Next Article in Journal
Pharmacotherapy Treatment Options for Insomnia: A Primer for Clinicians
Next Article in Special Issue
Identification of Serum Peptidome Signatures of Non-Small Cell Lung Cancer
Previous Article in Journal
Innovative Target Therapies Are Able to Block the Inflammation Associated with Dysfunction of the Cholesterol Biosynthesis Pathway
Previous Article in Special Issue
Alisertib Induces Cell Cycle Arrest, Apoptosis, Autophagy and Suppresses EMT in HT29 and Caco-2 Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Notch Signaling in Pancreatic Development

1
College of Life Science, Northeast Forestry University, Harbin 150040, China
2
College of Life Sciences, Agriculture and Forestry, Qiqihar University, Qiqihar 161006, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2016, 17(1), 48; https://doi.org/10.3390/ijms17010048
Submission received: 22 November 2015 / Revised: 22 December 2015 / Accepted: 24 December 2015 / Published: 30 December 2015
(This article belongs to the Special Issue Molecular Classification of Human Cancer: Diagnosis and Treatment)

Abstract

:
The Notch signaling pathway plays a significant role in embryonic cell fate determination and adult tissue homeostasis. Various studies have demonstrated the deep involvement of Notch signaling in the development of the pancreas and the lateral inhibition of Notch signaling in pancreatic progenitor differentiation and maintenance. The targeted inactivation of the Notch pathway components promotes premature differentiation of the endocrine pancreas. However, there is still the contrary opinion that Notch signaling specifies the endocrine lineage. Here, we review the current knowledge of the Notch signaling pathway in pancreatic development and its crosstalk with the Wingless and INT-1 (Wnt) and fibroblast growth factor (FGF) pathways.

Graphical Abstract

1. Introduction

Notch signaling is an evolutionarily conserved pathway for cell-cell communication and cell-fate determination during embryonic development and tissue homeostasis [1,2]; it includes canonical and non-canonical pathways. The former is initiated by ligand-receptor interactions between adjacent cells [3,4], which results in the activation of the Hes1 gene by a complex consisting of the Notch intracellular domain (NICD), Rbp-Jκ family of nuclear proteins (Rbp-J (mammalian), Su(H) (Drosophila), and Lag-1 (Caenorhabditis elegans)), and the Mastermind co-activator. However, non-canonical Notch signaling broadly encompasses several modes of Notch activity that do not go through the Rbp-J and activation of the Hes/Hey genes [5,6]. Here, we review the role of the canonical pathway during pancreatic development. Notch-mediated lateral inhibition represents an important conserved mechanism that regulates cell differentiation, cell proliferation and cell survival in stem cells [7,8,9]. Abnormalities in Notch signaling have been linked to various syndromes and diseases, including developmental malformation, neurodegenerative diseases, metabolic disorders, and malignant disease [10,11,12,13].
Over the past two decades, numerous reports have revealed the pivotal role of Notch signaling in pancreatic specification, cell proliferation, differentiation and plasticity [9,14,15,16]. The first evidence of the involvement of the Notch signaling pathway in pancreatic development focused on its lateral inhibition role in controlling pancreatic fate decision. The activation of Notch signaling in pancreatic progenitors prevents their differentiation into the endocrine or exocrine cell lineage [17,18]. In contrast, the blockage of the Notch signaling pathway causes premature differentiation of the multipotent progenitor cells (MPCs) into endocrine cells [19,20]. A series of studies have revealed that Notch signaling functions as a negative regulator of the pro-endocrine factor neurogenin3 (Ngn3), and the formation of insulin-producing β-cells is significantly enhanced by induction of pro-endocrine factors or inhibition of Notch processing. However, recently, some researchers have disagreed with the notion that the Notch pathway is an inhibitor of endocrine cell differentiation. They have proposed that the Notch pathway specifies the pancreatic progenitors differentiating towards endocrine lineage [21,22] or the inactivated Notch pathway promotes acinar cell differentiation [23,24,25]. Some recent findings have revealed that Notch signaling does not act through an on or off mode, but at a Notch level-dependent manner to regulate the quiescence, self-renewal and differentiation of pancreatic progenitor cells during pancreas development [22,26]. Studies on animal models of pancreatic regeneration and diseases have revealed that Notch signaling is involved in controlling the plasticity of terminally differentiated adult pancreatic cells [25,27,28]. During pancreatic development, the formation of islets of Langerhans is enhanced by specific transcription factors and regulated by multiple intercellular signaling pathways, including the Wingless and INT-1 (Wnt), fibroblast growth factor (FGF), Notch pathways, etc. [29,30,31]. These pathways independently or collaboratively perform regulatory functions at different time-points. This mini review summarizes the current knowledge of the roles of Notch signaling in pancreatic development, including pancreatic cell lineage commitment, pancreatic progenitor maintenance, and adult pancreatic cell plasticity, and it also discusses the crosstalk between the Notch and Wnt/FGF pathways.

2. An Outline of Pancreatic Development

The mammalian pancreas is derived from two independent ventral and dorsal buds and experiences three stages of transition [32,33]. In mice, the primary transition is marked by the specification and proliferation of pancreatic progenitors and the appearance of glucagon-producing cells during E9.5 and E12.5 [34,35]. The secondary transition is from E13.5 to E15.5, during which all five hormone-expressing endocrine lineages (α-, β-, δ-, ε-, and PP-cells) begin to emerge rapidly and amylase-expressing acinar cells arise from the extending tip epithelium [36,37]. The third transition occurs from E16.5 to E19. During this period, endocrine cells migrate and cluster into numerous islets, and acinar cells further expand [38]. The pancreatic buds contain undifferentiated progenitor cells, which contribute to all pancreatic cell lineages, the exocrine, ductal and endocrine cell lineages [39,40]. In the MPCs, Notch signaling is critical and essential for their proliferation and commitment [22,24] (Figure 1).
Figure 1. A schematic of pancreatic development.
Figure 1. A schematic of pancreatic development.
Ijms 17 00048 g001
Lineage-specific transcription factors control the differentiation of pancreatic progenitor cells towards a specific type [40,41]. Pancreatic cells arise from pancreatic duodenal homeobox 1 (Pdx1)-expressing progenitors (Figure 1). As pancreatic development proceeds, the Pdx1 gene becomes progressively confined to endocrine β-cells, where it plays an essential role in the transcriptional activation of the insulin gene [42]. The progenitors co-express sex-determining region Y (Sry) box 9 (Sox9) and pancreas specific transcription factor1a (Ptf1a, also known as P48) [43,44] (Figure 1). However, Sox9 expression is eventually limited to a subset of ductal and centroacinar cells (CACs) in adults, and Ptf1a is expressed solely in mature acinar cells [45,46] (Figure 1). The Basic Helix-Loop-Helix (bHLH) transcription factor Ngn3 drives MPCs towards the endocrine cell fate [47]. The specification of the endocrine subtypes is essentially under the control of the opposing actions of aristaless related homeobox (Arx) and paired box 4 (Pax4) acting downstream of Ngn3 [48] (Figure 1). Ngn3-null mice fail to generate pancreatic endocrine cells and lose the expression of islet transcription factors, including islet-1 (Isl1), NK2 homeobox 2 (Nkx2.2), paired box 4 (Pax4), paired box 6 (Pax6) and NeuroD1, which are all important for endocrine cell differentiation [49]. During the maturation of islet cells, the cell cycle regulatory proteins play a pivotal role in cell division and differentiation. Prior to and during the secondary transition, cyclin-dependent kinase 4 (Cdk4) and E2F transcription factor 1 (E2F1) promote β-cell development by activating Ngn3 to increase the numbers of endocrine precursors [50]. In the mouse embryonic pancreas, P21 protein (Cdc42/Rac)-activated kinase 3 (Pak3) acts downstream of Ngn3 to promote cell cycle exit and cell differentiation by repressing cyclin D1 in Ngn3+ endocrine progenitors [51]. The PTF1 complex initiates exocrine differentiation [52]. The PTF1 complex, which is composed of p64/HEB, p75/E2A and Ptf1a [52], directly binds to the promoter regions of the acinar digestive enzyme genes, and leads to acinar cell differentiation and cell cycle exit [46,53]. More interestingly, a recent study on the Ptf1a mutant zebrafish model has found that the down-regulation of Ptf1a induces acinar-to-endocrine fate conversion [54]. Using genetic loss- and gain-of-function approaches, Schaffer et al. [55], have demonstrated that the cross-repressive interactions between Nkx6 (Nkx6.1/Nkx6.2) and Ptf1a commit the fate of pancreatic progenitor cells. Nkx6 induces endocrine cell determination, however, Ptf1a promotes acinar cell specification.The cross-antagonistic switch between Nkx6 and Ptf1a is controlled by Notch signaling [55].
Growing evidence suggests that miRNAs play an important role in the embryonic development and physiological function of pancreas. Examples of these miRNAs include miR-7, miR-375, the miR17-92 cluster, miR-26 and miR-15 et al. [56]. Dicer-null mice display gross defects in all pancreatic lineages, especially the insulin-producing-cells. The endocrine defect in Dicer-null mice is associated with an increase in the expression of the Notch signaling target Hes1 [56]. miR-7 is dispensable for the maintenance of β-cell mass and functions [57]. miR-375 is required for normal pancreatic genesis and maintains α- and β-cell mass [58]. We have found that, during pancreatic progenitor cell differentiation, miR-375 inhibited pancreatic progenitor cell proliferation by targeting the Hippo signaling effector Yap1 [59]. miR-375 also plays a role in regulating insulin secretion through targeting the myotrophin (Mtpn) and pyruvate dehydrogenase kinase (Pdk1) genes. miR-19b and miR-18a have been shown to directly act on NeuroD1 and Ptf1a 3′ UTR, respectively [60,61]. These two miRNAs may contribute to the regulation of the differentiation and function of β-cells and acinar cells during pancreatic development. Pancreatic regeneration is accompanied by the high expression of miR-15a, miR-15b, miR-16 and miR-195, which can potentially bind to the Ngn3 transcript [62].
The development of the pancreas depends on the spatio-temporal expression of a number of transcription factors and associated signaling pathways [63,64]. Studies on the transcriptional regulation of the pancreas have been reviewed extensively, and thus, not discussed in detail here.

