scholar : PubMed/NCBI

87 

Heller C, Kuhn MC, Mulders-Opgenoorth B, Schott M, Willenberg HS, Scherbaum WA and Schinner S: Exendin-4 upregulates the expression of wnt-4, a novel regulator of pancreatic β-cell proliferation. Am J Physiol Endocrinol Metab. 301:E864–E872. 2011. View Article : Google Scholar : PubMed/NCBI

88 

Xue G, Romano E, Massi D and MandaIa M: Wnt/β-catenin signaling in melanoma: Preclinical rationale and novel therapeutic insights. Cancer Treat Rev. 49:1–12. 2016. View Article : Google Scholar : PubMed/NCBI

89 

Schinner S, Ulgen F, Papewalis C, Schott M, Woelk A, Vidal-Puig A and Scherbaum WA: Regulation of insulin secretion, glucokinase gene transcription and beta cell proliferation by adipicyte-derived wnt signaling molecules. Diabetologia. 51:147–154. 2008. View Article : Google Scholar : PubMed/NCBI

90 

Bader E, Migliorini A, Gegg M, Moruzzi N, Gerdes J, Roscioni SS, Bakhti M, Brandl E, Irmler M, Beckers J, et al: Indentification of proliferative and mature β-cells in the islets of Langerhans. Nature. 535:430–434. 2016. View Article : Google Scholar : PubMed/NCBI

91 

Krutzfeldt J and Stoffel M: Regulation of wingless-type MMTV integration site family (WNT) signaling in pancreatic islets from wild-type and obese mice. Diabetologia. 53:123–127. 2010. View Article : Google Scholar : PubMed/NCBI

92 

Gui S, Yuan G, Wang L, Zhou L, Xue Y, Yu Y, Zhang J, Zhang M, Yang Y and Wang DW: Wnt3a regulates proliferation, apoptosis and function of pancreatic NIT-1 β cells via activation of IRS2/PI3K signaling. J Cell Biochem. 114:1488–1497. 2013. View Article : Google Scholar : PubMed/NCBI

93 

Rulifson IC, Karnik SK, Heiser PW, Ten Berge D, Chen H, Gu X, Taketo MM, Nusse R, Hebro M and Kim SK: Wnt signaling regulates pancreatic beta cell proliferation. Proc Nati Acad Sci USA. 104:6247–6252. 2007. View Article : Google Scholar

94 

He X, Han W, Hu SX, Zhang MZ, Hua JL and Peng S: Canonical wnt signaling pathway contributes to the proliferation and survival in porcine pancreatic stem cells (PSCs). Cell Tissue Res. 362:379–388. 2015. View Article : Google Scholar : PubMed/NCBI

95 

Sarkar S and Mandal C, Sangwan R and Mandal C: Coupling G2/M arrest to the wnt/β-catenin pathway restrains pancreatic adcnocarcinoma. Endocr Relat Cancer. 21:113–125. 2014. View Article : Google Scholar : PubMed/NCBI

96 

Zhang YQ, Morris JP, Yan W, Schofield HK, Gurney A, Simeone DM, Millar SE, Hoey T, Hebrok M and Pasca di Magliano M: Canonical wnt signaling is required for pancreatic carcinogenesis. Cancer Res. 73:4909–4922. 2013. View Article : Google Scholar : PubMed/NCBI

97 

Afelik S, Pool B, Schmerr M, Penton C and Jensen J: Wnt7b is required for epithelial progenitor growth and operates during epithelial-to-mesenchymal signaling in pancreatic development. Dev Biol. 399:204–217. 2015. View Article : Google Scholar : PubMed/NCBI

98 

Lyssenko V: The transcription factor 7-like 2 gene and increased risk of type 2 diabetes: An update. Curr Opin Clin Nutr Metab Care. 11:385–392. 2008. View Article : Google Scholar : PubMed/NCBI