3. An Overview of the Notch Signaling Cascade

Notch genes encode a single-pass transmembrane receptor family of molecules, including Notch1–4 in mammals [65]. Notch proteins contain a large Notch extracellular domain (NECD) composed of 29–36 tandem epidermal growth factor-like repeats, a short Notch transmembrane fragment (NTM), and a Notch intracellular domain (NICD) [66]. As part of the biosynthetic process of Notch, a Furin-like protease in the Golgi cuts nascent Notch proteins at the S1 cleavage site into two fragments [67,68], the NECD and non-NECD domains. Then, they conjugate non-covalently as a heterodimer and target to the cellular surface. The initiation of Notch signaling is trigged by the binding of the NECD domain to type I transmembrane ligands (δ-like 1, 3, 4 (Dll1, 3, 4) and Serrate/Jagged1, 2 (Ser/Jag1, 2)) on adjacent cells. Then, the membrane-bound proteases are activated and continue to cleave the non-NECD domain into two domains, the NTM and NICD domains. This cleavage is catalyzed by A Disintegrin and Metalloprotease (ADAM)-family of metalloproteases at a luminal juxtamembrane site 2 (S2) and by a tetrameric γ-secretase complex at an intramembrane site 3 (S3) of non-NECD domain. After the transcriptionally activated NICD is released and translocates into the nucleus, it binds to Rbp-J [69]. Then, Rbp-J recruits the coactivator Mastermind-like (Maml) to activate downstream genes, including the Hairy enhance of split (Hes) and Hairy/enhancer of spit related with YRPW motif (Hey) families, nuclear factor-κB (NF-κB), vascular growth factor receptor (VEGF), etc. [70,71] (Figure 2). The Hes genes, which encode bHLH transcriptional repressors, play crucial roles in the fate choice and differentiation of stem cells, e.g., in the inner ear, neurons and pancreas development [72,73,74]. Activated Hes1 inhibits the expression of Ngn3 by binding to the proximal promoter and specifically blocking promoter activity [75,76]. However, when Notch signaling is limited or nonexistent, Rbp-J recruits a corepressor (CoR) complex to repress the expression of the Notch target genes (Figure 2).
Figure 2. A schematic of Notch signaling during pancreatic development. Notch signaling is initiated by the binding of the ligands and receptors of two neighboring cells. Upon activation, Notch is cleaved, releasing the Notch intracellular domain (NICD). The NICD can subsequently translocate into the nucleus to transcriptionally activate Notch target genes. Hes1 inhibits the expression of Ngn3 by blocking its promoter activity.
Figure 2. A schematic of Notch signaling during pancreatic development. Notch signaling is initiated by the binding of the ligands and receptors of two neighboring cells. Upon activation, Notch is cleaved, releasing the Notch intracellular domain (NICD). The NICD can subsequently translocate into the nucleus to transcriptionally activate Notch target genes. Hes1 inhibits the expression of Ngn3 by blocking its promoter activity.
Ijms 17 00048 g002
Several Notch-related regulators have also been found to function in normal organism development and tumorigenesis. In the left-right asymmetric development of Xenopus, the transcriptional repressor B cell leukemia/lymphoma 6 (Bcl6) competes with Maml to repress the Notch1 target genes [77]. Mind bomb1 (Mib1), which encodes an E3 ubiquitin ligase, promotes the internalization of Δ, which paradoxically increases the efficiency with which Δ activates Notch [78]. It is reported that Mib1 is required for the formation of pancreatic β-cells and the specification of neurons and glia in the spinal cord [79,80]. In neural progenitors, the multifunctional protein kinase Nemo-like kinase (NLK) negatively regulates the formation of the Rbp-J-NICD-Maml complex through the phosphorylation of Notch1 to fine-tune the timing of neuronal differentiation [81]. DEAD (Asp-Glu-Ala-Asp) box helicase5 (DDX5, also known as p68) is found to act as a component of the Mastermind-like1(Maml1) protein complex for transcriptional coactivation and is also a novel regulator of oncogenic Notch signaling in T-ALL leukemic cells [82]. Several reports have shown that the Notch glycosylating enzyme Manic Fringe (Mfng) is expressed in Ngn3+ cells, and the ectopic expression of Mfng is sufficient to induce chick endodermal cells to differentiate towards endocrine fate [83]. However, the deletion of Mfng in mice has no effect on pancreatic development, cell differentiation or function [84]. The glycosylation of the Notch receptors has been shown to prevent the binding of Jagged, thus inhibiting Jagged-mediated activation [85,86]. The ADAM family of metalloproteases have proteolytic activity and play a major role in the ectodomain shedding of proteins involved in paracrine signaling, cell adhesion and intracellular signaling [87,88,89]. During embryogenesis, ADAM9 and ADAM17 are expressed separately in insulin-producing β-cells and other islet cells, and ADAM10 is predominantly detectable in acinar cells [90]. ADAM10 and ADAM17 have been shown to cleave Notch at the S2 site, and ADAM9 and ADAM15 are also found to be involved in Notch signaling indirectly in neurons [91,92]. These ADAMs may play an essential role in the activation of Notch signaling during pancreatic development.

4. Notch Signaling Pathway in the Pancreas

4.1. Notch Signaling in Pancreatic Progenitor Cell Differentiation

Notch signaling is critical at multiple steps during pancreatic development [16,93]. The expression levels of the Notch ligand and receptor change during pancreatic development. Notch1 is the first expressed receptor in a subset of pancreatic epithelium cells at E9.5 and then is broadly expressed in the pancreatic epithelium at E14.5 [94]. Notch2 is strongly expressed at E11.5, and its expression is absolutely restricted to ductal cells at E15.5 [94]. Notch3 and Notch4 are expressed in the early pancreatic mesenchyme and then in the endothelial cells of the pancreas at E15.5 [94]. Dll1 is transiently expressed in the pancreatic duct epithelium between E9.5 and E11.5 [93,95], and Jag1 is the most abundant ligand during mid-gestation pancreatic development [96]. Recent studies have identified that CACs and terminal duct cells are unique locations of activated Notch signaling in the pancreas of adult mice and zebrafish [97,98]. In the adult pancreas, the reactivation of Notch is involved in phenotype modulation of adult rat exocrine cells and the proliferation of metaplastic exocrine cells during pancreas regeneration [99,100]. In the caerulein-induced pancreatitis mouse model, the expression of the exocrine genes disappeared in pancreatic exocrine cells, while genes normally associated with Notch components, such as Notch1, Notch2, Hes1, Jag2, and a low level of Dll1 were induced [101].
During the secondary transition, Notch regulates endocrine differentiation via a lateral inhibition mechanism. The lateral inhibition model proposes that the onset of Ngn3 expression initiates endocrine differentiation and activates the Notch ligand Delta. Subsequently, Delta binds with Notch receptors in neighboring cells to initiate the Notch signaling cascade and release the NICD. The activated NICD enters the nucleus to activate the target gene Hes1, which inhibits the expression of Ngn3 [75,76]. Ultimately, the activated Notch pathway prevents adjacent cells from adopting an endocrine fate (Figure 3). Most early studies have shown that the inactivation of Notch signaling accelerates the premature differentiation of the endocrine pancreas [20]. In Dll1-deficient mice, Pdx1+ progenitor cells within the pancreatic buds lacked expansion and differentiated prematurely into endocrine cells [93]. In zebrafish embryos, the inhibition of Jagged caused ectopic islet-cell differentiation [19]. The loss of Rbp-J at the initial stage of pancreatic development resulted in the rapid differentiation of α-and PP-cells and decreased numbers of Ngn3+ cells [102]. Furthermore, Rbp-J KO mice exhibited insulin-deficient diabetes [20,102]. Hes1-deficient mice showed pancreatic hypoplasia, which is caused by the depletion of pancreatic epithelial progenitors from the accelerated differentiation of pancreatic endocrine cells [72,103]. A similar phenotype has been observed in mice that over-express Ngn3 [104] or the intracellular form of Notch3 (a repressor of Notch signaling) [105]. Conversely, the enforced activation of Notch signaling blocks pancreatic progenitors differentiation into the endocrine and exocrine cells. In chicken embryos, activated Notch1 can block the expression of endocrine genes and prevent endocrine differentiation [18]. The lentiviral-mediated activation of Hes1 and intracellular domain of Notch (Notch-IC) in mouse dorsal pancreatic buds at E10.5 represses endocrine and exocrine differentiation [106]. Ectopic Notch activation in zebrafish embryos can inhibit acinar and β-cell differentiation [106]. When Notch was overexpressed in the E9.5 or E11.5 mouse embryonic pancreas, the differentiation of Hes1+ cells was blocked; however, ectopic Notch activation at E15.5 does not perturb exocrine differentiation [107]. The sustained Notch1 signaling in Pdx1-NICD transgenic mice prevents the differentiation of pancreatic acinar and endocrine cells [17].
Figure 3. Notch-mediated lateral inhibition in pancreatic multipotent progenitor cell differentiation.
Figure 3. Notch-mediated lateral inhibition in pancreatic multipotent progenitor cell differentiation.
Ijms 17 00048 g003
Furthermore, recent studies have implied that the Notch-mediated regulation of endocrine differentiation is more complex than the classical lateral inhibition model suggests. For example, E9.5 Dll1 mutant mice show a dramatic decrease in their number of Ngn3+ cells, which suggests that Dll1 is required for the continuous formation of Ngn3+ endocrine precursors [21]. Shih et al. [22], demonstrated that Notch induces the expression of the Ngn3 gene activator Sox9, which may provide further evidence that Notch initiates the endocrine lineage. Some current gene-targeting studies focused on the Notch signaling components found that inactivated Notch signaling promotes the differentiation or transdifferentiation of MPCs, CACs or adult duct cells to the acinar lineage, which suggests that Notch may specify the endocrine lineage. Zebrafish embryos that were injected with RNA encoding a dominant-negative Suppressor of Hairless showed accelerated exocrine cells compared with the controls [106]. Similarly, a study by Cras and his colleagues demonstrated that the deletion of presenilin1 (Psen1) and presenilin2 (Psen2) drives Ngn3-expressing cells to differentiate into acinar cells [108]. Once cultured in the presence of the γ-secretase inhibitor DAPT, mouse pancreatic progenitors differentiate into acinar cells [23]. When Notch signaling is suppressed through the mosaic overexpression of a Notch signaling antagonist, dominant-negative mastermind-like1 (dnMaml), the pancreatic progenitor cells subsequently differentiate into acinar cells [24]. The inactivation of Mib1 in the endoderm causes a loss of endocrine cells and an increase in exocrine cells in the proximal domain, which suggested that Notch signalling is required to prompt MPCs to adopt an endocrine fate [79]. Upon Rbp-J deletion, mouse CACs undergo a rapid conversion to acinar cells [109]. In addition, the differentiation of acinar cells is accelerated by Hes1 depletion but is suppressed by NICD induction in adult mouse Sox9-expressing cells [25].
The causal role of Notch signaling in the patterning of MPCs into endocrine cells has been evaluated in some studies in different aspects. Genetic interactions have suggested that γ-secretase and Notch2 act in a non-canonical mechanism to sequester Rbp-J away from Ptf1a, which secures mouse Ngn3-positive progenitor cells to the endocrine fate [108]. Afelik et al. [24], found that Notch signaling is required to establish a duct and endocrine identity through the activation of Nkx6.1, which is bound to Rbp-J. Some studies revealed that Notch does not function in an on-off mode, but that Notch signaling seems to act in a concentration-dependent manner [22,26,76]. A study by Shih et al. [22], proposed a model in which high Notch expression activates Hes1 and Sox9 expression, resulting in the generation of pancreatic duct cells, while low Notch expression activates Sox9 but not Hes1, resulting in Ngn3 activation and endocrine differentiation. A study on the dynamic assessment of Notch signaling in the zebrafish intrapancreatic duct found that the proliferation and differentiation of MPCs are regulated by different levels of Notch signaling [26]. It has been reported that the hyperactivation of Notch signaling could convert the proliferative Notch-responsive MPCs to the quiescent state, hypo-activation of the Notch pathway induces the quiescent MPCs to the proliferative state, and strong down-regulation of Notch signaling promotes MPCs differentiate towards endocrine cells [26]. However, there are gaps in our understanding of the timing and extent of Notch ligand-receptor interactions and how this affects the behavior of MPCs. Therefore, there may be even more complexity in the Notch-mediated regulation of pancreatic development.

4.2. Notch Signaling in Pancreatic Progenitors Maintenance

The Notch signaling pathway has been found to maintain proliferation and prevent the precocious differentiation of pancreatic progenitor cells [18,110]. The activation of Notch at the “primary transition” maintains the pancreatic state, allowing the coordination of epithelial outgrowth and helping the pancreatic buds reach their destined size.
The transcription factor network is crucial for the maintenance and expansion of MPCs. Pdx1 and Ptf1a can form autoregulatory loops and feed-forward loop to retain and expand the progenitor cells during early pancreatic development [111]. The inactivation of Ptf1a in mice results in pancreatic hypoplasia, glucose intolerance and the transformation of pancreatic progenitors to a duodenal fate [112,113]. The expression of Pdx1 and Ptf1a is also regulated by fibroblast growth factor 10 (FGF10) and Notch signaling [111]. FGF10 signaling promotes the expansion of pancreatic epithelial cells through Sox9 and Hes1 activation [31,110,114,115]. Hes1 is reported to regulate the binary decision choice of pancreatic progenitors, cell cycle exist or self-renewal maintenance, through suppression of P57 and P27, which are cyclin-dependent kinase inhibitors [116,117] (Figure 4). In pancreatic progenitors, the inactivation of Hes1 could increase the expression of the P57 gene, which leads to cell cycle arrest, early differentiation and the depletion of the progenitor pool [117]. Moreover, Sox9 promotes pancreatic progenitor expansion by modulating the FGF-receptor (FGFR), Notch and Wnt signal transduction [31,45,118]. It has been reported that the pancreas-specific Sox9-deficient progenitors exhibit reduced proliferation and a low level of Hes1 [45]. Sox9 cell-autonomously controls the expression of FGFR2b in pancreatic progenitors [31]. In isolated human islet-epithelial clusters, the knockdown of Sox9 resulted in a decrease in pGSK3β, nuclear β-catenin and the Wnt signaling target gene cyclin D1 [118]. Notch signaling also regulates Sox9 expression in the pancreas [22,25]. Notch positively regulates Sox9 expression in a Hes1-independent manner in the pancreatic duct cells of Sox9CreERT2 mice [25]. The progenitor-intrinsic transcription factors Pdx1, Ptf1a and Sox9, as well as Notch, Wnt and FGF10 signaling, compose a complex network for the maintenance of MPCs.
Figure 4. Notch signaling in the proliferation of pancreatic multipotent progenitor cells.
Figure 4. Notch signaling in the proliferation of pancreatic multipotent progenitor cells.
Ijms 17 00048 g004

4.3. Notch Signaling in Adult Pancreatic Cell Plasticity

Because the components of the Notch signaling pathway are not normally expressed in terminally differentiated pancreatic cells, most mature cells lose their responsiveness to Notch signaling. However, in-depth studies have indicated that Notch signaling is involved in controlling the plasticity of adult, terminally differentiated pancreatic cells. Importantly, the dedifferentiation and transdifferentiation of terminally differentiated pancreatic cells are associated with Notch pathway reactivation during regeneration following pancreatitis [28,99,119], pancreatic neoplasia [120], and acino-ductal metaplasia [121,122]. For instance, when the pancreas is injured by pancreatitis, the exocrine acinar cells lose their differentiated characteristics and present acino-ductal metaplasia, which strongly upregulates the expression of the receptors Notch1 and 2 and the target genes Hes1, Hey1, and Hey2 [27]. Adult pancreatic duct cells retain their plasticity to differentiate into endocrine or acinar cell types, which is controlled by Notch signaling and Sox9 cooperatively [25]. When dissociated adult human β-cells are cultured in serum-containing medium, the β-cells dedifferentiate, and they enter into the cell cycle, which correlates with the activation of the Notch pathway and the down-regulation of P57 [123]. When Hes1 expression was down-regulated by shRNA, the dedifferentiated β cells redifferentiate into insulin-expressing cells [124]. Notch signaling is also reported as a gatekeeper of acinar-to-β-cell conversion in vitro [100,125]. Baeyens et al. [100], found that the growth factor-induced conversion of adult acinar cells to β-cells is negatively regulated by activated Notch1, which has the ability to prevent the re-expression of the pro-endocrine transcription factor Ngn3. Notch re-expression is deeply associated with the modulation of the proliferation of metaplastic cells and possible plays an important role in pancreatic regeneration.

5. The Crosstalk between Notch Signaling and the Wingless and INT-1 (Wnt)/Fibroblast Growth Factor (FGF) Pathway in the Pancreas

5.1. Notch/Wnt Crosstalk

Notch and Wnt signaling are key pathways that control the expansion and differentiation of stem/progenitor cells during embryogenesis, tissue formation and maintenance in adult homeostasis [126]. The Wnt pathway inhibits the specification of the pancreas in the early endoderm, whereas the pathway promotes the growth of the dorsal and ventral primordial buds, specifically the proliferation of acinar cells [127]. In the developing mouse pancreatic epithelium, the deletion of Wnt7b leads to pancreatic hypoplasia because of the reduced proliferation of pancreatic progenitor cells [128]. The ectopic stabilization of β-catenin before E11.5 in mouse embryos prevents the proper differentiation and expansion of early pancreatic progenitor cells [129]. Ectopic expression of β-catenin in mouse embryos at E18.5 causes the gross enlargement of the exocrine pancreas, which results in a dramatic increase in pancreas organ size [129]. Conversely, in pancreas-specific β-catenin knockout mice, the pancreas almost completely lacks acinar cells [130]. Meanwhile, gain- and loss-of-function experiments have proved that the Wnt pathway is involved in β-cell growth and survival. The proliferation of mouse islet cells and β-cells increases when they are treated with Wnt3a [131,132]. Conditional activated β-catenin promotes the expansion of β-cells in mice [129]. However, the addition of the soluble Wnt inhibitor Fz8-cysteine-rich domain (Fz8-CRD) or the conditional knock-in of the Wnt inhibitor Axin impaired the proliferation of neonatal mouse β-cells [129,131]. Likewise, studies have found that the Wnt signaling pathway plays a role in regulating glucose-stimulated insulin secretion in mature β-cells and is involved in lipid metabolism and glucose homeostasis [133,134]. The knockout of the low-density lipoprotein receptor-related protein 5 (LRP5), which is a Wnt co-receptor, results in glucose intolerance in mice [133].
In the early pancreatic lineage commitment, there is a repressive crosstalk between Notch and Wnt signaling. Notch pathway promotes the lineage commitment and differentiation of pancreatic progenitors, whereas Wnt signaling maintains the stem cell state. The opposing activities of Notch and Wnt have also been found in the skin, mammary glands and intestinal stem cells [135,136,137]. Similarly, both pathways can act on the same cell during the development of sensory bristle and epidermal cells in Drosophila and Xenopus [138,139]. In recent years, genetic analysis and proposed mathematical models have explained the dynamics of the crosstalk between the Notch and Wnt pathways. Here, we will summarize the major findings concerning the molecular mechanisms of the interactions between the two signaling pathways. First, Dishevelled 2 (Dvl2) plays a dual role, acting as an activator of Wnt signaling and an inhibitor of Notch activity. Dvl2 blocks Notch signaling directly after interacting with the Notch carboxyl terminus, which results in the disruption of the lateral inhibition signal mediated by Notch in the sensory mother cells (SMCs) of Drosophila [138] (Figure 5). Collu et al. [139], found that Dvl2 binds and inhibits Rbp-J proteins in order to suppress the transcriptional activity of the Rbp-J-NICD-Maml transcriptional activator complex during Xenopus epidermal development (Figure 5). The crosstalk mechanism is conserved between vertebrates and invertebrates, as Dvl2 targets the unique and common pathway component, Rbp-J, a core player in conventional Notch signaling. The similar inhibition of Notch signaling by Dvl2 has been shown during the establishment of planar polarity in the Drosophila eye and leg epithelium [140,141]. The regulation of Notch signaling by the Wnt pathway is shown by the negative effect of Wnt on the Dvl2-mediated GSK3β activity. GSK3β stabilizes the Notch-IC by binding and phosphorylating Notch-IC in the embryonic fibroblasts and N2a cells [142], and GSK-3β inhibition leads to the degradation of Notch-IC mediated by the proteasome (Figure 5). Furthermore, Notch can directly interact with β-catenin. Notch regulates the stability and activity of Armadillo/β-catenin and negatively regulates β-catenin/TCF transcription in Drosophila [143]. In neural precursor cells and vascular progenitors, NICD inhibits β-catenin activity directly by forming a Notch/β-catenin/Rbp-J complex to prevent β-catenin from binding its target sites [144]. The protein complex may play a critical role in cell fate determination in various organs. Lastly, Wnt signaling inhibits Notch activity through Pygopus2 (Pygo2) to promote self-renewal and to prevent the premature differentiation of mammary stem cells (MaSCs) [145]. Pygo2, a histone methylation reader and a context-dependent Wnt/β-catenin coactivator, facilitates the binding of β-catenin to the Notch3 locus and maintains Notch3 in a bivalent chromatin structure in MaSCs [145]. The crosstalk between Wnt and Notch reinforces the balance among stem cells, progenitors and differentiated cells within a tissue, and these confirmed molecular interactions might occur in the pancreas.
Figure 5. Inhibitory crosstalk between the Wingless and INT-1 (Wnt) and Notch pathways in pancreatic progenitor cells. The dashed lines signify that the mechanisms existing in the pancreas are unknown. The arrows represent activation whereas bar-headed lines represent inhibition.
Figure 5. Inhibitory crosstalk between the Wingless and INT-1 (Wnt) and Notch pathways in pancreatic progenitor cells. The dashed lines signify that the mechanisms existing in the pancreas are unknown. The arrows represent activation whereas bar-headed lines represent inhibition.
Ijms 17 00048 g005