99 

Takamoto I, Kubota N, Nakaya K, Kumagai K, Hashimoto S, Kubota T, Inoue M, Kajiwara E, Katsuyama H, Obata A, et al: TCF7L2 in mouse pancreatic beta cells plays a crucial role in glucose homeostasis by regulating beta cell mass. Diabetologia. 57:542–553. 2014. View Article : Google Scholar : PubMed/NCBI

100 

Zhou Y, Oskolkov N, Shcherbina L, Ratti J, Kock KH, Su J, Martin B, Oskolkova MZ, Goransson O, Bacon J, et al: HMGB1 binds to the rs7903146 locus in TCF7L2 in human pancreatic islets. Mol Cell Endocrinol. 430:138–145. 2016. View Article : Google Scholar : PubMed/NCBI

101 

Yao DD, Yang L, Wang Y, Liu C, Wei YJ, Jia XB, Yin W and Shu L: Geniposide promotes beta-cell regeneration and survival through regulating β-catenin/TCF7L2 pathway. Cell Death Dis. 6:e17462015. View Article : Google Scholar : PubMed/NCBI

102 

Kiu H and Nicholson SE: Biology and significance of the JAK/STAT signaling pathways. Growth Factors. 30:88–106. 2012. View Article : Google Scholar : PubMed/NCBI

103 

O'Shea JJ and Plenge R: JAK and STAT signaling molecules in immunoregulation and immune-mediated disease. Immunity. 36:542–550. 2012. View Article : Google Scholar : PubMed/NCBI

104 

Zhuang S: Regulation of STAT signaling by acetylation. Cell Signal. 25:1942–1931. 2013. View Article : Google Scholar

105 

Brooks AJ, Dai W, O'Mara ML, Abankwa D, Chhabra Y, Pelekanos RA, Gardon O, Tunny KA, Blucher KM, Morton CJ, et al: Mechanism of activation of protein kinase JAK2 by the growth hormone recetor. Science. 344:12497832014. View Article : Google Scholar : PubMed/NCBI

106 

Liongue C, Taznin T and Ward AC: Signaling via the CytoR/JAK/STAT/SOCS pathway: Emergence during evolution. Mol Immunol. 71:166–175. 2016. View Article : Google Scholar : PubMed/NCBI

107 

Linossi EM, Babon JJ, Hilton DJ and Nicholson SE: Suppression of cytokine signaling: The SOCS perspective. Cytokine Growth Factor Rev. 24:241–248. 2013. View Article : Google Scholar : PubMed/NCBI

108 

Shuai K and Liu B: Regulation of gene-activation pathways by PIAS protein in the immune system. Nat Rev Immunol. 5:593–605. 2005. View Article : Google Scholar : PubMed/NCBI

109 

Trengove MC and Ward AC: SOCS proteins in development and disease. Am J Exp Clin Immunol. 2:1–29. 2013.

110 

Fleyel T, Brorsson C, Nielsen LB, Miani M, Bang-Berthelsen CH, Friedrichsen M, Overgaard AJ, Berchtold LA, Wiberg A, Poulsen P, et al: CTSH regulates β-cell function and disease progression in newly diagnosed type 1 diabetes patients. Proc Natl Acad Sci USA. 111:10305–10310. 2014. View Article : Google Scholar : PubMed/NCBI

111 

Stanley WJ, Litwak SA, Quah HS, Tan SM, Kay TW, Tiganis T, de Haan JB, Thomas HE and Gurzov EN: Inactivation of protein tyrosine phosphatases enhances interferon signaling in pancreatic islets. Diabetes. 64:2489–2496. 2015. View Article : Google Scholar : PubMed/NCBI

112 

Chou DH, Vetere A, Choudhary A, Scully SS, Schenone M, Tang A, Gomez R, Burns SM, Lundh M, Vital T, et al: Kinase-independent small-molecule inhibition of JAK-STAT signaling. J Am Chem Soc. 137:7929–7934. 2015. View Article : Google Scholar : PubMed/NCBI

113 

Chen H, Kleinberger JW, Takane KK, Salim F, Fiaschi-Taesch N, Pappas K, Parsons R, Jiang J, Zhang Y, Liu H, et al: Augmented stat5 signaling bypasses multiple impediments to lactogen-mediated proliferation in human β-cells. Diabetes. 64:3784–3797. 2015. View Article : Google Scholar : PubMed/NCBI