5.2. Notch/FGF Crosstalk

FGF signaling from the pancreatic mesenchyme has been shown to play an essential role in pancreatic development and the pancreatic disease process. Studies using recombined models of embryonic tissues have shown that pancreatic buds can develop in vitro but will not undergo any growth or branching morphogenesis without the presence of the mesenchyme [146,147]. FGF10 signaling plays a crucial mitogenic role in driving the proliferation of pancreatic progenitor cells [64,148,149]. In FGF10−/− mouse embryos, the proliferation of Pdx1+ progenitor cells was reduced, but the growth, differentiation and branching morphogenesis of the pancreatic epithelia were arrested [148]. In mice, the persistent expression of FGF10 mediated by the Ipf1/Pdx1 promoter increased the proliferation of pancreatic progenitor cells and arrested them in a pluripotent state [110]. Furthermore, the expression of Notch1 and Hes1 was maintained in the pancreatic epithelium, along with the reduction of Ngn3 [110]. The persistent expression of FGF10 perturbs the expression of Suppressor enhancer lin12/Notch 1-like (Sel1l), which regulates pancreatic epithelial growth and differentiation by suppressing Notch signaling in mice [110,150]. The phenotype of mice with an overexpression of FGF10 is similar to that of the mice with Notch overexpression described above. Thus, Notch signaling is a critical downstream effector of FGF pathway-induced embryonic pancreatic epithelial proliferation (Figure 6). FGF-stimulated progenitor cell maintenance via Notch signaling has been previously reported in several developmental contexts. During tracheal cell invagination in Drosophila, the FGF-like ligand Branchless activates the FGF-receptor, and the downstream MAPK signaling causes the upregulation of Δ [151]. Likewise, FGF10 is capable of maintaining the dental epithelial precursor pool via the stimulation of Hes1 in Fringe-dependent or Fringe-independent manners in the developing tooth [152]. However, whether the above mechanisms exist in the pancreas is unknown (Figure 6). Recently, mouse genetic studies have revealed that there is a FGF10/FGFR2b/Sox9 feed-forward loop in early pancreatic progenitors to maintain their proliferation [31] (Figure 6). Thus, Sox9 acts as the conduit between FGF10 and Notch in the proliferation of progenitors.
Figure 6. The crosstalk between the fibroblast growth factor (FGF) and Notch pathways in pancreatic progenitors. The dashed lines signify that the mechanisms existing in the pancreas are unknown. The arrows represent activation whereas bar-headed lines represent inhibition.
Figure 6. The crosstalk between the fibroblast growth factor (FGF) and Notch pathways in pancreatic progenitors. The dashed lines signify that the mechanisms existing in the pancreas are unknown. The arrows represent activation whereas bar-headed lines represent inhibition.
Ijms 17 00048 g006
Classic culture explant experiments highlight the importance of the mesenchyme for exocrine pancreatic growth and differentiation [153,154]. In Pdx1-FGF4 transgenic mice, the pancreas has degenerated ductal and destructive endocrine tissue [155]. However, the exact function of FGF signaling on β-cell development remains controversial. For example, studies on the FGF10 knockout mice have suggested that FGF10 could directly and positively control the final number of β-cells [148,156,157]. Mice with attenuated FGFR1c have a decreased number of β-cells and develop diabetes. [158]. On the other hand, the overexpression of FGF10 in the pancreas inhibited endocrine fate by increasing Notch signaling in both mice and rats [110,159].

6. Conclusions and Future Perspective

The Notch pathway regulates cell fate and homeostasis during the development and postnatal life of self-renewing tissues. It not only plays important roles in the pancreas but also regulates self-renewal, lineage specification and differentiation of stem cells in other systems. Notch signaling functions at different time and at different levels during the development of the central nervous system (CNS). Initially, the Notch pathway enhances the neural precursor proliferation and represses their differentiation [160,161]. At later stages, Notch signaling promotes astrocyte differentiation and inhibits oligodendrocyte generation [162]. Notch signaling maintains self-renewal of the early muscle progenitors and regulates their differentiation during embryonic development and adulthood [163,164]. Studies with flies, nematodes, and vertebrates have revealed that Notch signaling is an evolutionarily conserved mechanism and specifies cell fates through local cell interactions in nearly all tissues [165,166,167]. However, there may be species- and tissue-specific differences in the precise roles of the Notch pathway in regulating stem cells and their fate. Notch signaling is an important player that operates sequentially and spatially to affect different aspects of pancreas formation. During early pancreatic development, distinct levels of Notch signaling at two different transition stages trigger the proliferation and differentiation of progenitors; strict control of the time and dosage of the Notch signaling components is necessary for proper organ homeostasis. We speculate that the Notch-mediated regulation of endocrine or exocrine differentiation may be more complex than we currently understand.

Acknowledgments

We apologize to the authors of those primary works that are not cited in this review due to space constraints. We are thankful to Yicheng Zhao and Delin Kong for their editing work on our manuscript and figures. This work was supported by the National Natural Science Foundation of China (No. 31472159, J1210053).

Author Contributions

Xu-Yan Li contributed to the drafting and editing of the manuscript. Wen-Jun Zhai participated in literature search. Chun-Bo Teng conceived the paper design, and supervised the writing, editing and proofreading of the paper. All authors have read and approved the final manuscript for publication.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Artavanis-Tsakonas, S.; Matsuno, K.; Fortini, M.E. Notch signaling. Science 1995, 268, 225–232. [Google Scholar] [CrossRef] [PubMed]
  2. Bray, S. Notch signalling in drosophila: Three ways to use a pathway. Semin. Cell Dev. Biol. 1998, 9, 591–597. [Google Scholar] [CrossRef] [PubMed]
  3. Buono, K.D.; Robinson, G.W.; Martin, C.; Shi, S.; Stanley, P.; Tanigaki, K.; Honjo, T.; Hennighausen, L. The canonical Notch/Rbp-j signaling pathway controls the balance of cell lineages in mammary epithelium during pregnancy. Dev. Biol. 2006, 293, 565–580. [Google Scholar] [CrossRef] [PubMed]
  4. Kopan, R.; Ilagan, M.X. The canonical Notch signaling pathway: Unfolding the activation mechanism. Cell 2009, 137, 216–233. [Google Scholar] [CrossRef] [PubMed]
  5. Yugawa, T.; Nishino, K.; Ohno, S.; Nakahara, T.; Fujita, M.; Goshima, N.; Umezawa, A.; Kiyono, T. Noncanonical Notch signaling limits self-renewal of human epithelial and induced pluripotent stem cells through rock activation. Mol. Cell. Biol. 2013, 33, 4434–4447. [Google Scholar] [CrossRef] [PubMed]
  6. Layden, M.J.; Martindale, M.Q. Non-canonical Notch signaling represents an ancestral mechanism to regulate neural differentiation. Evodevo 2014, 5, 30. [Google Scholar] [CrossRef] [PubMed]
  7. Pezeron, G.; Millen, K.; Boukhatmi, H.; Bray, S. Notch directly regulates the cell morphogenesis genes Reck, talin and trio in adult muscle progenitors. J. Cell Sci. 2014, 127, 4634–4644. [Google Scholar] [CrossRef] [PubMed]
  8. De Back, W.; Zhou, J.X.; Brusch, L. On the role of lateral stabilization during early patterning in the pancreas. J. R. Soc. Interface 2013, 10, 20120766. [Google Scholar] [CrossRef] [PubMed]
  9. Kim, W.; Shin, Y.K.; Kim, B.J.; Egan, J.M. Notch signaling in pancreatic endocrine cell and diabetes. Biochem. Biophys. Res. Commun. 2010, 392, 247–251. [Google Scholar] [CrossRef] [PubMed]
  10. Le Caignec, C. Human diseases and Notch receptors. Med. Sci. 2011, 27, 593–595. [Google Scholar]
  11. Polychronidou, E.; Vlachakis, D.; Vlamos, P.; Baumann, M.; Kossida, S. Notch signaling and ageing. Adv. Exp. Med. Biol. 2015, 822, 25–36. [Google Scholar] [PubMed]
  12. Bi, P.; Kuang, S. Notch signaling as a novel regulator of metabolism. Trends Endocrinol. Metab. 2015, 26, 248–255. [Google Scholar] [CrossRef] [PubMed]
  13. Grishina, I.B. Mini-review: Does Notch promote or suppress cancer? New findings and old controversies. Am. J. Clin. Exp. Urol. 2015, 3, 24–27. [Google Scholar] [PubMed]
  14. Murtaugh, L.C.; Stanger, B.Z.; Kwan, K.M.; Melton, D.A. Notch signaling controls multiple steps of pancreatic differentiation. Proc. Natl. Acad. Sci. USA 2003, 100, 14920–14925. [Google Scholar] [CrossRef] [PubMed]
  15. Greenwood, A.L.; Li, S.; Jones, K.; Melton, D.A. Notch signaling reveals developmental plasticity of pax4(+) pancreatic endocrine progenitors and shunts them to a duct fate. Mech. Dev. 2007, 124, 97–107. [Google Scholar] [CrossRef] [PubMed]
  16. Afelik, S.; Jensen, J. Notch signaling in the pancreas: Patterning and cell fate specification. Dev. Biol. 2013, 2, 531–544. [Google Scholar] [CrossRef] [PubMed]
  17. Hald, J.; Hjorth, J.P.; German, M.S.; Madsen, O.D.; Serup, P.; Jensen, J. Activated Notch1 prevents differentiation of pancreatic acinar cells and attenuate endocrine development. Dev. Biol. 2003, 260, 426–437. [Google Scholar] [CrossRef]
  18. Ahnfelt-Ronne, J.; Hald, J.; Bodker, A.; Yassin, H.; Serup, P.; Hecksher-Sorensen, J. Preservation of proliferating pancreatic progenitor cells by δ-Notch signaling in the embryonic chicken pancreas. BMC Dev. Biol. 2007, 7, 63. [Google Scholar] [CrossRef] [PubMed]
  19. Zecchin, E.; Filippi, A.; Biemar, F.; Tiso, N.; Pauls, S.; Ellertsdottir, E.; Gnugge, L.; Bortolussi, M.; Driever, W.; Argenton, F. Distinct δ and jagged genes control sequential segregation of pancreatic cell types from precursor pools in zebrafish. Dev. Biol. 2007, 301, 192–204. [Google Scholar] [CrossRef] [PubMed]
  20. Nakhai, H.; Siveke, J.T.; Klein, B.; Mendoza-Torres, L.; Mazur, P.K.; Algul, H.; Radtke, F.; Strobl, L.; Zimber-Strobl, U.; Schmid, R.M. Conditional ablation of Notch signaling in pancreatic development. Development 2008, 135, 2757–2765. [Google Scholar] [CrossRef] [PubMed]
  21. Ahnfelt-Ronne, J.; Jorgensen, M.C.; Klinck, R.; Jensen, J.N.; Fuchtbauer, E.M.; Deering, T.; MacDonald, R.J.; Wright, C.V.; Madsen, O.D.; Serup, P. Ptf1a-mediated control of Dll1 reveals an alternative to the lateral inhibition mechanism. Development 2012, 139, 33–45. [Google Scholar] [CrossRef] [PubMed]
  22. Shih, H.P.; Kopp, J.L.; Sandhu, M.; Dubois, C.L.; Seymour, P.A.; Grapin-Botton, A.; Sander, M. A Notch-dependent molecular circuitry initiates pancreatic endocrine and ductal cell differentiation. Development 2012, 139, 2488–2499. [Google Scholar] [CrossRef] [PubMed]
  23. Shimajiri, Y.; Kosaka, Y.; Scheel, D.W.; Lynn, F.C.; Kishimoto, N.; Wang, J.; Zhao, S.; German, M.S. A mouse model for monitoring islet cell genesis and developing therapies for diabetes. Dis. Model. Mech. 2011, 4, 268–276. [Google Scholar] [CrossRef] [PubMed]
  24. Afelik, S.; Qu, X.; Hasrouni, E.; Bukys, M.A.; Deering, T.; Nieuwoudt, S.; Rogers, W.; Macdonald, R.J.; Jensen, J. Notch-mediated patterning and cell fate allocation of pancreatic progenitor cells. Development 2012, 139, 1744–1753. [Google Scholar] [CrossRef] [PubMed]
  25. Hosokawa, S.; Furuyama, K.; Horiguchi, M.; Aoyama, Y.; Tsuboi, K.; Sakikubo, M.; Goto, T.; Hirata, K.; Tanabe, W.; Nakano, Y.; et al. Impact of Sox9 dosage and Hes1-mediated Notch signaling in controlling the plasticity of adult pancreatic duct cells in mice. Sci. Rep. 2015, 5, 8518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Ninov, N.; Borius, M.; Stainier, D.Y. Different levels of Notch signaling regulate quiescence, renewal and differentiation in pancreatic endocrine progenitors. Development 2012, 139, 1557–1567. [Google Scholar] [CrossRef] [PubMed]
  27. Bhanot, U.; Kohntop, R.; Hasel, C.; Moller, P. Evidence of Notch pathway activation in the ectatic ducts of chronic pancreatitis. J. Pathol. 2008, 214, 312–319. [Google Scholar] [CrossRef] [PubMed]
  28. Pinho, A.V.; Rooman, I.; Reichert, M.; de Medts, N.; Bouwens, L.; Rustgi, A.K.; Real, F.X. Adult pancreatic acinar cells dedifferentiate to an embryonic progenitor phenotype with concomitant activation of a senescence programme that is present in chronic pancreatitis. Gut 2011, 60, 958–966. [Google Scholar] [CrossRef] [PubMed]
  29. Chetty, R.; Serra, S.; Salahshor, S.; Alsaad, K.; Shih, W.; Blaszyk, H.; Woodgett, J.R.; Tsao, M.S. Expression of wnt-signaling pathway proteins in intraductal papillary mucinous neoplasms of the pancreas: A tissue microarray analysis. Hum. Pathol. 2006, 37, 212–217. [Google Scholar] [CrossRef] [PubMed]
  30. Lin, C.L.; Vuguin, P.M. Determinants of pancreatic islet development in mice and men: A focus on the role of transcription factors. Horm. Res. Paediatr. 2012, 77, 205–213. [Google Scholar] [CrossRef] [PubMed]
  31. Seymour, P.A.; Shih, H.P.; Patel, N.A.; Freude, K.K.; Xie, R.; Lim, C.J.; Sander, M. A Sox9/FGF feed-forward loop maintains pancreatic organ identity. Development 2012, 139, 3363–3372. [Google Scholar] [CrossRef] [PubMed]
  32. Wolfe-Coote, S.A.; Louw, J.; Poerstamper, H.M.; Du Toit, D.F. Do the pancreatic primordial buds in embryogenesis have the potential to produce all pancreatic endocrine cells? Med. Hypotheses 1990, 31, 313–317. [Google Scholar] [CrossRef]
  33. Merino, P.L. Developmental biology of the pancreas. Cell Biochem. Biophys. 2004, 40, 127–142. [Google Scholar] [CrossRef]
  34. Herrera, P.L. Adult insulin- and glucagon-producing cells differentiate from two independent cell lineages. Development 2000, 127, 2317–2322. [Google Scholar] [PubMed]
  35. Teitelman, G.; Alpert, S.; Polak, J.M.; Martinez, A.; Hanahan, D. Precursor cells of mouse endocrine pancreas coexpress insulin, glucagon and the neuronal proteins tyrosine hydroxylase and neuropeptide y, but not pancreatic polypeptide. Development 1993, 118, 1031–1039. [Google Scholar] [PubMed]
  36. Kemp, J.D.; Walther, B.T.; Rutter, W.J. Protein synthesis during the secondary developmental transition of the embryonic rat pancreas. J. Biol. Chem. 1972, 247, 3941–3952. [Google Scholar] [PubMed]
  37. Wang, J.; Kilic, G.; Aydin, M.; Burke, Z.; Oliver, G.; Sosa-Pineda, B. Prox1 activity controls pancreas morphogenesis and participates in the production of “secondary transition” pancreatic endocrine cells. Dev. Biol. 2005, 286, 182–194. [Google Scholar] [CrossRef] [PubMed]
  38. Bouwens, L.; Rooman, I. Regulation of pancreatic β-cell mass. Physiol. Rev. 2005, 85, 1255–1270. [Google Scholar] [CrossRef] [PubMed]
  39. Offield, M.F.; Jetton, T.L.; Labosky, P.A.; Ray, M.; Stein, R.W.; Magnuson, M.A.; Hogan, B.L.; Wright, C.V. Pdx-1 is required for pancreatic outgrowth and differentiation of the rostral duodenum. Development 1996, 122, 983–995. [Google Scholar] [PubMed]
  40. Arda, H.E.; Benitez, C.M.; Kim, S.K. Gene regulatory networks governing pancreas development. Dev. Cell 2013, 25, 5–13. [Google Scholar] [CrossRef] [PubMed]
  41. Bonal, C.; Herrera, P.L. Genes controlling pancreas ontogeny. Int. J. Dev. Biol. 2008, 52, 823–835. [Google Scholar] [CrossRef] [PubMed]
  42. Wu, H.; MacFarlane, W.M.; Tadayyon, M.; Arch, J.R.; James, R.F.; Docherty, K. Insulin stimulates pancreatic-duodenal homoeobox factor-1 (PDX1) DNA-binding activity and insulin promoter activity in pancreatic β cells. Biochem. J. 1999, 344, 813–818. [Google Scholar] [CrossRef] [PubMed]
  43. Lynn, F.C.; Smith, S.B.; Wilson, M.E.; Yang, K.Y.; Nekrep, N.; German, M.S. Sox9 coordinates a transcriptional network in pancreatic progenitor cells. Proc. Natl. Acad. Sci. USA 2007, 104, 10500–10505. [Google Scholar] [CrossRef] [PubMed]