114 

Choi D, Schroer SA, Lu SY, Wang L, Wu X, Liu Y, Zhang Y, Gaisano HY, Wagner KY, Wu H, et al: Erythropoietin protects against diabetes through direct effects on pancreatic beta cells. J Exp Med. 207:2831–2842. 2010. View Article : Google Scholar : PubMed/NCBI

115 

De-Groef S, Renmans D, Cai Y, Leuckx G, Roels S, Staels W, GradwohI G, Baeyens L, Heremans Y, Martens GA, et al: STAT3 modulates β-cell cycling in injured mouse pancreas and protects against DNA damage. Cell Death Dis. 7:e22722016. View Article : Google Scholar : PubMed/NCBI

116 

Shao L, Zhang P, Zhang Y and Ma A: Inflammatory unbalance of TLR3 and TLR4 in PCI patients with or without type 2 diabetes mellitus. Immunol Lett. 161:81–88. 2014. View Article : Google Scholar : PubMed/NCBI

117 

Kruger B, Yin N, Zhang N, Yadav A, Coward W, Lai G, Zang W, S Heeger P, Bromberg JS, Murphy B, et al: Islet-expressed TLR2 and TLR4 sense injury and mediate early graft failure after transplantation. Eur J Immunol. 40:2914–2924. 2010. View Article : Google Scholar : PubMed/NCBI

118 

Yang X, Haghiac M, Glazebrook P, Minium J, Catalano PM and Hanguel-de Mouzon S: Saturated fatty acids enhance TLR4 immune pathways in human trophoblasts. Hum Reprod. 30:2152–2159. 2015. View Article : Google Scholar : PubMed/NCBI

119 

Sepehri Z, Kiani Z, Nasiri AA, Mashhadi MA, Javadian F, Haghighi A, Kohan F, Bahari A and Sargazi A: Human Toll like receptor 4 gene expression of PBMCs in diabetes mellitus type 2 patients. Cell Mol Biol. 61:92–95. 2015.PubMed/NCBI

120 

Verzila D, Cappuccino L, D'Amato E, Villaggio B, Gianiorio F, Mij M, Simonato A, Viazzi F, Salvidia G and Garibotto G: Enhanced glomerular Toll-like receptor 4 expression and signaling in patients with type 2 diabetic nephropathy and microalbuminuria. Kidney Int. 86:1229–1243. 2014. View Article : Google Scholar : PubMed/NCBI

121 

Baldan A, Ferronato S, Olivato S, Malerba G, Scuro A, Veraldi GF, Gelati M, Ferrari S, Mariotto S, Pignati PF, et al: Cyclooxygenase 2, toll-like receptor 4 and interleukin 1β mRNA expression in atherosclerotic plaques of type 2 diabetic patients. Inflamm Res. 63:851–858. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2018
Volume 16 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jiang WJ, Peng YC and Yang KM: Cellular signaling pathways regulating β‑cell proliferation as a promising therapeutic target in the treatment of diabetes (Review). Exp Ther Med 16: 3275-3285, 2018
APA
Jiang, W., Peng, Y., & Yang, K. (2018). Cellular signaling pathways regulating β‑cell proliferation as a promising therapeutic target in the treatment of diabetes (Review). Experimental and Therapeutic Medicine, 16, 3275-3285. https://doi.org/10.3892/etm.2018.6603
MLA
Jiang, W., Peng, Y., Yang, K."Cellular signaling pathways regulating β‑cell proliferation as a promising therapeutic target in the treatment of diabetes (Review)". Experimental and Therapeutic Medicine 16.4 (2018): 3275-3285.
Chicago
Jiang, W., Peng, Y., Yang, K."Cellular signaling pathways regulating β‑cell proliferation as a promising therapeutic target in the treatment of diabetes (Review)". Experimental and Therapeutic Medicine 16, no. 4 (2018): 3275-3285. https://doi.org/10.3892/etm.2018.6603