  44. Hald, J.; Sprinkel, A.E.; Ray, M.; Serup, P.; Wright, C.; Madsen, O.D. Generation and characterization of Ptf1a antiserum and localization of Ptf1a in relation to Nkx6.1 and Pdx1 during the earliest stages of mouse pancreas development. J. Histochem. Cytochem. 2008, 56, 587–595. [Google Scholar] [CrossRef] [PubMed]
  45. Seymour, P.A.; Freude, K.K.; Tran, M.N.; Mayes, E.E.; Jensen, J.; Kist, R.; Scherer, G.; Sander, M. Sox9 is required for maintenance of the pancreatic progenitor cell pool. Proc. Natl. Acad. Sci. USA 2007, 104, 1865–1870. [Google Scholar] [CrossRef] [PubMed]
  46. Rodolosse, A.; Chalaux, E.; Adell, T.; Hagege, H.; Skoudy, A.; Real, F.X. Ptf1α/p48 transcription factor couples proliferation and differentiation in the exocrine pancreas [corrected]. Gastroenterology 2004, 127, 937–949. [Google Scholar] [CrossRef] [PubMed]
  47. Gradwohl, G.; Dierich, A.; LeMeur, M.; Guillemot, F. Neurogenin3 is required for the development of the four endocrine cell lineages of the pancreas. Proc. Natl. Acad. Sci. USA 2000, 97, 1607–1611. [Google Scholar] [CrossRef] [PubMed]
  48. Beucher, A.; Gjernes, E.; Collin, C.; Courtney, M.; Meunier, A.; Collombat, P.; Gradwohl, G. The homeodomain-containing transcription factors Arx and Pax4 control enteroendocrine subtype specification in mice. PLoS ONE 2012, 7, e36449. [Google Scholar] [CrossRef] [PubMed]
  49. Juhl, K.; Sarkar, S.A.; Wong, R.; Jensen, J.; Hutton, J.C. Mouse pancreatic endocrine cell transcriptome defined in the embryonic Ngn3-null mouse. Diabetes 2008, 57, 2755–2761. [Google Scholar] [CrossRef] [PubMed]
  50. Kim, S.Y.; Rane, S.G. The Cdk4-E2F1 pathway regulates early pancreas development by targeting PDX1+ progenitors and Ngn3+ endocrine precursors. Development 2011, 138, 1903–1912. [Google Scholar] [CrossRef] [PubMed]
  51. Piccand, J.; Meunier, A.; Merle, C.; Jia, Z.; Barnier, J.V.; Gradwohl, G. Pak3 promotes cell cycle exit and differentiation of β-cells in the embryonic pancreas and is necessary to maintain glucose homeostasis in adult mice. Diabetes 2014, 63, 203–215. [Google Scholar] [CrossRef] [PubMed]
  52. Krapp, A.; Knofler, M.; Frutiger, S.; Hughes, G.J.; Hagenbuchle, O.; Wellauer, P.K. The p48 DNA-binding subunit of transcription factor Ptf1 is a new exocrine pancreas-specific basic helix-loop-helix protein. EMBO J. 1996, 15, 4317–4329. [Google Scholar] [PubMed]
  53. Cockell, M.; Stevenson, B.J.; Strubin, M.; Hagenbuchle, O.; Wellauer, P.K. Identification of a cell-specific DNA-binding activity that interacts with a transcriptional activator of genes expressed in the acinar pancreas. Mol. Cell. Biol. 1989, 9, 2464–2476. [Google Scholar] [CrossRef] [PubMed]
  54. Dong, P.D.; Provost, E.; Leach, S.D.; Stainier, D.Y. Graded levels of Ptf1a differentially regulate endocrine and exocrine fates in the developing pancreas. Genes Dev. 2008, 22, 1445–1450. [Google Scholar] [CrossRef] [PubMed]
  55. Schaffer, A.E.; Freude, K.K.; Nelson, S.B.; Sander, M. Nkx6 transcription factors and Ptf1a function as antagonistic lineage determinants in multipotent pancreatic progenitors. Dev. Cell 2010, 18, 1022–1029. [Google Scholar] [CrossRef] [PubMed]
  56. Lynn, F.C.; Skewes-Cox, P.; Kosaka, Y.; McManus, M.T.; Harfe, B.D.; German, M.S. Microrna expression is required for pancreatic islet cell genesis in the mouse. Diabetes 2007, 56, 2938–2945. [Google Scholar] [CrossRef] [PubMed]
  57. Latreille, M.; Hausser, J.; Stutzer, I.; Zhang, Q.; Hastoy, B.; Gargani, S.; Kerr-Conte, J.; Pattou, F.; Zavolan, M.; Esguerra, J.L.; et al. Microrna-7a regulates pancreatic β cell function. J. Clin. Investig. 2014, 124, 2722–2735. [Google Scholar] [CrossRef] [PubMed]
  58. Poy, M.N.; Hausser, J.; Trajkovski, M.; Braun, M.; Collins, S.; Rorsman, P.; Zavolan, M.; Stoffel, M. miR-375 maintains normal pancreatic α- and β-cell mass. Proc. Natl. Acad. Sci. USA 2009, 106, 5813–5818. [Google Scholar] [CrossRef] [PubMed]
  59. Zhang, Z.W.; Men, T.; Feng, R.C.; Li, Y.C.; Zhou, D.; Teng, C.B. miR-375 inhibits proliferation of mouse pancreatic progenitor cells by targeting YAP1. Cell. Physiol. Biochem. 2013, 32, 1808–1817. [Google Scholar] [PubMed]
  60. Zhang, Z.W.; Zhang, L.Q.; Ding, L.; Wang, F.; Sun, Y.J.; An, Y.; Zhao, Y.; Li, Y.H.; Teng, C.B. Microrna-19b downregulates insulin 1 through targeting transcription factor neurod1. FEBS Lett. 2011, 585, 2592–2598. [Google Scholar] [PubMed]
  61. Yang, Y.; Ding, L.; An, Y.; Zhang, Z.W.; Lang, Y.; Tai, S.; Guo, F.; Teng, C.B. miR-18a regulates expression of the pancreatic transcription factor Ptf1a in pancreatic progenitor and acinar cells. FEBS Lett. 2012, 586, 422–427. [Google Scholar] [PubMed]
  62. Joglekar, M.V.; Parekh, V.S.; Mehta, S.; Bhonde, R.R.; Hardikar, A.A. Microrna profiling of developing and regenerating pancreas reveal post-transcriptional regulation of neurogenin3. Dev. Biol. 2007, 311, 603–612. [Google Scholar] [PubMed]
  63. Cano, D.A.; Soria, B.; Martin, F.; Rojas, A. Transcriptional control of mammalian pancreas organogenesis. Cell. Mol. Life Sci. 2014, 71, 2383–2402. [Google Scholar] [CrossRef] [PubMed]
  64. Talavera-Adame, D.; Dafoe, D.C. Endothelium-derived essential signals involved in pancreas organogenesis. World J. Exp. Med. 2015, 5, 40–49. [Google Scholar] [CrossRef] [PubMed]
  65. Fleming, R.J.; Purcell, K.; Artavanis-Tsakonas, S. The Notch receptor and its ligands. Trends Cell Biol. 1997, 7, 437–441. [Google Scholar] [CrossRef]
  66. Bray, S. Notch. Curr. Biol. 2000, 10, R433–R435. [Google Scholar] [CrossRef]
  67. Logeat, F.; Bessia, C.; Brou, C.; LeBail, O.; Jarriault, S.; Seidah, N.G.; Israel, A. The Notch1 receptor is cleaved constitutively by a furin-like convertase. Proc. Natl. Acad. Sci. USA 1998, 95, 8108–8112. [Google Scholar] [CrossRef] [PubMed]
  68. Lake, R.J.; Grimm, L.M.; Veraksa, A.; Banos, A.; Artavanis-Tsakonas, S. In vivo analysis of the Notch receptor s1 cleavage. PLoS ONE 2009, 4, e6728. [Google Scholar] [CrossRef] [PubMed]
  69. Lubman, O.Y.; Ilagan, M.X.; Kopan, R.; Barrick, D. Quantitative dissection of the Notch:CSL interaction: Insights into the Notch-mediated transcriptional switch. J. Mol. Biol. 2007, 365, 577–589. [Google Scholar] [CrossRef] [PubMed]
  70. Bray, S.; Furriols, M. Notch pathway: Making sense of suppressor of hairless. Curr. Biol. 2001, 11, R217–R221. [Google Scholar] [CrossRef]
  71. Bray, S.; Bernard, F. Notch targets and their regulation. Curr. Top. Dev. Biol. 2010, 92, 253–275. [Google Scholar] [PubMed]
  72. Fukuda, A.; Kawaguchi, Y.; Furuyama, K.; Kodama, S.; Horiguchi, M.; Kuhara, T.; Koizumi, M.; Boyer, D.F.; Fujimoto, K.; Doi, R.; et al. Ectopic pancreas formation in Hes1 -knockout mice reveals plasticity of endodermal progenitors of the gut, bile duct, and pancreas. J. Clin. Investig. 2006, 116, 1484–1493. [Google Scholar] [CrossRef] [PubMed]
  73. Murai, K.; Philpott, A.; Jones, P.H. Hes6 is required for the neurogenic activity of neurogenin and neurod. PLoS ONE 2011, 6, e27880. [Google Scholar] [CrossRef] [PubMed]
  74. Petrovic, J.; Galvez, H.; Neves, J.; Abello, G.; Giraldez, F. Differential regulation of Hes/Hey genes during inner ear development. Dev. Neurobiol. 2015, 75, 703–720. [Google Scholar] [CrossRef] [PubMed]
  75. Lee, J.C.; Smith, S.B.; Watada, H.; Lin, J.; Scheel, D.; Wang, J.; Mirmira, R.G.; German, M.S. Regulation of the pancreatic pro-endocrine gene neurogenin3. Diabetes 2001, 50, 928–936. [Google Scholar] [CrossRef] [PubMed]
  76. Qu, X.; Afelik, S.; Jensen, J.N.; Bukys, M.A.; Kobberup, S.; Schmerr, M.; Xiao, F.; Nyeng, P.; Veronica Albertoni, M.; Grapin-Botton, A.; et al. Notch-mediated post-translational control of Ngn3 protein stability regulates pancreatic patterning and cell fate commitment. Dev. Biol. 2013, 376, 1–12. [Google Scholar] [CrossRef] [PubMed]
  77. Sakano, D.; Kato, A.; Parikh, N.; McKnight, K.; Terry, D.; Stefanovic, B.; Kato, Y. Bcl6 canalizes Notch-dependent transcription, excluding mastermind-like1 from selected target genes during left-right patterning. Dev. Cell 2010, 18, 450–462. [Google Scholar] [CrossRef] [PubMed]
  78. Itoh, M.; Kim, C.H.; Palardy, G.; Oda, T.; Jiang, Y.J.; Maust, D.; Yeo, S.Y.; Lorick, K.; Wright, G.J.; Ariza-McNaughton, L.; et al. Mind bomb is a ubiquitin ligase that is essential for efficient activation of Notch signaling by δ. Dev. Cell 2003, 4, 67–82. [Google Scholar] [CrossRef]
  79. Horn, S.; Kobberup, S.; Jorgensen, M.C.; Kalisz, M.; Klein, T.; Kageyama, R.; Gegg, M.; Lickert, H.; Lindner, J.; Magnuson, M.A.; et al. Mind bomb 1 is required for pancreatic β-cell formation. Proc. Natl. Acad. Sci. USA 2012, 109, 7356–7361. [Google Scholar] [CrossRef] [PubMed]
  80. Kang, K.; Lee, D.; Hong, S.; Park, S.G.; Song, M.R. The E3 ligase mind bomb-1 (Mib1) modulates δ-Notch signaling to control neurogenesis and gliogenesis in the developing spinal cord. J. Biol. Chem. 2013, 288, 2580–2592. [Google Scholar] [CrossRef] [PubMed]
  81. Ishitani, T.; Hirao, T.; Suzuki, M.; Isoda, M.; Ishitani, S.; Harigaya, K.; Kitagawa, M.; Matsumoto, K.; Itoh, M. Nemo-like kinase suppresses Notch signalling by interfering with formation of the Notch active transcriptional complex. Nat. Cell Biol. 2010, 12, 278–285. [Google Scholar] [CrossRef] [PubMed]
  82. Lin, S.; Tian, L.; Shen, H.; Gu, Y.; Li, J.L.; Chen, Z.; Sun, X.; You, M.J.; Wu, L. DDX5 is a positive regulator of oncogenic Notch1 signaling in T cell acute lymphoblastic leukemia. Oncogene 2013, 32, 4845–4853. [Google Scholar] [CrossRef] [PubMed]
  83. Xu, Y.; Wang, S.; Zhang, J.; Zhao, A.; Stanger, B.Z.; Gu, G. The fringe molecules induce endocrine differentiation in embryonic endoderm by activating cMyt1/cMyt3. Dev. Biol. 2006, 297, 340–349. [Google Scholar] [CrossRef] [PubMed]
  84. Svensson, P.; Bergqvist, I.; Norlin, S.; Edlund, H. Mfng is dispensable for mouse pancreas development and function. Mol. Cell. Biol. 2009, 29, 2129–2138. [Google Scholar] [CrossRef] [PubMed]
  85. Rana, N.A.; Haltiwanger, R.S. Fringe benefits: Functional and structural impacts of O-glycosylation on the extracellular domain of Notch receptors. Curr. Opin. Struct. Biol. 2011, 21, 583–589. [Google Scholar] [CrossRef] [PubMed]
  86. Takeuchi, H.; Haltiwanger, R.S. Significance of glycosylation in Notch signaling. Biochem. Biophys. Res. Commun. 2014, 453, 235–242. [Google Scholar] [CrossRef] [PubMed]
  87. Blobel, C.P. Adams: Key components in egfr signalling and development. Nat. Rev. Mol. Cell Biol. 2005, 6, 32–43. [Google Scholar] [CrossRef] [PubMed]
  88. Groot, A.J.; Vooijs, M.A. The role of adams in Notch signaling. Adv. Exp. Med. Biol. 2012, 727, 15–36. [Google Scholar] [PubMed]
  89. Christian, L.M. The adam family: Insights into Notch proteolysis. Fly 2012, 6, 30–34. [Google Scholar] [CrossRef] [PubMed]
  90. Asayesh, A.; Alanentalo, T.; Khoo, N.K.; Ahlgren, U. Developmental expression of metalloproteases ADAM 9, 10, and 17 becomes restricted to divergent pancreatic compartments. Dev. Dny. 2005, 232, 1105–1114. [Google Scholar] [CrossRef] [PubMed]
  91. Dyczynska, E.; Sun, D.; Yi, H.; Sehara-Fujisawa, A.; Blobel, C.P.; Zolkiewska, A. Proteolytic processing of δ-like 1 by ADAM proteases. J. Biol. Chem. 2007, 282, 436–444. [Google Scholar] [CrossRef] [PubMed]
  92. Tousseyn, T.; Thathiah, A.; Jorissen, E.; Raemaekers, T.; Konietzko, U.; Reiss, K.; Maes, E.; Snellinx, A.; Serneels, L.; Nyabi, O.; et al. ADAM10, the rate-limiting protease of regulated intramembrane proteolysis of Notch and other proteins, is processed by ADAMs-9, ADAMs-15, and the γ-secretase. J. Biol. Chem. 2009, 284, 11738–11747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Apelqvist, A.; Li, H.; Sommer, L.; Beatus, P.; Anderson, D.J.; Honjo, T.; Hrabe de Angelis, M.; Lendahl, U.; Edlund, H. Notch signalling controls pancreatic cell differentiation. Nature 1999, 400, 877–881. [Google Scholar] [PubMed]
  94. Lammert, E.; Brown, J.; Melton, D.A. Notch gene expression during pancreatic organogenesis. Mech. Dev. 2000, 94, 199–203. [Google Scholar] [CrossRef]
  95. Bettenhausen, B.; Hrabe de Angelis, M.; Simon, D.; Guenet, J.L.; Gossler, A. Transient and restricted expression during mouse embryogenesis of Dll1, a murine gene closely related to Drosophila δ. Development 1995, 121, 2407–2418. [Google Scholar] [PubMed]
  96. Golson, M.L.; le Lay, J.; Gao, N.; Bramswig, N.; Loomes, K.M.; Oakey, R.; May, C.L.; White, P.; Kaestner, K.H. Jagged1 is a competitive inhibitor of Notch signaling in the embryonic pancreas. Mech. Dev. 2009, 126, 687–699. [Google Scholar] [CrossRef] [PubMed]
  97. Parsons, M.J.; Pisharath, H.; Yusuff, S.; Moore, J.C.; Siekmann, A.F.; Lawson, N.; Leach, S.D. Notch-responsive cells initiate the secondary transition in larval zebrafish pancreas. Mech. Dev. 2009, 126, 898–912. [Google Scholar] [CrossRef] [PubMed]
  98. Rovira, M.; Scott, S.G.; Liss, A.S.; Jensen, J.; Thayer, S.P.; Leach, S.D. Isolation and characterization of centroacinar/terminal ductal progenitor cells in adult mouse pancreas. Proc. Natl. Acad. Sci. USA 2010, 107, 75–80. [Google Scholar] [CrossRef] [PubMed]
  99. Su, Y.; Buchler, P.; Gazdhar, A.; Giese, N.; Reber, H.A.; Hines, O.J.; Giese, T.; Buchler, M.W.; Friess, H. Pancreatic regeneration in chronic pancreatitis requires activation of the Notch signaling pathway. J. Gastrointest. Surg. 2006, 10, 1230–1241. [Google Scholar] [CrossRef] [PubMed]
  100. Baeyens, L.; Bonne, S.; Bos, T.; Rooman, I.; Peleman, C.; Lahoutte, T.; German, M.; Heimberg, H.; Bouwens, L. Notch signaling as gatekeeper of rat acinar-to-β-cell conversion in vitro. Gastroenterology 2009, 136, 1750–1760. [Google Scholar] [CrossRef] [PubMed]
  101. Jensen, J.N.; Cameron, E.; Garay, M.V.; Starkey, T.W.; Gianani, R.; Jensen, J. Recapitulation of elements of embryonic development in adult mouse pancreatic regeneration. Gastroenterology 2005, 128, 728–741. [Google Scholar] [CrossRef] [PubMed]
  102. Fujikura, J.; Hosoda, K.; Iwakura, H.; Tomita, T.; Noguchi, M.; Masuzaki, H.; Tanigaki, K.; Yabe, D.; Honjo, T.; Nakao, K. Notch/Rbp-j signaling prevents premature endocrine and ductal cell differentiation in the pancreas. Cell Matab. 2006, 3, 59–65. [Google Scholar] [CrossRef] [PubMed]
  103. Jensen, J.; Pedersen, E.E.; Galante, P.; Hald, J.; Heller, R.S.; Ishibashi, M.; Kageyama, R.; Guillemot, F.; Serup, P.; Madsen, O.D. Control of endodermal endocrine development by Hes-1. Nat. Genet. 2000, 24, 36–44. [Google Scholar] [PubMed]
  104. Sommer, L.; Ma, Q.; Anderson, D.J. Neurogenins, a novel family of atonal-related bHLH transcription factors, are putative mammalian neuronal determination genes that reveal progenitor cell heterogeneity in the developing CNS and PNS. Mol. Cell. Neurosci. 1996, 8, 221–241. [Google Scholar] [CrossRef] [PubMed]
  105. Beatus, P.; Lundkvist, J.; Oberg, C.; Lendahl, U. The Notch 3 intracellular domain represses Notch 1-mediated activation through hairy/enhancer of split (Hes) promoters. Development 1999, 126, 3925–3935. [Google Scholar] [PubMed]
  106. Esni, F.; Ghosh, B.; Biankin, A.V.; Lin, J.W.; Albert, M.A.; Yu, X.; MacDonald, R.J.; Civin, C.I.; Real, F.X.; Pack, M.A.; et al. Notch inhibits Ptf1 function and acinar cell differentiation in developing mouse and zebrafish pancreas. Development 2004, 131, 4213–4224. [Google Scholar] [CrossRef] [PubMed]
  107. Kopinke, D.; Brailsford, M.; Shea, J.E.; Leavitt, R.; Scaife, C.L.; Murtaugh, L.C. Lineage tracing reveals the dynamic contribution of Hes1+ cells to the developing and adult pancreas. Development 2011, 138, 431–441. [Google Scholar] [CrossRef] [PubMed]
  108. Cras-Meneur, C.; Li, L.; Kopan, R.; Permutt, M.A. Presenilins, Notch dose control the fate of pancreatic endocrine progenitors during a narrow developmental window. Genes Dev. 2009, 23, 2088–2101. [Google Scholar] [CrossRef] [PubMed]
  109. Kopinke, D.; Brailsford, M.; Pan, F.C.; Magnuson, M.A.; Wright, C.V.; Murtaugh, L.C. Ongoing Notch signaling maintains phenotypic fidelity in the adult exocrine pancreas. Dev. Biol. 2012, 362, 57–64. [Google Scholar] [CrossRef] [PubMed]
  110. Hart, A.; Papadopoulou, S.; Edlund, H. FGF10 maintains Notch activation, stimulates proliferation, and blocks differentiation of pancreatic epithelial cells. Dev. Dny. 2003, 228, 185–193. [Google Scholar] [CrossRef] [PubMed]
  111. Thompson, N.; Gesina, E.; Scheinert, P.; Bucher, P.; Grapin-Botton, A. RNA profiling and chromatin immunoprecipitation-sequencing reveal that Ptf1a stabilizes pancreas progenitor identity via the control of MNX1/HLXB9 and a network of other transcription factors. Mol. Cell. Biol. 2012, 32, 1189–1199. [Google Scholar] [CrossRef] [PubMed]
  112. Kawaguchi, Y.; Cooper, B.; Gannon, M.; Ray, M.; MacDonald, R.J.; Wright, C.V. The role of the transcriptional regulator Ptf1a in converting intestinal to pancreatic progenitors. Nat. Genet. 2002, 32, 128–134. [Google Scholar] [CrossRef] [PubMed]
  113. Fukuda, A.; Kawaguchi, Y.; Furuyama, K.; Kodama, S.; Horiguchi, M.; Kuhara, T.; Kawaguchi, M.; Terao, M.; Doi, R.; Wright, C.V.; et al. Reduction of Ptf1a gene dosage causes pancreatic hypoplasia and diabetes in mice. Diabetes 2008, 57, 2421–2431. [Google Scholar] [CrossRef] [PubMed]
  114. Norgaard, G.A.; Jensen, J.N.; Jensen, J. FGF10 signaling maintains the pancreatic progenitor cell state revealing a novel role of Notch in organ development. Dev. Biol. 2003, 264, 323–338. [Google Scholar] [CrossRef] [PubMed]
  115. Miralles, F.; Lamotte, L.; Couton, D.; Joshi, R.L. Interplay between FGF10 and Notch signalling is required for the self-renewal of pancreatic progenitors. Int. J. Dev. Biol. 2006, 50, 17–26. [Google Scholar] [CrossRef] [PubMed]
  116. Murata, K.; Hattori, M.; Hirai, N.; Shinozuka, Y.; Hirata, H.; Kageyama, R.; Sakai, T.; Minato, N. Hes1 directly controls cell proliferation through the transcriptional repression of p27kip1. Mol. Cell. Biol. 2005, 25, 4262–4271. [Google Scholar] [CrossRef] [PubMed]
  117. Georgia, S.; Soliz, R.; Li, M.; Zhang, P.; Bhushan, A. P57 and Hes1 coordinate cell cycle exit with self-renewal of pancreatic progenitors. Dev. Biol. 2006, 298, 22–31. [Google Scholar] [CrossRef] [PubMed]
  118. McDonald, E.; Li, J.; Krishnamurthy, M.; Fellows, G.F.; Goodyer, C.G.; Wang, R. Sox9 regulates endocrine cell differentiation during human fetal pancreas development. Int. J. Biochem. Cell Biol. 2012, 44, 72–83. [Google Scholar] [CrossRef] [PubMed]
  119. Siveke, J.T.; Lubeseder-Martellato, C.; Lee, M.; Mazur, P.K.; Nakhai, H.; Radtke, F.; Schmid, R.M. Notch signaling is required for exocrine regeneration after acute pancreatitis. Gastroenterology 2008, 134, 544–555. [Google Scholar] [CrossRef] [PubMed]
  120. De la, O.J.; Emerson, L.L.; Goodman, J.L.; Froebe, S.C.; Illum, B.E.; Curtis, A.B.; Murtaugh, L.C. Notch and Kras reprogram pancreatic acinar cells to ductal intraepithelial neoplasia. Proc. Natl. Acad. Sci. USA 2008, 105, 18907–18912. [Google Scholar] [CrossRef] [PubMed]
  121. Rooman, I.; de Medts, N.; Baeyens, L.; Lardon, J.; de Breuck, S.; Heimberg, H.; Bouwens, L. Expression of the Notch signaling pathway and effect on exocrine cell proliferation in adult rat pancreas. Am. J. Pathol. 2006, 169, 1206–1214. [Google Scholar] [CrossRef] [PubMed]
  122. Mukhi, S.; Brown, D.D. Transdifferentiation of tadpole pancreatic acinar cells to duct cells mediated by Notch and stromelysin-3. Dev. Biol. 2011, 351, 311–317. [Google Scholar] [CrossRef] [PubMed]
  123. Bar, Y.; Russ, H.A.; Knoller, S.; Ouziel-Yahalom, L.; Efrat, S. Hes-1 is involved in adaptation of adult human β-cells to proliferation in vitro. Diabetes 2008, 57, 2413–2420. [Google Scholar] [CrossRef] [PubMed]
  124. Bar, Y.; Russ, H.A.; Sintov, E.; Anker-Kitai, L.; Knoller, S.; Efrat, S. Redifferentiation of expanded human pancreatic β-cell-derived cells by inhibition of the Notch pathway. J. Biol. Chem. 2012, 287, 17269–17280. [Google Scholar] [CrossRef] [PubMed]
  125. Minami, K.; Seino, S. Pancreatic acinar-to-β cell transdifferentiation in vitro. Front. Biosci. 2008, 13, 5824–5837. [Google Scholar] [CrossRef] [PubMed]
  126. Pires-daSilva, A.; Sommer, R.J. The evolution of signalling pathways in animal development. Nat. Rev. Genet. 2003, 4, 39–49. [Google Scholar] [CrossRef] [PubMed]
  127. Baumgartner, B.K.; Cash, G.; Hansen, H.; Ostler, S.; Murtaugh, L.C. Distinct requirements for β-catenin in pancreatic epithelial growth and patterning. Dev. Biol. 2014, 391, 89–98. [Google Scholar] [CrossRef] [PubMed]
  128. Afelik, S.; Pool, B.; Schmerr, M.; Penton, C.; Jensen, J. Wnt7b is required for epithelial progenitor growth and operates during epithelial-to-mesenchymal signaling in pancreatic development. Dev. Biol. 2015, 399, 204–217. [Google Scholar] [CrossRef] [PubMed]
  129. Heiser, P.W.; Lau, J.; Taketo, M.M.; Herrera, P.L.; Hebrok, M. Stabilization of β-catenin impacts pancreas growth. Development 2006, 133, 2023–2032. [Google Scholar] [CrossRef] [PubMed]
  130. Murtaugh, L.C.; Law, A.C.; Dor, Y.; Melton, D.A. β-Catenin is essential for pancreatic acinar but not islet development. Development 2005, 132, 4663–4674. [Google Scholar] [CrossRef] [PubMed]
  131. Rulifson, I.C.; Karnik, S.K.; Heiser, P.W.; ten Berge, D.; Chen, H.; Gu, X.; Taketo, M.M.; Nusse, R.; Hebrok, M.; Kim, S.K. Wnt signaling regulates pancreatic β cell proliferation. Proc. Natl. Acad. Sci. USA 2007, 104, 6247–6252. [Google Scholar] [CrossRef] [PubMed]
  132. Gui, S.; Yuan, G.; Wang, L.; Zhou, L.; Xue, Y.; Yu, Y.; Zhang, J.; Zhang, M.; Yang, Y.; Wang, D.W. Wnt3a regulates proliferation, apoptosis and function of pancreatic NIT-1 β cells via activation of IRS2/PI3K signaling. J. Cell. Biochem. 2013, 114, 1488–1497. [Google Scholar] [CrossRef] [PubMed]
  133. Fujino, T.; Asaba, H.; Kang, M.J.; Ikeda, Y.; Sone, H.; Takada, S.; Kim, D.H.; Ioka, R.X.; Ono, M.; Tomoyori, H.; et al. Low-density lipoprotein receptor-related protein 5 (LRP5) is essential for normal cholesterol metabolism and glucose-induced insulin secretion. Proc. Natl. Acad. Sci. USA 2003, 100, 229–234. [Google Scholar] [CrossRef] [PubMed]
  134. Jin, T. The wnt signalling pathway and diabetes mellitus. Diabetologia 2008, 51, 1771–1780. [Google Scholar] [CrossRef] [PubMed]
  135. Nusse, R.; Fuerer, C.; Ching, W.; Harnish, K.; Logan, C.; Zeng, A.; ten Berge, D.; Kalani, Y. Wnt signaling and stem cell control. Cold Spring Harb. Symp. Quant. Biol. 2008, 73, 59–66. [Google Scholar] [CrossRef] [PubMed]
  136. Zeng, Y.A.; Nusse, R. Wnt proteins are self-renewal factors for mammary stem cells and promote their long-term expansion in culture. Cell Stem Cell 2010, 6, 568–577. [Google Scholar] [CrossRef] [PubMed]
  137. Tian, H.; Biehs, B.; Chiu, C.; Siebel, C.W.; Wu, Y.; Costa, M.; de Sauvage, F.J.; Klein, O.D. Opposing activities of Notch and wnt signaling regulate intestinal stem cells and gut homeostasis. Cell Rep. 2015, 11, 33–42. [Google Scholar] [CrossRef] [PubMed]
  138. Axelrod, J.D.; Matsuno, K.; Artavanis-Tsakonas, S.; Perrimon, N. Interaction between wingless and Notch signaling pathways mediated by dishevelled. Science 1996, 271, 1826–1832. [Google Scholar] [CrossRef] [PubMed]
  139. Collu, G.M.; Hidalgo-Sastre, A.; Acar, A.; Bayston, L.; Gildea, C.; Leverentz, M.K.; Mills, C.G.; Owens, T.W.; Meurette, O.; Dorey, K.; et al. Dishevelled limits Notch signalling through inhibition of CSL. Development 2012, 139, 4405–4415. [Google Scholar] [CrossRef] [PubMed]
  140. Strutt, D.; Johnson, R.; Cooper, K.; Bray, S. Asymmetric localization of frizzled and the determination of Notch-dependent cell fate in the drosophila eye. Curr. Biol. 2002, 12, 813–824. [Google Scholar] [CrossRef]
  141. Capilla, A.; Johnson, R.; Daniels, M.; Benavente, M.; Bray, S.J.; Galindo, M.I. Planar cell polarity controls directional Notch signaling in the drosophila leg. Development 2012, 139, 2584–2593. [Google Scholar] [CrossRef] [PubMed]
  142. Foltz, D.R.; Santiago, M.C.; Berechid, B.E.; Nye, J.S. Glycogen synthase kinase-3β modulates Notch signaling and stability. Curr. Biol. 2002, 12, 1006–1011. [Google Scholar] [CrossRef]
  143. Hayward, P.; Brennan, K.; Sanders, P.; Balayo, T.; DasGupta, R.; Perrimon, N.; Martinez Arias, A. Notch modulates Wnt signalling by associating with Armadillo/β-catenin and regulating its transcriptional activity. Development 2005, 132, 1819–1830. [Google Scholar] [CrossRef] [PubMed]
  144. Yamamizu, K.; Matsunaga, T.; Uosaki, H.; Fukushima, H.; Katayama, S.; Hiraoka-Kanie, M.; Mitani, K.; Yamashita, J.K. Convergence of Notch and β-catenin signaling induces arterial fate in vascular progenitors. J. Cell Biol. 2010, 189, 325–338. [Google Scholar] [CrossRef] [PubMed]
  145. Gu, B.; Watanabe, K.; Sun, P.; Fallahi, M.; Dai, X. Chromatin effector Pygo2 mediates wnt-Notch crosstalk to suppress luminal/alveolar potential of mammary stem and basal cells. Cell Stem Cell 2013, 13, 48–61. [Google Scholar] [CrossRef] [PubMed]
  146. Golosow, N.; Grobstein, C. Epitheliomesenchymal interaction in pancreatic morphogenesis. Dev. Biol. 1962, 4, 242–255. [Google Scholar] [CrossRef]
  147. Shih, H.P.; Sander, M. Pancreas development ex vivo: Culturing embryonic pancreas explants on permeable culture inserts, with fibronectin-coated glass microwells, or embedded in three-dimensional matrigel. Methods Mol. Biol. 2014, 1210, 229–237. [Google Scholar] [PubMed]
  148. Bhushan, A.; Itoh, N.; Kato, S.; Thiery, J.P.; Czernichow, P.; Bellusci, S.; Scharfmann, R. FGF10 is essential for maintaining the proliferative capacity of epithelial progenitor cells during early pancreatic organogenesis. Development 2001, 128, 5109–5117. [Google Scholar] [PubMed]
  149. Ye, F.; Duvillie, B.; Scharfmann, R. Fibroblast growth factors 7 and 10 are expressed in the human embryonic pancreatic mesenchyme and promote the proliferation of embryonic pancreatic epithelial cells. Diabetologia 2005, 48, 277–281. [Google Scholar] [CrossRef] [PubMed]
  150. Li, S.; Francisco, A.B.; Munroe, R.J.; Schimenti, J.C.; Long, Q. SEL1L deficiency impairs growth and differentiation of pancreatic epithelial cells. BMC Dev. Biol. 2010, 10, 19. [Google Scholar] [CrossRef] [PubMed]
  151. Ikeya, T.; Hayashi, S. Interplay of Notch and FGF signaling restricts cell fate and mapk activation in the drosophila trachea. Development 1999, 126, 4455–4463. [Google Scholar] [PubMed]
  152. Mustonen, T.; Tummers, M.; Mikami, T.; Itoh, N.; Zhang, N.; Gridley, T.; Thesleff, I. Lunatic fringe, FGF, and BMP regulate the Notch pathway during epithelial morphogenesis of teeth. Dev. Biol. 2002, 248, 281–293. [Google Scholar] [CrossRef] [PubMed]
  153. Gittes, G.K.; Galante, P.E.; Hanahan, D.; Rutter, W.J.; Debase, H.T. Lineage-specific morphogenesis in the developing pancreas: Role of mesenchymal factors. Development 1996, 122, 439–447. [Google Scholar] [PubMed]
  154. Blauer, M.; Sand, J.; Laukkarinen, J. Cryopreserved mouse pancreatic acinar cells from long-term explant outgrowth cultures maintain their secretory phenotype after thawing. Pancreatology 2013, 13, 610–614. [Google Scholar] [CrossRef] [PubMed]
  155. Dichmann, D.S.; Miller, C.P.; Jensen, J.; Scott Heller, R.; Serup, P. Expression and misexpression of members of the FGF and TGFβ families of growth factors in the developing mouse pancreas. Dev. Dyn. 2003, 226, 663–674. [Google Scholar] [CrossRef] [PubMed]
  156. Kobberup, S.; Schmerr, M.; Dang, M.L.; Nyeng, P.; Jensen, J.N.; MacDonald, R.J.; Jensen, J. Conditional control of the differentiation competence of pancreatic endocrine and ductal cells by FGF10. Mech. Dev. 2010, 127, 220–234. [Google Scholar] [CrossRef] [PubMed]
  157. Duvillie, B.; Heinis, M.; Stetsyuk, V. In vivo and in vitro techniques to study pancreas development and islet cell function. Endocr. Dev. 2007, 12, 46–54. [Google Scholar] [PubMed]
  158. Hart, A.W.; Baeza, N.; Apelqvist, A.; Edlund, H. Attenuation of FGF signalling in mouse β-cells leads to diabetes. Nature 2000, 408, 864–868. [Google Scholar] [PubMed]
  159. Duvillie, B.; Attali, M.; Bounacer, A.; Ravassard, P.; Basmaciogullari, A.; Scharfmann, R. The mesenchyme controls the timing of pancreatic β-cell differentiation. Diabetes 2006, 55, 582–589. [Google Scholar] [CrossRef] [PubMed]
  160. Chitnis, A.; Henrique, D.; Lewis, J.; Ish-Horowicz, D.; Kintner, C. Primary neurogenesis in xenopus embryos regulated by a homologue of the drosophila neurogenic gene δ. Nature 1995, 375, 761–766. [Google Scholar] [CrossRef] [PubMed]
  161. Sakamoto, M.; Hirata, H.; Ohtsuka, T.; Bessho, Y.; Kageyama, R. The basic helix-loop-helix genes Hesr1/Hey1 and Hesr2/Hey2 regulate maintenance of neural precursor cells in the brain. J. Biol. Chem. 2003, 278, 44808–44815. [Google Scholar] [CrossRef] [PubMed]
  162. Grandbarbe, L.; Bouissac, J.; Rand, M.; Hrabe de Angelis, M.; Artavanis-Tsakonas, S.; Mohier, E. δ-Notch signaling controls the generation of neurons/glia from neural stem cells in a stepwise process. Development 2003, 130, 1391–1402. [Google Scholar] [CrossRef] [PubMed]
  163. Schuster-Gossler, K.; Cordes, R.; Gossler, A. Premature myogenic differentiation and depletion of progenitor cells cause severe muscle hypotrophy in δ1 mutants. Proc. Natl. Acad. Sci. USA 2007, 104, 537–542. [Google Scholar] [CrossRef] [PubMed]
  164. Brohl, D.; Vasyutina, E.; Czajkowski, M.T.; Griger, J.; Rassek, C.; Rahn, H.P.; Purfurst, B.; Wende, H.; Birchmeier, C. Colonization of the satellite cell niche by skeletal muscle progenitor cells depends on Notch signals. Dev. Cell 2012, 23, 469–481. [Google Scholar] [CrossRef] [PubMed]
  165. Skeath, J.B. At the nexus between pattern formation and cell-type specification: The generation of individual neuroblast fates in the drosophila embryonic central nervous system. Bioessays 1999, 21, 922–931. [Google Scholar] [CrossRef]
  166. Sanalkumar, R.; Dhanesh, S.B.; James, J. Non-canonical activation of Notch signaling/target genes in vertebrates. Cell. Mol. Life Sci. 2010, 67, 2957–2968. [Google Scholar] [CrossRef] [PubMed]
  167. Gazave, E.; Lapebie, P.; Richards, G.S.; Brunet, F.; Ereskovsky, A.V.; Degnan, B.M.; Borchiellini, C.; Vervoort, M.; Renard, E. Origin and evolution of the Notch signalling pathway: An overview from eukaryotic genomes. BMC Evol. Biol. 2009, 9, 249. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Li, X.-Y.; Zhai, W.-J.; Teng, C.-B. Notch Signaling in Pancreatic Development. Int. J. Mol. Sci. 2016, 17, 48. https://doi.org/10.3390/ijms17010048

AMA Style

Li X-Y, Zhai W-J, Teng C-B. Notch Signaling in Pancreatic Development. International Journal of Molecular Sciences. 2016; 17(1):48. https://doi.org/10.3390/ijms17010048

Chicago/Turabian Style

Li, Xu-Yan, Wen-Jun Zhai, and Chun-Bo Teng. 2016. "Notch Signaling in Pancreatic Development" International Journal of Molecular Sciences 17, no. 1: 48. https://doi.org/10.3390/ijms17010048

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop