Heterozygous mutations in the human HNF1B gene are associated with maturity-onset diabetes of the young type 5 (MODY5) and pancreas hypoplasia. In mouse, Hnf1b heterozygous mutants do not exhibit any phenotype, whereas the homozygous deletion in the entire epiblast leads to pancreas agenesis associated with abnormal gut regionalization. Here, we examine the specific role of Hnf1b during pancreas development, using constitutive and inducible conditional inactivation approaches at key developmental stages. Hnf1b early deletion leads to a reduced pool of pancreatic multipotent progenitor cells (MPCs) due to decreased proliferation and increased apoptosis. Lack of Hnf1b either during the first or the secondary transitions is associated with cystic ducts. Ductal cells exhibit aberrant polarity and decreased expression of several cystic disease genes, some of which we identified as novel Hnf1b targets. Notably, we show that Glis3, a transcription factor involved in duct morphogenesis and endocrine cell development, is downstream Hnf1b. In addition, a loss and abnormal differentiation of acinar cells are observed. Strikingly, inactivation of Hnf1b at different time points results in the absence of Ngn3+ endocrine precursors throughout embryogenesis. We further show that Hnf1b occupies novel Ngn3 putative regulatory sequences in vivo. Thus, Hnf1b plays a crucial role in the regulatory networks that control pancreatic MPC expansion, acinar cell identity, duct morphogenesis and generation of endocrine precursors. Our results uncover an unappreciated requirement of Hnf1b in endocrine cell specification and suggest a mechanistic explanation of diabetes onset in individuals with MODY5.

The pancreas is an abdominal gland that is essential for nutrient homeostasis formed by acinar, ductal and endocrine cells. Acinar cells, organized in acini, secrete digestive enzymes into the duodenum via the ductal network. Endocrine cells are organized in the islets of Langerhans, notably composed of α, β and δ cells, which secrete the hormones glucagon, insulin and somatostatin, respectively. In mice, the pancreas is specified by embryonic day (E) 8.5. Early pancreas morphogenesis, known as the primary transition, is characterized by active proliferation of epithelial multipotent progenitor cells (MPCs), followed by a period of growth and differentiation from E12.5, called the secondary transition, to form acinar, ductal and endocrine cells. Regionalization of the early epithelium results in a pre-acinar domain in the tips of the branching organ and a central bipotential ductal/endocrine trunk domain, from which endocrine cells differentiate (Seymour and Sander, 2011). Notch signaling plays an important role in this process (Afelik and Jensen, 2013). Numerous transcription factors have been implicated in the regulation of pancreas development (Pan and Wright, 2011). Foxa1/a2 are dominant regulators of Pdx1 expression (Gao et al., 2008). Pdx1 and Ptf1a determine fate specification of pancreatic MPCs (Pan and Wright, 2011), whereas Sox9 maintains MPCs, by preventing apoptosis and promoting proliferation (Seymour et al., 2007). Ngn3 is required for endocrine cell differentiation (Gradwohl et al., 2000). During the secondary transition, Sox9 and Hnf6 are both expressed in the bipotent duct/endocrine domain and required for maintaining Ngn3 expression (Dubois et al., 2011; Jacquemin et al., 2000; Seymour et al., 2008). After the secondary transition, endocrine and exocrine cell populations expand and differentiate to generate the mature hormone- and enzyme-producing cell types of islets and acini, respectively.

Among transcription factors, hepatocyte nuclear factor 1b (Hnf1b) is expressed in the pre-pancreatic foregut endoderm and in pancreatic MPCs. A sequential transcriptional cascade of Hnf1b→Hnf6→Pdx1 was found to direct differentiation of endodermal cells into pancreatic progenitors (Poll et al., 2006). From ∼E14.5 to adulthood, Hnf1b expression is restricted to the embryonic ductal cords that later form the adult ductal cells (Haumaitre et al., 2005; Kopp et al., 2011; Maestro et al., 2003; Nammo et al., 2008). We have previously shown that Hnf1b-deficient mouse embryos rescued by tetraploid aggregation exhibit pancreas agenesis, with a transient dorsal bud expressing Pdx1 but not Ptf1a, showing that Hnf1b is required for pancreas specification (Haumaitre et al., 2005). Regionalization of the gut was also affected, as revealed by ectopic expression of Shh, which could contribute to reduce the Pdx1+ pre-pancreatic domain. Hnf1b-deficient embryos also exhibit impaired specification of other organs derived from the ventral endoderm, including liver (Lokmane et al., 2008). Thus, it was difficult to distinguish the role of Hnf1b in the pancreas from its role in regionalizing the primitive intestine. Moreover, the severity of the phenotype precluded the analysis of crucial later roles of Hnf1b during pancreas differentiation. Indeed, lineage-tracing analyses revealed that embryonic Hnf1b+ cells of the branching pancreas are precursors of acinar, duct and endocrine lineages (Solar et al., 2009). In humans, HNF1B heterozygous mutations are associated with ‘maturity onset diabetes of the young type 5’ (MODY5) syndrome, which is characterized by early onset of diabetes, pancreas hypoplasia and multicystic kidney dysplasia (Bellanné-Chantelot et al., 2004; Chen et al., 2010; Edghill et al., 2006; Haldorsen et al., 2008). The identification of two fetuses carrying distinct HNF1B mutations, associated with polycystic kidneys and severe pancreatic hypoplasia (Haumaitre et al., 2006), further suggested an early developmental role of HNF1B in human pancreas, which might be an important cause of MODY5.

In order to elucidate the specific role of Hnf1b in pancreas development, we conditionally inactivated Hnf1b in pancreatic MPCs and at later stages. Combined early and late deletion analyses demonstrate the crucial function of Hnf1b in the regulatory networks controlling pancreatic MPC expansion, duct morphogenesis, acinar cell identity and generation of endocrine precursors.

Hnf1b deficiency in pancreatic progenitors leads to severe pancreatic hypoplasia and perinatal lethality

We performed a conditional deletion of Hnf1b in pancreatic MPCs using a Hnf1b-floxed mouse line (Heliot et al., 2013) crossed with the Pdx1-Cre (Wells et al., 2007) or the tamoxifen (TM)-inducible Sox9-CreERT2 line (Kopp et al., 2011), as Pdx1 and Sox9 share a common expression domain with Hnf1b in the early pancreas (Dubois et al., 2011; Maestro et al., 2003; Seymour et al., 2007). Pdx1-Cre;Hnf1b+/LacZ and Hnf1bFlox/Flox mice were crossed to generate Pdx1-Cre;Hnf1bFlox/LacZ embryos, referred to as mutants. Hnf1bFlox/LacZ and Hnf1b+/Flox embryos without the Pdx1-Cre transgene were referred to as controls, as haploinsufficient embryos with the LacZ-null Hnf1b allele did not show any phenotype (Barbacci et al., 1999; Kornfeld et al., 2013). Heterozygous Pdx1-Cre;Hnf1b+/Flox embryos also showed no phenotype (Fig. 1E; supplementary material Fig. S1). By contrast, Pdx1-Cre;Hnf1bFlox/LacZ mutant embryos displayed severe pancreatic hypoplasia at E18.5 (Fig. 1A-D), corresponding to a 45% and 90% decrease in pancreatic weight at E16.5 and E18.5, respectively (Fig. 1E). We also generated Pdx1-Cre;Hnf1bFlox/LacZ;R26R+/YFP mutants and observed uniform YFP labeling in the remnant pancreatic epithelium, revealing the high efficiency of the Pdx1-driven Cre recombination (Fig. 1C′,D′). Hnf1b/GFP co-immunostainings at E10.5 confirmed the extensive deletion of Hnf1b in mutants, showing only 16% of remaining Hnf1b+/GFP cells due to a slight mosaic expression of the Pdx1-Cre line (Fig. 1F-G′). In accordance, we found a 70% decrease in wild-type (WT) Hnf1b transcripts at E12.5 (Fig. 1H). Histological analysis by Hematoxylin and Eosin staining revealed a severe decrease in acinar cells with dispersed clusters of acini, cystic ducts and an apparent absence of endocrine islets in mutant pancreata at E16.5 and E18.5 (Fig. 1I-L). This phenotype was associated with high lethality of mutant pups, as 70% died during the first week of life (Fig. 1M). Interestingly, we found that mutant newborns were hypoglycemic, with a 30% decrease of blood glucose (Fig. 1O). This phenotype correlates with a 93% decrease in glucagon-expressing cells (see Fig. 7O). Hypoglycemia is likely the main cause of mutant lethality at P0/P1 (40%), because immediately after birth, glycogenolysis stimulated by glucagon allows mobilisation of hepatic glycogen, which is the only energetic source available at this stage. Hnf3a−/− mice also die perinatally of hypoglycemia, associated with a 50% decrease in glucagon (Kaestner et al., 1999; Shih et al., 1999). By contrast, at P2, mutants became hyperglycemic, with a 44% increase of blood glucose (Fig. 1O), in correlation with the 93% decrease in insulin-expressing cells (see Fig. 7O). Furthermore, we found a massive increase of blood amylase in mutants (250%) (Fig. 1P), as is the case in pancreatitis. This was associated with acinar cell defects (see Fig. 4) and a 32% decrease in mutant body weight at P2 (Fig. 1N). This suggests that mutant lethality after P2 may be due to hyperglycemia and to chronic malabsorption.

Fig. 1.

Hnf1b inactivation in pancreatic MPCs leads to a strong pancreas hypoplasia. (A,B) Digestive tracts at E18.5. p, pancreas; st, stomach; sp, spleen; d, duodenum; li, liver. (C,D) E18.5 dissected pancreata. (C′,D′) Recombination shown by the YFP+ signal (green) in the Pdx1-Cre;Hnf1bFlox/LacZ;R26R+/YFP mutant pancreas. (E) Pancreas weight at E16.5 and E18.5 in controls, heterozygous (Pdx1-Cre;Hnf1bFlox/+) and mutants (Pdx1-Cre;Hnf1bFlox/LacZ) (E16.5: control n=9, heterozygous n=7, mutant n=9; E18.5: control n=9, heterozygous n=6, mutant n=5). (F,G) Hnf1b inactivation efficiency in Pdx1-Cre;Hnf1bFlox/LacZ;R26R+/YFP mutant pancreata at E10.5. Hnf1b (red) and GFP (green) co-immunostaining. (F′,G′) Same section showing GFP staining (green) and nuclei stained with DAPI (blue). Only a few Hnf1b+ cells are observed in the mutants, which are GFP (arrows in G,G′). (H) qRT-PCR of wild-type Hnf1b transcripts from E12.5 control and Pdx1-Cre;Hnf1bFlox/LacZ mutant pancreata (control, n=6; mutant, n=4; n being a pool of three pancreata). (I-L) Hematoxylin/Eosin staining of pancreata at E16.5 and E18.5. Asterisks indicate cystic ducts and arrows indicate enlarged acinar lumen in mutants. (M) Lethality of Hnf1b mutant mice. (No lethality was observed for control mice.) (N,O) Body weight and glycemia of P0/P1 (control n=16, mutant n=8) and P2 pups (control n=5, mutant n=3). (P) Blood amylase in pups (P0-P2: control n=15, mutant n=5). *P<0.05; ***P<0.001. Scale bars: 200 µm in A-D; 50 µm in F,G and I-L.

Fig. 1.

Hnf1b inactivation in pancreatic MPCs leads to a strong pancreas hypoplasia. (A,B) Digestive tracts at E18.5. p, pancreas; st, stomach; sp, spleen; d, duodenum; li, liver. (C,D) E18.5 dissected pancreata. (C′,D′) Recombination shown by the YFP+ signal (green) in the Pdx1-Cre;Hnf1bFlox/LacZ;R26R+/YFP mutant pancreas. (E) Pancreas weight at E16.5 and E18.5 in controls, heterozygous (Pdx1-Cre;Hnf1bFlox/+) and mutants (Pdx1-Cre;Hnf1bFlox/LacZ) (E16.5: control n=9, heterozygous n=7, mutant n=9; E18.5: control n=9, heterozygous n=6, mutant n=5). (F,G) Hnf1b inactivation efficiency in Pdx1-Cre;Hnf1bFlox/LacZ;R26R+/YFP mutant pancreata at E10.5. Hnf1b (red) and GFP (green) co-immunostaining. (F′,G′) Same section showing GFP staining (green) and nuclei stained with DAPI (blue). Only a few Hnf1b+ cells are observed in the mutants, which are GFP (arrows in G,G′). (H) qRT-PCR of wild-type Hnf1b transcripts from E12.5 control and Pdx1-Cre;Hnf1bFlox/LacZ mutant pancreata (control, n=6; mutant, n=4; n being a pool of three pancreata). (I-L) Hematoxylin/Eosin staining of pancreata at E16.5 and E18.5. Asterisks indicate cystic ducts and arrows indicate enlarged acinar lumen in mutants. (M) Lethality of Hnf1b mutant mice. (No lethality was observed for control mice.) (N,O) Body weight and glycemia of P0/P1 (control n=16, mutant n=8) and P2 pups (control n=5, mutant n=3). (P) Blood amylase in pups (P0-P2: control n=15, mutant n=5). *P<0.05; ***P<0.001. Scale bars: 200 µm in A-D; 50 µm in F,G and I-L.

We also conditionally inactivated Hnf1b with the Sox9-CreERT2 line. Pregnant females from crosses between Hnf1bFlox/Flox and Sox9-CreERT2;Hnf1b+/LacZ mice were injected with TM at E9.5 (Kopp et al., 2011). Sox9-CreERT2;Hnf1bFlox/LacZ mutants exhibited strong pancreatic hypoplasia at E18.5 (supplementary material Fig. S2A,B), corresponding to a 40% decrease in pancreas weight at E16.5 (supplementary material Fig. S2G). Hematoxylin and Eosin staining of mutant pancreata showed cystic ducts with a severe decrease in acinar cells and absence of islet structures (supplementary material Fig. S2C-F). Efficient Sox9-driven Cre recombination was observed with a 70% decrease in Hnf1b transcripts at E14.5 (supplementary material Fig. S2H), and further confirmed by Hnf1b and GFP co-immunostainings at E11.5 in Sox9-CreERT2;Hnf1bFlox/LacZ;R26R+/YFP mutants (supplementary material Fig. S2I-J′). These data strongly corroborate the findings obtained with the Pdx1-Cre line. Therefore, Hnf1b inactivation in MPCs leads to severe pancreatic hypoplasia, associated with cystic ducts, a decrease in acinar cells and absence of endocrine cells.

Hnf1b is required for proliferation and survival of MPCs

To investigate the underlying cause of pancreatic hypoplasia in Pdx1-Cre;Hnf1bFlox/LacZ mutants, we analyzed the pool of MPCs at E12.5 using Pdx1 immunostaining and observed a 35% decrease in Pdx1+ progenitor cells (Fig. 2A). We further analyzed proliferation and apoptosis. We quantified the percentage of mitotic and apoptotic Pdx1+ cells using phospho-histone H3 (PHH3) and TUNEL assay, respectively. A 20% decrease in Pdx1+ cell proliferation (Fig. 2B,D,E), as well as an 11-fold increase in Pdx1+ cell apoptosis (Fig. 2C,F,G) were observed in mutants compared with controls. Thus, both decreased proliferation and increased cell death contribute to the reduction of MPCs in mutants.

Fig. 2.

Hnf1b is required for proliferation and survival of pancreatic MPCs. (A) Percentage of Pdx1+ progenitors in control and Pdx1-Cre;Hnf1bFlox/LacZ pancreata at E12.5. (B,C) Proliferation and apoptosis of Pdx1+ progenitors at E12.5. (D,E) Phospho-histone H3 (PHH3) (red) and Pdx1 (green) immunostaining. (F,G) TUNEL assay (green). The epithelium is encircled in red. Scale bars: 50 µm. (H) qRT-PCR of Fgfr2b and Fgfr4 on E12.5 pancreata. (I) ChIP showing Hnf1b fold enrichment in regulatory regions of Fgfr4 from E12.5 pancreata immunoprecipitated with an Hnf1b antibody versus control IgG. Hnf1-binding sites are shown in red with their positions relative to Fgfr4 TSS. *P<0.05; **P<0.005; ***P<0.001.

Fig. 2.

Hnf1b is required for proliferation and survival of pancreatic MPCs. (A) Percentage of Pdx1+ progenitors in control and Pdx1-Cre;Hnf1bFlox/LacZ pancreata at E12.5. (B,C) Proliferation and apoptosis of Pdx1+ progenitors at E12.5. (D,E) Phospho-histone H3 (PHH3) (red) and Pdx1 (green) immunostaining. (F,G) TUNEL assay (green). The epithelium is encircled in red. Scale bars: 50 µm. (H) qRT-PCR of Fgfr2b and Fgfr4 on E12.5 pancreata. (I) ChIP showing Hnf1b fold enrichment in regulatory regions of Fgfr4 from E12.5 pancreata immunoprecipitated with an Hnf1b antibody versus control IgG. Hnf1-binding sites are shown in red with their positions relative to Fgfr4 TSS. *P<0.05; **P<0.005; ***P<0.001.

Activation of the fibroblast growth factor (FGF) pathway via binding of mesenchymal FGFs to epithelial FGF receptors (FGFRs) is fundamental to promote proliferation of early pancreatic MPCs, especially through the Fgf10/Fgfr2b pathway (Bhushan et al., 2001; Hart et al., 2003; Pulkkinen et al., 2003). Surprisingly, we observed no difference in Fgfr2b expression in Pdx1-Cre;Hnf1bFlox/LacZ mutants at E12.5, but we found a 70% decrease in Fgfr4 expression by qRT-PCR (Fig. 2H). Moreover, by in vivo chromatin immunoprecipitation (ChIP) on E12.5 pancreata (Fig. 2I), we found Hnf1b bound to a region containing two previously described Hnf1 DNA-binding sites (Shah et al., 2002), at +280 and +355 bp downstream of the Fgfr4 transcription start site (TSS) in its first intron. These data suggest that modulation of FGF signaling through direct regulation of Fgfr4 by Hnf1b may sustain MPC expansion.

The Notch signaling pathway also plays an important role in the maintenance, proliferation and differentiation of pancreatic MPCs (Apelqvist et al., 1999; Hald et al., 2003; Jensen et al., 2000; Murtaugh et al., 2003). In Pdx1-Cre;Hnf1bFlox/LacZ pancreata, we observed a downregulation of the Notch ligand Dll1 (40%), and an upregulation of the effectors Hey1, Hey2 and Heyl at E12.5 (Fig. 3A). Interestingly, we found that expression of Hey and other Notch members, such as Notch2 and Jag1, remained abnormally high in mutant pancreata at E14.5 contrary to controls, in which these genes are downregulated at this stage (Fig. 3B). Thus, lack of Hnf1b is associated with deregulation of some Notch pathway components. These findings show that Hnf1b is essential for proliferation and maintenance of MPCs, at least in part through modulation of FGF and Notch pathways.

Fig. 3.

Lack of Hnf1b is associated with a deregulated Notch pathway. (A,B) qRT-PCR analysis of Notch pathway genes in control and Pdx1-Cre;Hnf1bFlox/LacZ pancreata at E12.5 (A) and at E14.5 (B) (control, n=7; mutant, n=8). Dll1 is downregulated and Hey factors are upregulated at E12.5; high expression of Notch2, Jag1, Rbpj, Hey1 and Hey2 is maintained in mutants at E14.5. *P<0.05; **P<0.005; ***P<0.001.

Fig. 3.

Lack of Hnf1b is associated with a deregulated Notch pathway. (A,B) qRT-PCR analysis of Notch pathway genes in control and Pdx1-Cre;Hnf1bFlox/LacZ pancreata at E12.5 (A) and at E14.5 (B) (control, n=7; mutant, n=8). Dll1 is downregulated and Hey factors are upregulated at E12.5; high expression of Notch2, Jag1, Rbpj, Hey1 and Hey2 is maintained in mutants at E14.5. *P<0.05; **P<0.005; ***P<0.001.

Lack of Hnf1b leads to acinar cell differentiation defects

The severe loss of acinar cells and the impaired architecture of remaining acini with enlarged acinar lumens observed by Hematoxylin and Eosin staining in mutants (Fig 1I-L) led us to investigate acinar defects. Amylase immunostaining (Fig. 4A-H) and amylase+ cross-sectional area quantification at E16.5 showed a 20% decrease in acinar area in Pdx1-Cre;Hnf1bFlox/LacZ mutants (Fig. 4J). A 67% decrease in Amylase expression was observed at E16.5 by qRT-PCR (Fig. 4I), suggesting that Amylase expression might be reduced in the remaining mutant acinar cells. We also found an eightfold increase of apoptotic mutant acinar cells (Fig. 4L), which cannot be fully compensated for by a twofold increase of mitotic acinar cells (Fig. 4K). In addition, we analyzed expression of the early acinar markers Ptf1a, Mist1 (Bhlha15 – Mouse Genome Informatics) and Nr5a2 (Hale et al., 2014; Pin et al., 2001). Expression of Mist1 was severely reduced before and during the onset of acinar differentiation, with a 50% and an 80% decrease at E12.5 and E14.5, respectively (Fig. 4I). We also observed a 64% decrease of Ptf1a at E14.5 and a 35% decrease of Nr5a2 at E16.5 (Fig. 4I). Notably, an equivalent severe decrease in acinar cells was found in Sox9-CreERT2;Hnf1bFlox/LacZ (TM E9.5), with a 74% downregulation of amylase gene expression at E16.5 (supplementary material Fig. S3E).

Fig. 4.

Lack of Hnf1b leads to acinar cell differentiation defects. (A-D) Amylase (green) and Hnf6 (red) immunostaining in control and Pdx1-Cre;Hnf1bFlox/LacZ pancreata at E16.5. (A,B) Hnf6 expression is found in control ducts but not in in acinar amylase+ cells. (C,D) Absence of Hnf6 expression in mutant ducts and ectopic expression of Hnf6 in acinar amylase+ cells (arrow in D). Nuclei are stained with DAPI (blue). (E-H) Amylase (green) and pan-cytokeratin (PanCK, red) immunostaining at E16.5. (G,H) Increased cytokeratin expression in intercalated mutant ducts and centro-acinar cells. (I) qRT-PCR of Mist1, Ptf1a, Nr5a2, amylase and Ck19 at E12.5, E14.5 and E16.5 (control, n=11; mutant, n=11) (a.u., arbitrary unit). (J) Quantification of ductal Sox9+ and acinar amylase+ sectional areas. (K) Quantification of Sox9+ and amylase+ cells in proliferation by PHH3 immunostaining at E16.5. (L) Quantification of amylase+ acinar cells in apoptosis by TUNEL assay at E16.5. Scale bars: 50 µm in A,C,E,G; 25 µm in B,D,F,H. *P<0.05; ***P<0.001.

Fig. 4.

Lack of Hnf1b leads to acinar cell differentiation defects. (A-D) Amylase (green) and Hnf6 (red) immunostaining in control and Pdx1-Cre;Hnf1bFlox/LacZ pancreata at E16.5. (A,B) Hnf6 expression is found in control ducts but not in in acinar amylase+ cells. (C,D) Absence of Hnf6 expression in mutant ducts and ectopic expression of Hnf6 in acinar amylase+ cells (arrow in D). Nuclei are stained with DAPI (blue). (E-H) Amylase (green) and pan-cytokeratin (PanCK, red) immunostaining at E16.5. (G,H) Increased cytokeratin expression in intercalated mutant ducts and centro-acinar cells. (I) qRT-PCR of Mist1, Ptf1a, Nr5a2, amylase and Ck19 at E12.5, E14.5 and E16.5 (control, n=11; mutant, n=11) (a.u., arbitrary unit). (J) Quantification of ductal Sox9+ and acinar amylase+ sectional areas. (K) Quantification of Sox9+ and amylase+ cells in proliferation by PHH3 immunostaining at E16.5. (L) Quantification of amylase+ acinar cells in apoptosis by TUNEL assay at E16.5. Scale bars: 50 µm in A,C,E,G; 25 µm in B,D,F,H. *P<0.05; ***P<0.001.

Interestingly, the ductal marker Hnf6 was almost undetectable in Pdx1-Cre;Hnf1bFlox/LacZ pancreatic ducts, but was found to be ectopically expressed in some differentiated acinar cells at E16.5 (Fig. 4A-D). Ectopic expression of another ductal marker, Sox9, was also observed in mutant acini (Fig. 5C,D). This was accompanied by expanded expression of the ductal marker cytokeratin (pan-CK) (Fig. 4E-H) and by a twofold upregulation of Ck19 observed by qRT-PCR (Fig. 4I). In agreement with these observations, the Sox9+ cross-sectional area increased threefold in mutant pancreata (Fig. 4J), even though the proliferation rate in this compartment was not significantly changed (Fig. 4K).

Fig. 5.

Hnf1b is crucial for duct morphogenesis. (A-F) Sox9 immunohistochemistry (brown) in control and Pdx1-Cre;Hnf1bFlox/LacZ pancreata at E14.5, E16.5 and E18.5. In mutants, note the epithelium multistratified regions at E14.5 (B; arrow), the cystic ducts from E16.5 (D,F; asterisks) with multilayered epithelia (D; arrow), a group of acinar cells ectopically expressing Sox9 (D; encircled in black) and the loss of ductal marker expression at E18.5 (F; arrows). Immunostaining at E16.5 for dystroglycan (green) and β-catenin (β-Cat, red) (G,H); laminin (green) (I,J); AQP1 (green) (K,L); ezrin (green) and PanCK (red) (M,N); PKCz (green) and β-Cat (red) (O,P); mucin 1 (MUC1, green) and β-Cat (red) (Q,R); and MUC1 (green) and acetylated α-Tubulin (Ac-Tub, red) (S,T). There is a strong decrease in dystroglycan and laminin basal marker staining in mutant acinar cells (H′,J′), and increased β-catenin staining in the apical region of acinar cells (H′) and in cystic ducts (R′). Ezrin, PKCz and MUC1 staining shows the loss of polarity of cystic ducts with absence or disruption of the apical staining (N′,P′,R′). Mutant ductal epithelial cells stained with MUC1 are devoid of primary cilia stained for Ac-Tub (T). Nuclei are stained with DAPI (blue). (U) qRT-PCR of ductal and cystic disease genes in E14.5 pancreata (control, n=7; mutant, n=8). (V) ChIP showing Hnf1b fold-enrichment in regulatory regions of Hnf6, Cys1, Pkhd1, Glis3 and Bicc1 from E12.5 pancreata immunoprecipitated with an Hnf1b antibody versus control IgG. A scheme showing Hnf1-binding sites (red), relative to TSS (grey), is presented for each gene.

Fig. 5.

Hnf1b is crucial for duct morphogenesis. (A-F) Sox9 immunohistochemistry (brown) in control and Pdx1-Cre;Hnf1bFlox/LacZ pancreata at E14.5, E16.5 and E18.5. In mutants, note the epithelium multistratified regions at E14.5 (B; arrow), the cystic ducts from E16.5 (D,F; asterisks) with multilayered epithelia (D; arrow), a group of acinar cells ectopically expressing Sox9 (D; encircled in black) and the loss of ductal marker expression at E18.5 (F; arrows). Immunostaining at E16.5 for dystroglycan (green) and β-catenin (β-Cat, red) (G,H); laminin (green) (I,J); AQP1 (green) (K,L); ezrin (green) and PanCK (red) (M,N); PKCz (green) and β-Cat (red) (O,P); mucin 1 (MUC1, green) and β-Cat (red) (Q,R); and MUC1 (green) and acetylated α-Tubulin (Ac-Tub, red) (S,T). There is a strong decrease in dystroglycan and laminin basal marker staining in mutant acinar cells (H′,J′), and increased β-catenin staining in the apical region of acinar cells (H′) and in cystic ducts (R′). Ezrin, PKCz and MUC1 staining shows the loss of polarity of cystic ducts with absence or disruption of the apical staining (N′,P′,R′). Mutant ductal epithelial cells stained with MUC1 are devoid of primary cilia stained for Ac-Tub (T). Nuclei are stained with DAPI (blue). (U) qRT-PCR of ductal and cystic disease genes in E14.5 pancreata (control, n=7; mutant, n=8). (V) ChIP showing Hnf1b fold-enrichment in regulatory regions of Hnf6, Cys1, Pkhd1, Glis3 and Bicc1 from E12.5 pancreata immunoprecipitated with an Hnf1b antibody versus control IgG. A scheme showing Hnf1-binding sites (red), relative to TSS (grey), is presented for each gene.

Thus, lack of Hnf1b leads to increased acinar cell death and reduced expression of acinar cell markers, accompanied by ectopic expression of ductal markers. These results uncover an early role of Hnf1b in acinar differentiation and maintenance of acinar cell identity.

Hnf1b controls duct morphogenesis by regulating cystic disease-associated genes

We performed Sox9 immunostaining at different stages of pancreas development to further analyze duct morphogenesis (Fig. 5A-F). Cystic ducts at E16.5 in Pdx1-Cre;Hnf1bFlox/LacZ mutants showed some multilayered epithelium (Fig. 5B,D). Moreover, lack of Sox9 (Fig. 5E,F) or AQP1 expression (Fig. 5K-L′) revealed loss of ductal cell identity in large cystic ducts. We further analyzed the localization of polarity markers in mutants. Similar to PanCK (Fig. 4E-H), strong and expanded β-catenin expression to basal and apical membranes was observed in terminal enlarged ductal cells (Fig. 5G-H′). The basal expression of dystroglycan (Fig. 5G-H′) and laminin (Fig. 5I-J′) was also disrupted in most mutant acinar cells, further illustrating the defects in acinar cells and acquisition of ductal features. Importantly, although control ducts showed strong apical localization of ezrin, PKCz and MUC1 in epithelial cells around the duct lumen, expression of these apical markers in the cells lining cysts was reduced and discontinuous (Fig. 5M-R′). Cyst formation is often associated with an absence or dysfunction of primary cilia (Ware et al., 2011). Immunostaining of acetylated tubulin, a specific component of the cilium axoneme, revealed that cystic cells were devoid of primary cilia (Fig. 5S,T), whereas cilia were still present in non-cystic ducts, suggesting that Hnf1b is not required for primary cilium formation. Similar cystic ducts were observed in Sox9-CreERT2;Hnf1bFlox/LacZ (TM E9.5) mutants (supplementary material Fig. S4A,B), with altered ductal cell polarity and abnormal localization of β-catenin (supplementary material Fig. S4C,D). Notably, Sox9-CreERT2;Hnf1bFlox/LacZ (TM E12.5) mutants also displayed cystic ducts (supplementary material Fig. S5D,E). These data show that Hnf1b is required for morphogenesis and for epithelial polarization of ductal cells.

To gain insight into how Hnf1b controls ductal cell differentiation, we further investigated the expression of cystic disease genes in Pdx1-Cre;Hnf1bFlox/LacZ mutants. Although Hnf1b and Sox9 are both expressed in pancreatic ducts, and despite the cystic phenotype of pancreatic Sox9 mutants (Shih et al., 2012), we found no change in Sox9 expression in our mutants (Fig. 5U). By contrast, Hnf6 expression was strongly decreased in mutant ductal cells both by immunostaining (Fig. 4A,C) and by qRT-PCR at E14.5 (Fig. 5U), in line with the cystic phenotype of Hnf6−/− pancreata (Pierreux et al., 2006; Zhang et al., 2009). We also observed a decrease in Spp1 expression, which is directly regulated by Hnf1b in renal cells (Senkel et al., 2005). Importantly, we found a 90% downregulation of the autosomal recessive PKD gene Pkhd1 (Ward et al., 2002), and a dramatic decrease in the expression of the key cystic disease genes Kif12 (Mrug et al., 2005), Cys1 (Hou et al., 2002), Bicc1 (Cogswell et al., 2003; Lemaire et al., 2015) and Glis3 (Kang et al., 2009b) (Fig. 5U). Cys1 and Glis3 are of particular interest. Cys1 is responsible for congenital polycystic kidney (CPK) disease (Tao et al., 2009) and is involved in ciliogenesis and polarization of cholangiocytes (Raynaud et al., 2011), whereas Glis3 is implicated in polycystic disease in both kidney (Kang et al., 2009a) and pancreas (Kang et al., 2009b). Among these genes, Kif12, Pkhd1, Pkd2 and Bicc1 were identified as direct Hnf1b targets in the kidney (Gong et al., 2009; Gresh et al., 2004; Verdeguer et al., 2010). Thus, we analyzed whether Hnf1b could be a major regulator of these genes in the pancreas by ChIP experiments on E12.5 pancreata (Fig. 5V). Our results showed that Hnf1b is recruited to Hnf1-binding sites in the first intron of Hnf6, within a region known to drive Hnf6 expression in E8.75 pancreatic endoderm (Poll et al., 2006). Moreover, Hnf1b bound to a region carrying a site in the Pkhd1 promoter previously identified in the kidney (Gresh et al., 2004). Remarkably, we identified two novel Hnf1b target genes: Cys1 and Glis3 (Fig. 5V). These results suggest that Hnf1b is a key regulator of duct morphogenesis, exerting direct control of crucial genes involved in duct morphogenesis and cystogenesis.

Hnf1b expression in ducts controls exocrine morphogenesis

To examine the specific requirement of Hnf1b in the ductal compartment, we inactivated Hnf1b at late embryogenesis (∼E15), using the inducible Sox9-CreERT2 line with TM injection at E14.5. Overall pancreas morphology appeared to be normal in mutants at E18.5, and no change in expression of the ductal marker Sox9 and the acinar marker amylase were observed (Fig. 6A). However, we observed a dramatic decrease in expression of the cystic disease genes Pkhd1, Kif12, Cys1 and Glis3 (Fig. 6A). We further analyzed Sox9-CreERT2;Hnf1bFlox/LacZ (TM E14.5) mutant pancreata at postnatal day 8 (P8) and observed a significant reduction in pancreas weight (Fig. 6B). Histological analysis at P8 revealed many cystic ducts associated with a severe loss of acinar cells with enlarged acinar lumen often connected with enlarged terminal ducts (Fig. 6C,D). CPA1 immunostaining confirmed the decrease in the number of acinar cells in mutants (Fig. 6E,F). β-Catenin and MUC1 immunostaining revealed altered polarity of mutant ductal cells (Fig. 6G,H), which were also devoid of primary cilia (Fig. 6I,J). These data reinforce the specific role of Hnf1b in the control of duct morphogenesis, and suggest that its function in ducts contributes indirectly to the maintenance of acinar cells.

Fig. 6.

Hnf1b is required in ducts to maintain the exocrine compartment. (A) Hnf1b inactivation in ducts using the Sox9-CreERT2 line and TM injection at E14.5. qRT-PCR analysis of control and Sox9-CreERT2;Hnf1bFlox/LacZ (TM E14.5) mutant pancreata at E18.5 (control, n=14; mutant, n=6). (B) Relative pancreas weight/body weight of animals at P8 (control, n=8; mutant, n=3). *P<0.05; **P<0.005; ***P<0.001. (C,D) Hematoxylin/Eosin staining of control and Sox9-CreERT2;Hnf1bFlox/LacZ (TM E14.5) pancreata at P8. Arrow indicates a cystic duct and asterisks indicate dilated terminal ducts. (E,F) Carboxypeptidase (CPA1, green) staining showing fewer acinar cells in mutants. (G,H) MUC1 (green) and β-catenin (red) co-immunostaining shows cystic ducts and loss of polarity. (I,J) MUC1 (green) and Ac-Tub (red) co-immunostaining shows mutant ductal cells devoid of primary cilia. Nuclei are stained with DAPI (blue). Scale bars: 50 µm in C-H; 10 µm in I,J.

Fig. 6.

Hnf1b is required in ducts to maintain the exocrine compartment. (A) Hnf1b inactivation in ducts using the Sox9-CreERT2 line and TM injection at E14.5. qRT-PCR analysis of control and Sox9-CreERT2;Hnf1bFlox/LacZ (TM E14.5) mutant pancreata at E18.5 (control, n=14; mutant, n=6). (B) Relative pancreas weight/body weight of animals at P8 (control, n=8; mutant, n=3). *P<0.05; **P<0.005; ***P<0.001. (C,D) Hematoxylin/Eosin staining of control and Sox9-CreERT2;Hnf1bFlox/LacZ (TM E14.5) pancreata at P8. Arrow indicates a cystic duct and asterisks indicate dilated terminal ducts. (E,F) Carboxypeptidase (CPA1, green) staining showing fewer acinar cells in mutants. (G,H) MUC1 (green) and β-catenin (red) co-immunostaining shows cystic ducts and loss of polarity. (I,J) MUC1 (green) and Ac-Tub (red) co-immunostaining shows mutant ductal cells devoid of primary cilia. Nuclei are stained with DAPI (blue). Scale bars: 50 µm in C-H; 10 µm in I,J.

Hnf1b controls the generation of Ngn3+ endocrine precursors

As Hnf1b+ cells were defined as immediate precursors of Ngn3+ cells by immunohistochemistry (Maestro et al., 2003; Nammo et al., 2008; Rukstalis and Habener, 2007), and lineage-tracing analyses showed that embryonic Hnf1b+ cells give rise to precursors of endocrine cells (Solar et al., 2009), we analyzed whether Hnf1b is required for the generation of these cells. Interestingly, we observed an almost complete loss of Ngn3+ cells (Fig. 7A-F), associated with a 70%, 85% and 93% decrease in Ngn3 expression observed by qRT-PCR in Pdx1-Cre;Hnf1bFlox/LacZ pancreata at E12.5, E14.5 and E16.5, respectively (Fig. 7M). Endocrine cell differentiation was almost completely abrogated (Fig. 7G-L), as evidenced by a 93% decrease in insulin+ and glucagon+ areas at E16.5 (Fig. 7O), and decreased transcripts for insulin, glucagon and somatostatin (Fig. 7N). Sox9-CreERT2;Hnf1bFlox/LacZ (TM E9.5) mutants showed a similar dramatic loss of Ngn3+ cells (supplementary material Fig. S3A,B) and an 86% decrease in Ngn3 expression at E16.5 (supplementary material Fig. S3E). Insulin+ and glucagon+ areas were also severely reduced in size (supplementary material Fig. S3C,D), and insulin, glucagon and somatostatin mRNA levels decreased (supplementary material Fig. S3E).

Fig. 7.

Hnf1b controls the generation of endocrine precursors through Ngn3 regulation. (A,B) Immunostaining of Ngn3 (red) and E-Cadherin (E-CAD, green) in control and Pdx1-Cre;Hnf1bFlox/LacZ pancreata at E12.5. (C,D) Immunostaining of Ngn3 (red) and CPA1 (green) at E14.5. Arrows indicate a few remaining Ngn3+ cells in mutants (B,D). (E,F) Immunostaining of Ngn3 (red) and amylase (green) at E16.5. (G-L) Immunostaining of insulin (green) and glucagon (red) at E14.5, E16.5 and E18.5. Nuclei are stained with DAPI (blue). (M) qRT-PCR of Ngn3 at E12.5, E14.5 and E16.5. (N) qRT-PCR of glucagon, insulin and somatostatin at E14.5 and E16.5. (O) Quantification of glucagon+ and insulin+ sectional areas at E16.5. (P-U) Hnf1b inactivation during the secondary transition using the Sox9-CreERT2 line, with TM injection at E12.5 and analysis at E16.5. (P,Q) Immunostaining of amylase (green) and Ngn3 (red) in Sox9-CreERT2;Hnf1bFlox/LacZ (TM E12.5) pancreata at E16.5. (R,S) Immunostaining of insulin (green) and glucagon (red). Nuclei are stained with DAPI (blue). (T) qRT-PCR analysis in control and Sox9-CreERT2;Hnf1bFlox/LacZ (TM E12.5) pancreata at E16.5 (control, n=9; mutant, n=9). (U) Quantification of glucagon+ and insulin+ sectional areas at E16.5. (V) ChIP showing Hnf1b fold enrichment in regulatory regions of Ngn3 from E12.5 pancreata immunoprecipitated with an Hnf1b antibody versus control IgG. Hnf1-binding sites are shown in red with their positions relative to the Ngn3 TSS. *P<0.05; **P<0.005; ***P<0.001. Scale bars: 50 µm.

Fig. 7.

Hnf1b controls the generation of endocrine precursors through Ngn3 regulation. (A,B) Immunostaining of Ngn3 (red) and E-Cadherin (E-CAD, green) in control and Pdx1-Cre;Hnf1bFlox/LacZ pancreata at E12.5. (C,D) Immunostaining of Ngn3 (red) and CPA1 (green) at E14.5. Arrows indicate a few remaining Ngn3+ cells in mutants (B,D). (E,F) Immunostaining of Ngn3 (red) and amylase (green) at E16.5. (G-L) Immunostaining of insulin (green) and glucagon (red) at E14.5, E16.5 and E18.5. Nuclei are stained with DAPI (blue). (M) qRT-PCR of Ngn3 at E12.5, E14.5 and E16.5. (N) qRT-PCR of glucagon, insulin and somatostatin at E14.5 and E16.5. (O) Quantification of glucagon+ and insulin+ sectional areas at E16.5. (P-U) Hnf1b inactivation during the secondary transition using the Sox9-CreERT2 line, with TM injection at E12.5 and analysis at E16.5. (P,Q) Immunostaining of amylase (green) and Ngn3 (red) in Sox9-CreERT2;Hnf1bFlox/LacZ (TM E12.5) pancreata at E16.5. (R,S) Immunostaining of insulin (green) and glucagon (red). Nuclei are stained with DAPI (blue). (T) qRT-PCR analysis in control and Sox9-CreERT2;Hnf1bFlox/LacZ (TM E12.5) pancreata at E16.5 (control, n=9; mutant, n=9). (U) Quantification of glucagon+ and insulin+ sectional areas at E16.5. (V) ChIP showing Hnf1b fold enrichment in regulatory regions of Ngn3 from E12.5 pancreata immunoprecipitated with an Hnf1b antibody versus control IgG. Hnf1-binding sites are shown in red with their positions relative to the Ngn3 TSS. *P<0.05; **P<0.005; ***P<0.001. Scale bars: 50 µm.

To determine whether Hnf1b specifically controls the generation of endocrine precursors, excluding possible indirect effects due to the early Hnf1b deficiency in MPCs, we conditionally inactivated Hnf1b in the pancreatic epithelium during the secondary transition at ∼E13, when the major wave of endocrine cell neogenesis occurs. The Hnf1b-floxed locus was efficiently recombined in Sox9-CreERT2;Hnf1bFlox/LacZ (TM E12.5) pancreata at E16.5, as confirmed by a 75% decrease in Hnf1b expression (Fig. 7T). Mutant pancreas morphology and organ size appeared normal, and expression of the acinar markers Ptf1a, Mist1, Nr5a2 and amylase were only partially affected at E16.5 (supplementary material Fig. S5A). Strikingly, we found a dramatic loss of Ngn3+ endocrine precursors (Fig. 7P,Q) and a 90% decrease in Ngn3 expression at E16.5 (Fig. 7T). This coincided with a 81% decrease in insulin+ and a 69% decrease in glucagon+ sectional areas (Fig. 7R,S,U), and downregulation of insulin, glucagon and somatostatin transcripts (Fig. 7T). These results support the specific requirement for Hnf1b in the generation of Ngn3+ endocrine progenitors.

These data suggested that Hnf1b directly regulates Ngn3 expression. We performed ChIP experiments on E12.5 pancreata and found no Hnf1b enrichment −3318 base pairs (bp) upstream Ngn3 TSS, a region homologous to the human NGN3 cluster 1 enhancer containing a putative Hnf1-binding site (Lee et al., 2001). Importantly, we found that Hnf1b was recruited to a proximal and a distal region containing Hnf1-binding sites at −697 bp and −4890 bp, respectively (Fig. 7V).These results demonstrate that Hnf1b is specifically required to generate endocrine precursors, very likely by directly regulating Ngn3.

By Hnf1b conditional inactivation in the pancreas, our data demonstrate the essential functions exerted by Hnf1b in pancreatic MPC expansion and differentiation of exocrine and endocrine lineages, placing this transcription factor in a prominent position in the regulatory networks involved in these processes.

MPCs proliferation and survival

Pancreas hypoplasia observed in Pdx1-Cre;Hnf1bFlox/LacZ embryos is correlated with a reduced pool of MPCs, as it was previously shown that the progenitor pool defines the final pancreas size (Stanger et al., 2007). Hnf1b mutants display similar defects in MPC proliferation to Gata4/Gata6 compound pancreatic mutants (Xuan et al., 2012), and similar MPC apoptosis to Sox9 mutants (Seymour et al., 2007). Surprisingly, expression of the key transcription factors Pdx1, Sox9 and Ptf1a, which are involved in MPC expansion (Rieck et al., 2012), was not changed at E12.5 (data not shown), excluding their contribution to the MPC phenotype observed.

We observed that Notch signaling pathway is deregulated in Hnf1b mutant pancreata, showing a decrease in Dll1 expression and upregulation of Hey repressors. A direct regulation of Dll1 by Hnf1b, as recently shown in the kidney (Heliot et al., 2013; Massa et al., 2013), does not seem to occur in the pancreas, as ChIP on E12.5 pancreata showed no Hnf1b recruitment to a conserved region of Dll1 (data not shown). Recent data suggested a role for Dll1, which is activated by Ptf1a, in MPC proliferation (Ahnfelt-Ronne et al., 2012). Hes and Hey factors were found to inhibit the activity of the Ptf1 transcriptional complex by direct interaction with Ptf1a, without changing Ptf1a mRNA levels (Esni et al., 2004; Ghosh and Leach, 2006). Therefore, in Hnf1b mutants, the increased levels of Hey factors observed could inhibit Ptf1 transcriptional complex activity resulting in reduced Dll1 expression and MPC proliferation.

The reduced proliferation in Hnf1b-deficient pancreata could also be attributed to a decreased FGF signaling via downregulation of Fgfr4. Although the pancreatic phenotype of Fgfr4−/− embryos was not analyzed (Weinstein et al., 1998), several recent studies showed that Fgfr4 positively regulates proliferation and has anti-apoptotic effects in models of liver, prostate and gastric cancer (Drafahl et al., 2010; Ho et al., 2009; Miura et al., 2012; Ye et al., 2011). Interestingly, both Fgfr2b and Fgfr4 were downregulated in Sox9-deficient pancreas (Seymour et al., 2012), raising the possibility that they might serve partially redundant functions in MPC proliferation.

Acinar differentiation and duct morphogenesis

The severe reduction in the number of acinar cells in Pdx1-Cre;Hnf1bFlox/LacZ mutants was associated with a differentiation defect and apoptosis. The acinar compartment exhibited ectopic expression of the ductal markers Hnf6 and Sox9, which were shown to be required for acinar metaplasia and repression of acinar genes (Prevot et al., 2012). Moreover, the ductal compartment was increased in size without changing its proliferation rate, suggesting that acini were replaced by the expanded ductal compartment. This acinar defect could be associated with the dramatic decrease in Mist1 expression, as inhibition of Mist1 in acinar cells leads to severe defects, including acinar-to-ductal metaplasia (Zhu et al., 2004). The loss of acinar cell identity may also be explained by the previously described link between reduced Mist1 and Ptf1a expression, conversion of acinar cells into ductal cells and upregulation of Notch signaling (Rooman et al., 2006; Rovira et al., 2010; Shi et al., 2009). Hnf1b deletion at the onset of acinar cell differentiation (TM at E12.5) also resulted in increased Hey2 expression, even if less pronounced than in Pdx1-Cre;Hnf1bFlox/LacZ mutants, which correlated with downregulation of Mist1 and Ptf1a (supplementary material Fig. S5F), and fewer amylase-expressing cells (supplementary material Fig. S5B,C). These results suggest an early role for Hnf1b in the acquisition of pancreatic acinar cell identity from MPCs, possibly through Mist1, Ptf1a and Hey factors. Later Hnf1b deletion (TM at E14.5) was associated with a defect in acinar cell maintenance. As Hnf1b is not expressed in acinar cells, these late acinar defects might be an indirect consequence of abnormal duct morphogenesis, as described in pancreata deficient for Jag1 (Golson et al., 2009) or Kif3a (Cano et al., 2006).

Our results show that Hnf1b is required for both early and late control of duct morphogenesis; Hnf1b deletion resulted in cystic ducts with altered polarity and a lack of primary cilia. Cilia loss in ductal cells is an important event in pancreatic cyst development. However, despite lack of cilia throughout development in Kif3a mutants, duct dilatations do not occur before E17.5 (Cano et al., 2006). The pancreatic epithelium in Hnf1b mutants is dilated from E14.5, thus indicating that Hnf1b plays additional roles in duct morphogenesis. Conditional Hnf1b deletion at late embryogenesis also resulted in a polycystic pancreas postnatally, showing the direct requirement of Hnf1b in duct morphogenesis and further suggesting that the cystic phenotype is not a consequence of an early blocked endocrine differentiation (Magenheim et al., 2011). As in Hnf6−/− pancreata (Pierreux et al., 2006), cysts in Hnf1b mutants were not associated with increased epithelial cell proliferation. In Hnf6 and Sox9 mutants, cyst formation also seems to occur by deregulation of cystic-associated genes, but the genes involved are different. Sox9 mutants were characterized by a decrease in Pkd2 expression (Shih et al., 2012), whereas Hnf6 mutants displayed downregulation of Pkhd1 (Pierreux et al., 2006). We observed a marked decrease in Hnf6 expression in mutant pancreatic ducts at E14.5 upon deletion in MPCs, which correlated with the finding that Hnf1b is recruited at E12.5 to Hnf6 regulatory sequences known to drive expression in the pancreatic endoderm (Poll et al., 2006). Similarly, in Hnf6 mutants, Hnf1b expression is reduced during a narrow time window and then re-induced at late embryogenesis (Pierreux et al., 2006), illustrating the existence of a complex Hnf1b-Hnf6 feed-forward loop involved in duct morphogenesis at least between E12.5 and E16.5. However, duct morphogenetic defects of Hnf1b mutants are not simply explained by this transient cross-regulation. Indeed, duct morphogenesis is more largely affected in Hnf1b mutants than in Hnf6 mutants, as cysts affect all types of ducts, and not only intralobular and interlobular ducts, as in Hnf6 mutants (Pierreux et al., 2006). Moreover, whereas Glis3 expression was unaffected in Hnf6-null pancreas (Kang et al., 2009b), we found that Glis3 is downstream Hnf1b, which is particularly interesting as both factors are associated with cystogenesis (Kang et al., 2009a,,b) and endocrine cell development (Kim et al., 2012). Taking advantage of Hnf1b ChIP-seq analysis on embryonic kidneys, we identified Glis3 and Cys1 as novel Hnf1b targets in pancreas. Cys1 was found decreased in Hnf6 and Hnf1b mutant livers, although a direct regulation of Cys1 by Hnf1b was not established in biliary ducts (Raynaud et al., 2011). These studies show that Hnf1b is an essential regulator of key ductal genes related to cyst development in different organs. However, the regulatory circuits operating in ducts can diverge: Pkhd1 expression decreases in both pancreas and kidney, but not in the liver of Hnf1b mutants; Pkd2 expression decreases in the kidney, but not in the pancreas of Hnf1b mutants (Gresh et al., 2004; Hiesberger et al., 2005; Raynaud et al., 2011). Our study uncovers a crucial transcriptional network in pancreatic ductal cells, in which Hnf1b exerts a prominent role.

Control of endocrine progenitors

We show loss of Ngn3 expression upon Hnf1b inactivation during the first and secondary transitions, as well as the recruitment of Hnf1b to putative regulatory regions of Ngn3. This demonstrates an essential role of Hnf1b in the specification of endocrine progenitors. Additional transcription factors, including Hnf6, Glis3, Pdx1, Foxa2 and Sox9, were found to directly regulate Ngn3 expression (Jacquemin et al., 2000; Kim et al., 2012; Oliver-Krasinski et al., 2009; Seymour et al., 2008). Binding sites for some of these factors in the 5′ regulatory region of Ngn3 are listed in supplementary material Fig. S6. Hnf6−/− embryos exhibit markedly reduced numbers of Ngn3+ cells at mid-embryogenesis, associated with decreased Hnf1b expression (Jacquemin et al., 2000). By E17.5, Hnf1b expression is partially restored and reduction of Ngn3+ cells is clearly less pronounced when compared with earlier time points, supporting the notion that downregulation of Hnf1b in Hnf6 mutants might be important for Ngn3 downregulation (Maestro et al., 2003). Pdx1 also contributes to Ngn3 regulation and, together with Hnf6 (Oliver-Krasinski et al., 2009) and Glis3 (Kim et al., 2012; Yang et al., 2011), occupies an evolutionary conserved enhancer homologous to human cluster 1 (Lee et al., 2001). Ngn3 cluster 1 also contains putative Sox9-binding sites (at −3.3 kb), which were occupied by Sox9 in ChIP experiments on mPAC ductal cells (Lynn et al., 2007). However, ChIP on embryonic pancreata demonstrated that Sox9 was not bound to cluster 1, but to three other regions: one distal (at −4.0 kb) and two proximal (at −0.4 kb and −161 bp) (Seymour et al., 2008). Although there is a Hnf1-binding site in the homologous mouse cluster 1, Hnf1b, like Sox9, failed to transactivate this enhancer in transfected HePG2 cells (Oliver-Krasinski et al., 2009), which correlates with our findings showing no Hnf1b recruitment to this region by ChIP. Moreover, Hnf1b did not transactivate the Ngn3 promoter corresponding to −4864 bp to +88 bp (Ejarque et al., 2013), whereas this factor was able to activate the Ngn3 full promoter (−5800 to +40 bp) (Yang et al., 2011), suggesting that the Hnf1b-binding site we identified at −4890 bp might be important for Ngn3 activation. These studies support the existence of distinct enhancer modules with differential binding of essential transcription factors that contribute to the activation of Ngn3. Our results show the absolute requirement of Hnf1b for endocrine specification, placing this factor in a prominent position in the regulatory network controlling Ngn3 expression.

A model depicting Hnf1b functions during pancreas development is presented in Fig. 8. As increasing the pool of endocrine progenitors is a key step for the development of cell-based strategies for diabetes, these findings might be of clinical significance to improve in vitro protocols for cell-replacement therapies. Proper regulation of Hnf1b expression appears to be crucial for endocrine cell formation, and Hnf1b can be used as a marker of progenitor cells with the capacity to robustly produce endocrine cells in vitro. In addition, our results suggest that MODY might occur not only as a consequence of β-cell dysfunction, but also as a consequence of defects during development leading to diabetes later in life.

Fig. 8.

Proposed model for Hnf1b function in the regulatory networks governing pancreas development. Hnf1b appears to control MPC proliferation and survival by directly regulating Fgfr4 and acting as a negative regulator of the Hey factors, which are inhibitors of the Ptf1a transcriptional complex that in turn activates Dll1. Hnf1b indirectly controls acinar cell identity and maintenance, possibly through Mist1, Ptf1a and Hey factors. Subsequently, Hnf1b regulates epithelial duct morphogenesis through the direct control of the key cystic disease genes Glis3, Pkhd1, Cys1 and Hnf6. Hnf1b is absolutely required for the generation of endocrine progenitors, being an essential transcriptional activator of Ngn3. Hnf1b appears also to act directly on Glis3, but in parallel with Sox9 and Hes1 in the control of Ngn3. Arrows indicate epistatic or direct interactions, and double-headed arrows indicate protein-protein interactions. Double arrows indicate a Hnf1b-Hnf6 cross-regulatory loop between E12.5 and E16.5.

Fig. 8.

Proposed model for Hnf1b function in the regulatory networks governing pancreas development. Hnf1b appears to control MPC proliferation and survival by directly regulating Fgfr4 and acting as a negative regulator of the Hey factors, which are inhibitors of the Ptf1a transcriptional complex that in turn activates Dll1. Hnf1b indirectly controls acinar cell identity and maintenance, possibly through Mist1, Ptf1a and Hey factors. Subsequently, Hnf1b regulates epithelial duct morphogenesis through the direct control of the key cystic disease genes Glis3, Pkhd1, Cys1 and Hnf6. Hnf1b is absolutely required for the generation of endocrine progenitors, being an essential transcriptional activator of Ngn3. Hnf1b appears also to act directly on Glis3, but in parallel with Sox9 and Hes1 in the control of Ngn3. Arrows indicate epistatic or direct interactions, and double-headed arrows indicate protein-protein interactions. Double arrows indicate a Hnf1b-Hnf6 cross-regulatory loop between E12.5 and E16.5.

Mouse transgenic lines and physiological analyses

Mice carrying the Hnf1b-null allele (Hnf1btmsc1 known as Hnf1b+/LacZ), with the LacZ gene replacing the first exon of Hnf1b (Barbacci et al., 1999), were maintained as heterozygotes. The Hnf1b conditional knockout (Hnf1btm1Ics denoted as Hnf1bFlox/Flox) carrying LoxP sites flanking exon 4 (Heliot et al., 2013), Pdx1-Cre (Wells et al., 2007) and Sox9-CreERT2 (Kopp et al., 2011) lines have been previously described. The R26RYFP line (B6.129X1-Gt(ROSA)26Sortm1(EYFP)Cos/J) was from The Jackson Laboratory. 4-Hydroxytamoxifen (Sigma) was dissolved at 10 mg/ml in corn oil/10% ethanol and administrated intraperitoneally to pregnant females at a dose of 2 mg. Blood glucose levels were measured using the OneTouch Vita blood glucose meter (LifeScan), blood amylase using the Reflovet Plus analyzer and pancreatic amylase by reflotron assay (Roche). Animal experiments were conducted in accordance with French and European ethical legal guidelines and the local ethical committee for animal care.

Histology, immunohistochemistry and TUNEL assay

Tissues were fixed, embedded in paraffin, sectioned and analyzed by histology, immunohistochemistry and TUNEL assay as described previously (Haumaitre et al., 2005). Primary and secondary antibodies are listed in supplementary material Table S2. The percentage of Hnf1b+ cells was quantified by counting the number of Pdx1+ cells that were also Hnf1b+, on at least five sections per pancreas (n=4). Quantification of Pdx1+ cells at E12.5 was performed with at least five sections per pancreas (control, n=4; mutant, n=4). More than 10,000 Pdx1+ cells were counted for each genotype. Quantification of amylase+, Sox9+, insulin+ and glucagon+ cell surface was performed using ImageJ software, on at least three sections per pancreas at E16.5 co-immunostained with DAPI (control, n=3; mutant, n=5). Primary cilia were analyzed by confocal microscope (LEICA TSC SPE).

RNA extraction and quantitative PCR

Total RNA from embryonic pancreata was isolated using RNeasy Micro-kit (Qiagen) and reverse transcribed using the superscript II RT First-Strand Synthesis System (Life Technologies). qRT-PCR was performed using the Fast SYBR Green Master Mix (Life Technologies). Primer sequences are provided in supplementary material Table S2. The 2−ΔΔCt method was used to calculate expression levels (Livak and Schmittgen, 2001), normalized to cyclophilin A and relative to wild-type cDNA from E15.5 pancreata. Values are shown as mean+s.e.m. Statistical significance was determined using Student's t-test (NS, not significant; *P<0.05; **P<0.005; ***P<0.001).

Chromatin immunoprecipitation

Chromatin immunoprecipitation (ChIP) experiments were performed as described previously (Heliot and Cereghini, 2012), adapted for E12.5 wild-type pancreata. Immunoprecipitated chromatin were analyzed by qPCR and expressed as fold-enrichment values over IgG control. Primer sequences are provided in supplementary material Table S2. Values are shown as mean+s.e.m. Statistical significance was determined using Student's t-test (NS, not significant; *P<0.05; **P<0.005; ***P<0.001). A detailed protocol is provided in the methods in the supplementary material.

Funding

This work was supported by the European Union's Framework Program 7 (EU-FP7)- Marie Curie Initial Training Network (ITN)-Biology of Liver and Pancreatic Development and Disease (BOLD), by the Centre National de la Recherche Scientifique (CNRS), by the Université Pierre et Marie Curie (UPMC) and by the Institut National de la Santé et de la Recherche Médicale (INSERM) (to S.C. and C. Haumaitre); and by the programme Emergence UPMC and the Société Francophone du Diabète (SFD)-Allocation SFD-Industrie Ypsomed (to C. Haumaitre). M.G.D.V. and C. Heliot were recipients of PhD student fellowships from ITN BOLD and the Fondation pour la Recherche sur le Cancer (ARC), respectively. M.S. is supported by the National Institutes of Health (NIH) – NIDDK [R01-DK078803] and J.L.K. was supported by an Advanced Postdoctoral Fellowship from the Juvenile Diabetes Research Foundation (JDRF). Deposited in PMC for release after 12 months.

Author contributions

M.G.D.V. performed experiments, analyzed data and wrote the manuscript. M.S. and J.L.K. provided the Sox9-CreERT2 mouse line, advice on experimental procedures and carefully read the manuscript. C. Heliot performed ChIP-seq experiments. S.C. designed the study, contributed to discussions and revised the manuscript. C. Haumaitre designed the study, performed experiments, analyzed data and wrote the manuscript.

We thank C. Wright (Vanderbilt University, TN, USA), P. Jacquemin and F. Lemaigre (De Duve Institute, Belgium), and S. Louvet (UMR7622, France) for antibodies; the Centre d'Expérimentation Fonctionnelle (Faculté de Médecine Pitié-Salpêtrière, France) for blood amylase measures; mouse facilities of the UMR722-IBPS; and S. Gournet for illustration.

Afelik
,
S.
and
Jensen
,
J.
(
2013
).
Notch signaling in the pancreas: patterning and cell fate specification
.
Wiley Interdiscip. Rev. Dev. Biol.
2
,
531
-
544
.
Ahnfelt-Ronne
,
J.
,
Jorgensen
,
M. C.
,
Klinck
,
R.
,
Jensen
,
J. N.
,
Fuchtbauer
,
E.-M.
,
Deering
,
T.
,
MacDonald
,
R. J.
,
Wright
,
C. V. E.
,
Madsen
,
O. D.
and
Serup
,
P.
(
2012
).
Ptf1a-mediated control of Dll1 reveals an alternative to the lateral inhibition mechanism
.
Development
139
,
33
-
45
.
Apelqvist
,
A.
,
Li
,
H.
,
Sommer
,
L.
,
Beatus
,
P.
,
Anderson
,
D. J.
,
Honjo
,
T.
,
Hrabě de Angelis
,
M.
,
Lendahl
,
U.
and
Edlund
,
H.
(
1999
).
Notch signalling controls pancreatic cell differentiation
.
Nature
400
,
877
-
881
.
Barbacci
,
E.
,
Reber
,
M.
,
Ott
,
M. O.
,
Breillat
,
C.
,
Huetz
,
F.
and
Cereghini
,
S.
(
1999
).
Variant hepatocyte nuclear factor 1 is required for visceral endoderm specification
.
Development
126
,
4795
-
4805
.
Bellanné-Chantelot
,
C.
,
Chauveau
,
D.
,
Gautier
,
J.-F.
,
Dubois-Laforgue
,
D.
,
Clauin
,
S.
,
Beaufils
,
S.
,
Wilhelm
,
J.-M.
,
Boitard
,
C.
,
Noël
,
L.-H.
,
Velho
,
G.
, et al. 
(
2004
).
Clinical spectrum associated with hepatocyte nuclear factor-1beta mutations
.
Ann. Intern. Med.
140
,
510
-
517
.
Bhushan
,
A.
,
Itoh
,
N.
,
Kato
,
S.
,
Thiery
,
J. P.
,
Czernichow
,
P.
,
Bellusci
,
S.
and
Scharfmann
,
R.
(
2001
).
Fgf10 is essential for maintaining the proliferative capacity of epithelial progenitor cells during early pancreatic organogenesis
.
Development
128
,
5109
-
5117
.
Cano
,
D. A.
,
Sekine
,
S.
and
Hebrok
,
M.
(
2006
).
Primary cilia deletion in pancreatic epithelial cells results in cyst formation and pancreatitis
.
Gastroenterology
131
,
1856
-
1869
.
Chen
,
Y. Z.
,
Gao
,
Q.
,
Zhao
,
X. Z.
,
Chen
,
Y. Z.
,
Bennett
,
C. L.
,
Xiong
,
X. S.
,
Mei
,
C. L.
,
Shi
,
Y. Q.
and
Chen
,
X. M.
(
2010
).
Systematic review of TCF2 anomalies in renal cysts and diabetes syndrome/maturity onset diabetes of the young type 5
.
Chin. Med. J.
123
,
3326
-
3333
.
Cogswell
,
C.
,
Price
,
S. J.
,
Hou
,
X.
,
Guay-Woodford
,
L. M.
,
Flaherty
,
L.
and
Bryda
,
E. C.
(
2003
).
Positional cloning of jcpk/bpk locus of the mouse
.
Mamm. Genome
14
,
242
-
249
.
Drafahl
,
K. A.
,
McAndrew
,
C. W.
,
Meyer
,
A. N.
,
Haas
,
M.
and
Donoghue
,
D. J.
(
2010
).
The receptor tyrosine kinase FGFR4 negatively regulates NF-kappaB signaling
.
PLoS ONE
5
,
e14412
.
Dubois
,
C. L.
,
Shih
,
H. P.
,
Seymour
,
P. A.
,
Patel
,
N. A.
,
Behrmann
,
J. M.
,
Ngo
,
V.
and
Sander
,
M.
(
2011
).
Sox9-haploinsufficiency causes glucose intolerance in mice
.
PLoS ONE
6
,
e23131
.
Edghill
,
E. L.
,
Bingham
,
C.
,
Slingerland
,
A. S.
,
Minton
,
J. A. L.
,
Noordam
,
C.
,
Ellard
,
S.
and
Hattersley
,
A. T.
(
2006
).
Hepatocyte nuclear factor-1 beta mutations cause neonatal diabetes and intrauterine growth retardation: support for a critical role of HNF-1beta in human pancreatic development
.
Diabet. Med.
23
,
1301
-
1306
.
Ejarque
,
M.
,
Cervantes
,
S.
,
Pujadas
,
G.
,
Tutusaus
,
A.
,
Sanchez
,
L.
and
Gasa
,
R.
(
2013
).
Neurogenin3 cooperates with Foxa2 to autoactivate its own expression
.
J. Biol. Chem.
288
,
11705
-
11717
.
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. 
(
2004
).
Notch inhibits Ptf1 function and acinar cell differentiation in developing mouse and zebrafish pancreas
.
Development
131
,
4213
-
4224
.
Gao
,
N.
,
LeLay
,
J.
,
Vatamaniuk
,
M. Z.
,
Rieck
,
S.
,
Friedman
,
J. R.
and
Kaestner
,
K. H.
(
2008
).
Dynamic regulation of Pdx1 enhancers by Foxa1 and Foxa2 is essential for pancreas development
.
Genes Dev.
22
,
3435
-
3448
.
Ghosh
,
B.
and
Leach
,
S. D.
(
2006
).
Interactions between hairy/enhancer of split-related proteins and the pancreatic transcription factor Ptf1-p48 modulate function of the PTF1 transcriptional complex
.
Biochem. J.
393
,
679
-
685
.
Golson
,
M. L.
,
Loomes
,
K. M.
,
Oakey
,
R.
and
Kaestner
,
K. H.
(
2009
).
Ductal malformation and pancreatitis in mice caused by conditional Jag1 deletion
.
Gastroenterology
136
,
1761
-
1771.e1
.
Gong
,
Y.
,
Ma
,
Z.
,
Patel
,
V.
,
Fischer
,
E.
,
Hiesberger
,
T.
,
Pontoglio
,
M.
and
Igarashi
,
P.
(
2009
).
HNF-1beta regulates transcription of the PKD modifier gene Kif12
.
J. Am. Soc. Nephrol.
20
,
41
-
47
.
Gradwohl
,
G.
,
Dierich
,
A.
,
LeMeur
,
M.
and
Guillemot
,
F.
(
2000
).
neurogenin3 is required for the development of the four endocrine cell lineages of the pancreas
.
Proc. Natl. Acad. Sci. USA
97
,
1607
-
1611
.
Gresh
,
L.
,
Fischer
,
E.
,
Reimann
,
A.
,
Tanguy
,
M.
,
Garbay
,
S.
,
Shao
,
X.
,
Hiesberger
,
T.
,
Fiette
,
L.
,
Igarashi
,
P.
,
Yaniv
,
M.
, et al. 
(
2004
).
A transcriptional network in polycystic kidney disease
.
EMBO J.
23
,
1657
-
1668
.
Hald
,
J.
,
Hjorth
,
J. P.
,
German
,
M. S.
,
Madsen
,
O. D.
,
Serup
,
P.
and
Jensen
,
J.
(
2003
).
Activated Notch1 prevents differentiation of pancreatic acinar cells and attenuate endocrine development
.
Dev. Biol.
260
,
426
-
437
.
Haldorsen
,
I. S.
,
Vesterhus
,
M.
,
Raeder
,
H.
,
Jensen
,
D. K.
,
Søvik
,
O.
,
Molven
,
A.
and
Njølstad
,
P. R.
(
2008
).
Lack of pancreatic body and tail in HNF1B mutation carriers
.
Diabet. Med.
25
,
782
-
787
.
Hale
,
M. A.
,
Swift
,
G. H.
,
Hoang
,
C. Q.
,
Deering
,
T. G.
,
Masui
,
T.
,
Lee
,
Y.-K.
,
Xue
,
J.
and
MacDonald
,
R. J.
(
2014
).
The nuclear hormone receptor family member NR5A2 controls aspects of multipotent progenitor cell formation and acinar differentiation during pancreatic organogenesis
.
Development
141
,
3123
-
3133
.
Hart
,
A.
,
Papadopoulou
,
S.
and
Edlund
,
H.
(
2003
).
Fgf10 maintains notch activation, stimulates proliferation, and blocks differentiation of pancreatic epithelial cells
.
Dev. Dyn.
228
,
185
-
193
.
Haumaitre
,
C.
,
Barbacci
,
E.
,
Jenny
,
M.
,
Ott
,
M. O.
,
Gradwohl
,
G.
and
Cereghini
,
S.
(
2005
).
Lack of TCF2/vHNF1 in mice leads to pancreas agenesis
.
Proc. Natl. Acad. Sci. USA
102
,
1490
-
1495
.
Haumaitre
,
C.
,
Fabre
,
M.
,
Cormier
,
S.
,
Baumann
,
C.
,
Delezoide
,
A.-L.
and
Cereghini
,
S.
(
2006
).
Severe pancreas hypoplasia and multicystic renal dysplasia in two human fetuses carrying novel HNF1beta/MODY5 mutations
.
Hum. Mol. Genet.
15
,
2363
-
2375
.
Heliot
,
C.
and
Cereghini
,
S.
(
2012
).
Analysis of in vivo transcription factor recruitment by chromatin immunoprecipitation of mouse embryonic kidney
.
Methods Mol. Biol.
886
,
275
-
291
.
Heliot
,
C.
,
Desgrange
,
A.
,
Buisson
,
I.
,
Prunskaite-Hyyrylainen
,
R.
,
Shan
,
J.
,
Vainio
,
S.
,
Umbhauer
,
M.
and
Cereghini
,
S.
(
2013
).
HNF1B controls proximal-intermediate nephron segment identity in vertebrates by regulating Notch signalling components and Irx1/2
.
Development
140
,
873
-
885
.
Hiesberger
,
T.
,
Shao
,
X.
,
Gourley
,
E.
,
Reimann
,
A.
,
Pontoglio
,
M.
and
Igarashi
,
P.
(
2005
).
Role of the hepatocyte nuclear factor-1beta (HNF-1beta) C-terminal domain in Pkhd1 (ARPKD) gene transcription and renal cystogenesis
.
J. Biol. Chem.
280
,
10578
-
10586
.
Ho
,
H. K.
,
Pok
,
S.
,
Streit
,
S.
,
Ruhe
,
J. E.
,
Hart
,
S.
,
Lim
,
K. S.
,
Loo
,
H. L.
,
Aung
,
M. O.
,
Lim
,
S. G.
and
Ullrich
,
A.
(
2009
).
Fibroblast growth factor receptor 4 regulates proliferation, anti-apoptosis and alpha-fetoprotein secretion during hepatocellular carcinoma progression and represents a potential target for therapeutic intervention
.
J. Hepatol.
50
,
118
-
127
.
Hou
,
X.
,
Mrug
,
M.
,
Yoder
,
B. K.
,
Lefkowitz
,
E. J.
,
Kremmidiotis
,
G.
,
D'Eustachio
,
P.
,
Beier
,
D. R.
and
Guay-Woodford
,
L. M.
(
2002
).
Cystin, a novel cilia-associated protein, is disrupted in the cpk mouse model of polycystic kidney disease
.
J. Clin. Invest.
109
,
533
-
540
.
Jacquemin
,
P.
,
Durviaux
,
S. M.
,
Jensen
,
J.
,
Godfraind
,
C.
,
Gradwohl
,
G.
,
Guillemot
,
F.
,
Madsen
,
O. D.
,
Carmeliet
,
P.
,
Dewerchin
,
M.
,
Collen
,
D.
, et al. 
(
2000
).
Transcription factor hepatocyte nuclear factor 6 regulates pancreatic endocrine cell differentiation and controls expression of the proendocrine gene ngn3
.
Mol. Cell. Biol.
20
,
4445
-
4454
.
Jensen
,
J.
,
Pedersen
,
E. E.
,
Galante
,
P.
,
Hald
,
J.
,
Heller
,
R. S.
,
Ishibashi
,
M.
,
Kageyama
,
R.
,
Guillemot
,
F.
,
Serup
,
P.
and
Madsen
,
O. D.
(
2000
).
Control of endodermal endocrine development by Hes-1
.
Nat. Genet.
24
,
36
-
44
.
Kaestner
,
K. H.
,
Katz
,
J.
,
Liu
,
Y.
,
Drucker
,
D. J.
and
Schutz
,
G.
(
1999
).
Inactivation of the winged helix transcription factor HNF3alpha affects glucose homeostasis and islet glucagon gene expression in vivo
.
Genes Dev.
13
,
495
-
504
.
Kang
,
H. S.
,
Beak
,
J. Y.
,
Kim
,
Y.-S.
,
Herbert
,
R.
and
Jetten
,
A. M.
(
2009a
).
Glis3 is associated with primary cilia and Wwtr1/TAZ and implicated in polycystic kidney disease
.
Mol. Cell. Biol.
29
,
2556
-
2569
.
Kang
,
H. S.
,
Kim
,
Y.-S.
,
ZeRuth
,
G.
,
Beak
,
J. Y.
,
Gerrish
,
K.
,
Kilic
,
G.
,
Sosa-Pineda
,
B.
,
Jensen
,
J.
,
Foley
,
J.
and
Jetten
,
A. M.
(
2009b
).
Transcription factor Glis3, a novel critical player in the regulation of pancreatic beta-cell development and insulin gene expression
.
Mol. Cell. Biol.
29
,
6366
-
6379
.
Kim
,
Y.-S.
,
Kang
,
H. S.
,
Takeda
,
Y.
,
Hom
,
L.
,
Song
,
H.-Y.
,
Jensen
,
J.
and
Jetten
,
A. M.
(
2012
).
Glis3 regulates neurogenin 3 expression in pancreatic beta-cells and interacts with its activator, Hnf6
.
Mol. Cells
34
,
193
-
200
.
Kopp
,
J. L.
,
Dubois
,
C. L.
,
Schaffer
,
A. E.
,
Hao
,
E.
,
Shih
,
H. P.
,
Seymour
,
P. A.
,
Ma
,
J.
and
Sander
,
M.
(
2011
).
Sox9+ ductal cells are multipotent progenitors throughout development but do not produce new endocrine cells in the normal or injured adult pancreas
.
Development
138
,
653
-
665
.
Kornfeld
,
J.-W.
,
Baitzel
,
C.
,
Könner
,
A. C.
,
Nicholls
,
H. T.
,
Vogt
,
M. C.
,
Herrmanns
,
K.
,
Scheja
,
L.
,
Haumaitre
,
C.
,
Wolf
,
A. M.
,
Knippschild
,
U.
, et al. 
(
2013
).
Obesity-induced overexpression of miR-802 impairs glucose metabolism through silencing of Hnf1b
.
Nature
494
,
111
-
115
.
Lee
,
J. C.
,
Smith
,
S. B.
,
Watada
,
H.
,
Lin
,
J.
,
Scheel
,
D.
,
Wang
,
J.
,
Mirmira
,
R. G.
and
German
,
M. S.
(
2001
).
Regulation of the pancreatic pro-endocrine gene neurogenin3
.
Diabetes
50
,
928
-
936
.
Lemaire
,
L. A.
,
Goulley
,
J.
,
Kim
,
Y. H.
,
Carat
,
S.
,
Jacquemin
,
P.
,
Rougemont
,
J.
,
Constam
,
D. B.
and
Grapin-Botton
,
A.
(
2015
).
Bicaudal C1 promotes pancreatic NEUROG3+ endocrine progenitor differentiation and ductal morphogenesis
.
Development
142
,
858
-
870
.
Livak
,
K. J.
and
Schmittgen
,
T. D.
(
2001
).
Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method
.
Methods
25
,
402
-
408
.
Lokmane
,
L.
,
Haumaitre
,
C.
,
Garcia-Villalba
,
P.
,
Anselme
,
I.
,
Schneider-Maunoury
,
S.
and
Cereghini
,
S.
(
2008
).
Crucial role of vHNF1 in vertebrate hepatic specification
.
Development
135
,
2777
-
2786
.
Lynn
,
F. C.
,
Smith
,
S. B.
,
Wilson
,
M. E.
,
Yang
,
K. Y.
,
Nekrep
,
N.
and
German
,
M. S.
(
2007
).
Sox9 coordinates a transcriptional network in pancreatic progenitor cells
.
Proc. Natl. Acad. Sci. USA
104
,
10500
-
10505
.
Maestro
,
M. A.
,
Boj
,
S. F.
,
Luco
,
R. F.
,
Pierreux
,
C. E.
,
Cabedo
,
J.
,
Servitja
,
J. M.
,
German
,
M. S.
,
Rousseau
,
G. G.
,
Lemaigre
,
F. P.
and
Ferrer
,
J.
(
2003
).
Hnf6 and Tcf2 (MODY5) are linked in a gene network operating in a precursor cell domain of the embryonic pancreas
.
Hum. Mol. Genet.
12
,
3307
-
3314
.
Magenheim
,
J.
,
Klein
,
A. M.
,
Stanger
,
B. Z.
,
Ashery-Padan
,
R.
,
Sosa-Pineda
,
B.
,
Gu
,
G.
and
Dor
,
Y.
(
2011
).
Ngn3(+) endocrine progenitor cells control the fate and morphogenesis of pancreatic ductal epithelium
.
Dev. Biol.
359
,
26
-
36
.
Massa
,
F.
,
Garbay
,
S.
,
Bouvier
,
R.
,
Sugitani
,
Y.
,
Noda
,
T.
,
Gubler
,
M.-C.
,
Heidet
,
L.
,
Pontoglio
,
M.
and
Fischer
,
E.
(
2013
).
Hepatocyte nuclear factor 1beta controls nephron tubular development
.
Development
140
,
886
-
896
.
Miura
,
S.
,
Mitsuhashi
,
N.
,
Shimizu
,
H.
,
Kimura
,
F.
,
Yoshidome
,
H.
,
Otsuka
,
M.
,
Kato
,
A.
,
Shida
,
T.
,
Okamura
,
D.
and
Miyazaki
,
M.
(
2012
).
Fibroblast growth factor 19 expression correlates with tumor progression and poorer prognosis of hepatocellular carcinoma
.
BMC Cancer
12
,
56
.
Mrug
,
M.
,
Li
,
R.
,
Cui
,
X.
,
Schoeb
,
T. R.
,
Churchill
,
G. A.
and
Guay-Woodford
,
L. M.
(
2005
).
Kinesin family member 12 is a candidate polycystic kidney disease modifier in the cpk mouse
.
J. Am. Soc. Nephrol.
16
,
905
-
916
.
Murtaugh
,
L. C.
,
Stanger
,
B. Z.
,
Kwan
,
K. M.
and
Melton
,
D. A.
(
2003
).
Notch signaling controls multiple steps of pancreatic differentiation
.
Proc. Natl. Acad. Sci. USA
100
,
14920
-
14925
.
Nammo
,
T.
,
Yamagata
,
K.
,
Tanaka
,
T.
,
Kodama
,
T.
,
Sladek
,
F. M.
,
Fukui
,
K.
,
Katsube
,
F.
,
Sato
,
Y.
,
Miyagawa
,
J.-i.
and
Shimomura
,
I.
(
2008
).
Expression of HNF-4alpha (MODY1), HNF-1beta (MODY5), and HNF-1alpha (MODY3) proteins in the developing mouse pancreas
.
Gene Expr. Patterns
8
,
96
-
106
.
Oliver-Krasinski
,
J. M.
,
Kasner
,
M. T.
,
Yang
,
J.
,
Crutchlow
,
M. F.
,
Rustgi
,
A. K.
,
Kaestner
,
K. H.
and
Stoffers
,
D. A.
(
2009
).
The diabetes gene Pdx1 regulates the transcriptional network of pancreatic endocrine progenitor cells in mice
.
J. Clin. Invest.
119
,
1888
-
1898
.
Pan
,
F. C.
and
Wright
,
C.
(
2011
).
Pancreas organogenesis: from bud to plexus to gland
.
Dev. Dyn.
240
,
530
-
565
.
Pierreux
,
C. E.
,
Poll
,
A. V.
,
Kemp
,
C. R.
,
Clotman
,
F.
,
Maestro
,
M. A.
,
Cordi
,
S.
,
Ferrer
,
J.
,
Leyns
,
L.
,
Rousseau
,
G. G.
and
Lemaigre
,
F. P.
(
2006
).
The transcription factor hepatocyte nuclear factor-6 controls the development of pancreatic ducts in the mouse
.
Gastroenterology
130
,
532
-
541
.
Pin
,
C. L.
,
Rukstalis
,
J. M.
,
Johnson
,
C.
and
Konieczny
,
S. F.
(
2001
).
The bHLH transcription factor Mist1 is required to maintain exocrine pancreas cell organization and acinar cell identity
.
J. Cell Biol.
155
,
519
-
530
.
Poll
,
A. V.
,
Pierreux
,
C. E.
,
Lokmane
,
L.
,
Haumaitre
,
C.
,
Achouri
,
Y.
,
Jacquemin
,
P.
,
Rousseau
,
G. G.
,
Cereghini
,
S.
and
Lemaigre
,
F. P.
(
2006
).
A vHNF1/TCF2-HNF6 cascade regulates the transcription factor network that controls generation of pancreatic precursor cells
.
Diabetes
55
,
61
-
69
.
Prevot
,
P.-P.
,
Simion
,
A.
,
Grimont
,
A.
,
Colletti
,
M.
,
Khalaileh
,
A.
,
Van den Steen
,
G.
,
Sempoux
,
C.
,
Xu
,
X.
,
Roelants
,
V.
,
Hald
,
J.
, et al. 
(
2012
).
Role of the ductal transcription factors HNF6 and Sox9 in pancreatic acinar-to-ductal metaplasia
.
Gut
61
,
1723
-
1732
.
Pulkkinen
,
M.-A.
,
Spencer-Dene
,
B.
,
Dickson
,
C.
and
Otonkoski
,
T.
(
2003
).
The IIIb isoform of fibroblast growth factor receptor 2 is required for proper growth and branching of pancreatic ductal epithelium but not for differentiation of exocrine or endocrine cells
.
Mech. Dev.
120
,
167
-
175
.
Raynaud
,
P.
,
Tate
,
J.
,
Callens
,
C.
,
Cordi
,
S.
,
Vandersmissen
,
P.
,
Carpentier
,
R.
,
Sempoux
,
C.
,
Devuyst
,
O.
,
Pierreux
,
C. E.
,
Courtoy
,
P.
, et al. 
(
2011
).
A classification of ductal plate malformations based on distinct pathogenic mechanisms of biliary dysmorphogenesis
.
Hepatology
53
,
1959
-
1966
.
Rieck
,
S.
,
Bankaitis
,
E. D.
and
Wright
,
C. V. E.
(
2012
).
Lineage determinants in early endocrine development
.
Semin. Cell Dev. Biol.
23
,
673
-
684
.
Rooman
,
I.
,
De Medts
,
N.
,
Baeyens
,
L.
,
Lardon
,
J.
,
De Breuck
,
S.
,
Heimberg
,
H.
and
Bouwens
,
L.
(
2006
).
Expression of the Notch signaling pathway and effect on exocrine cell proliferation in adult rat pancreas
.
Am. J. Pathol.
169
,
1206
-
1214
.
Rovira
,
M.
,
Scott
,
S.-G.
,
Liss
,
A. S.
,
Jensen
,
J.
,
Thayer
,
S. P.
and
Leach
,
S. D.
(
2010
).
Isolation and characterization of centroacinar/terminal ductal progenitor cells in adult mouse pancreas
.
Proc. Natl. Acad. Sci. USA
107
,
75
-
80
.
Rukstalis
,
J. M.
and
Habener
,
J. F.
(
2007
).
Snail2, a mediator of epithelial-mesenchymal transitions, expressed in progenitor cells of the developing endocrine pancreas
.
Gene Expr. Patterns
7
,
471
-
479
.
Senkel
,
S.
,
Lucas
,
B.
,
Klein-Hitpass
,
L.
and
Ryffel
,
G. U.
(
2005
).
Identification of target genes of the transcription factor HNF1beta and HNF1alpha in a human embryonic kidney cell line
.
Biochim. Biophys. Acta
1731
,
179
-
190
.
Seymour
,
P. A.
and
Sander
,
M.
(
2011
).
Historical perspective: beginnings of the beta-cell: current perspectives in beta-cell development
.
Diabetes
60
,
364
-
376
.
Seymour
,
P. A.
,
Freude
,
K. K.
,
Tran
,
M. N.
,
Mayes
,
E. E.
,
Jensen
,
J.
,
Kist
,
R.
,
Scherer
,
G.
and
Sander
,
M.
(
2007
).
SOX9 is required for maintenance of the pancreatic progenitor cell pool
.
Proc. Natl. Acad. Sci. USA
104
,
1865
-
1870
.
Seymour
,
P. A.
,
Freude
,
K. K.
,
Dubois
,
C. L.
,
Shih
,
H.-P.
,
Patel
,
N. A.
and
Sander
,
M.
(
2008
).
A dosage-dependent requirement for Sox9 in pancreatic endocrine cell formation
.
Dev. Biol.
323
,
19
-
30
.
Seymour
,
P. A.
,
Shih
,
H. P.
,
Patel
,
N. A.
,
Freude
,
K. K.
,
Xie
,
R.
,
Lim
,
C. J.
and
Sander
,
M.
(
2012
).
A Sox9/Fgf feed-forward loop maintains pancreatic organ identity
.
Development
139
,
3363
-
3372
.
Shah
,
R. N. H.
,
Ibbitt
,
J. C.
,
Alitalo
,
K.
and
Hurst
,
H. C.
(
2002
).
FGFR4 overexpression in pancreatic cancer is mediated by an intronic enhancer activated by HNF1alpha
.
Oncogene
21
,
8251
-
8261
.
Shi
,
G.
,
Zhu
,
L.
,
Sun
,
Y.
,
Bettencourt
,
R.
,
Damsz
,
B.
,
Hruban
,
R. H.
and
Konieczny
,
S. F.
(
2009
).
Loss of the acinar-restricted transcription factor Mist1 accelerates Kras-induced pancreatic intraepithelial neoplasia
.
Gastroenterology
136
,
1368
-
1378
.
Shih
,
D. Q.
,
Navas
,
M. A.
,
Kuwajima
,
S.
,
Duncan
,
S. A.
and
Stoffel
,
M.
(
1999
).
Impaired glucose homeostasis and neonatal mortality in hepatocyte nuclear factor 3alpha-deficient mice
.
Proc. Natl. Acad. Sci. USA
96
,
10152
-
10157
.
Shih
,
H. P.
,
Kopp
,
J. L.
,
Sandhu
,
M.
,
Dubois
,
C. L.
,
Seymour
,
P. A.
,
Grapin-Botton
,
A.
and
Sander
,
M.
(
2012
).
A Notch-dependent molecular circuitry initiates pancreatic endocrine and ductal cell differentiation
.
Development
139
,
2488
-
2499
.
Solar
,
M.
,
Cardalda
,
C.
,
Houbracken
,
I.
,
Martín
,
M.
,
Maestro
,
M. A.
,
De Medts
,
N.
,
Xu
,
X.
,
Grau
,
V.
,
Heimberg
,
H.
,
Bouwens
,
L.
, et al. 
(
2009
).
Pancreatic exocrine duct cells give rise to insulin-producing beta cells during embryogenesis but not after birth
.
Dev. Cell
17
,
849
-
860
.
Stanger
,
B. Z.
,
Tanaka
,
A. J.
and
Melton
,
D. A.
(
2007
).
Organ size is limited by the number of embryonic progenitor cells in the pancreas but not the liver
.
Nature
445
,
886
-
891
.
Tao
,
B.
,
Bu
,
S.
,
Yang
,
Z.
,
Siroky
,
B.
,
Kappes
,
J. C.
,
Kispert
,
A.
and
Guay-Woodford
,
L. M.
(
2009
).
Cystin localizes to primary cilia via membrane microdomains and a targeting motif
.
J. Am. Soc. Nephrol.
20
,
2570
-
2580
.
Verdeguer
,
F.
,
Le Corre
,
S.
,
Fischer
,
E.
,
Callens
,
C.
,
Garbay
,
S.
,
Doyen
,
A.
,
Igarashi
,
P.
,
Terzi
,
F.
and
Pontoglio
,
M.
(
2010
).
A mitotic transcriptional switch in polycystic kidney disease
.
Nat. Med.
16
,
106
-
110
.
Ward
,
C. J.
,
Hogan
,
M. C.
,
Rossetti
,
S.
,
Walker
,
D.
,
Sneddon
,
T.
,
Wang
,
X.
,
Kubly
,
V.
,
Cunningham
,
J. M.
,
Bacallao
,
R.
,
Ishibashi
,
M.
, et al. 
(
2002
).
The gene mutated in autosomal recessive polycystic kidney disease encodes a large, receptor-like protein
.
Nat. Genet.
30
,
259
-
269
.
Ware
,
S. M.
,
Aygun
,
M. G.
and
Hildebrandt
,
F.
(
2011
).
Spectrum of clinical diseases caused by disorders of primary cilia
.
Proc. Am. Thorac. Soc.
8
,
444
-
450
.
Weinstein
,
M.
,
Xu
,
X.
,
Ohyama
,
K.
and
Deng
,
C. X.
(
1998
).
FGFR-3 and FGFR-4 function cooperatively to direct alveogenesis in the murine lung
.
Development
125
,
3615
-
3623
.
Wells
,
J. M.
,
Esni
,
F.
,
Boivin
,
G. P.
,
Aronow
,
B. J.
,
Stuart
,
W.
,
Combs
,
C.
,
Sklenka
,
A.
,
Leach
,
S. D.
and
Lowy
,
A. M.
(
2007
).
Wnt/beta-catenin signaling is required for development of the exocrine pancreas
.
BMC Dev. Biol.
7
,
4
.
Xuan
,
S.
,
Borok
,
M. J.
,
Decker
,
K. J.
,
Battle
,
M. A.
,
Duncan
,
S. A.
,
Hale
,
M. A.
,
Macdonald
,
R. J.
and
Sussel
,
L.
(
2012
).
Pancreas-specific deletion of mouse Gata4 and Gata6 causes pancreatic agenesis
.
J. Clin. Invest.
122
,
3516
-
3528
.
Yang
,
Y.
,
Chang
,
B. H.-J.
,
Yechoor
,
V.
,
Chen
,
W.
,
Li
,
L.
,
Tsai
,
M.-J.
and
Chan
,
L.
(
2011
).
The Krüppel-like zinc finger protein GLIS3 transactivates neurogenin 3 for proper fetal pancreatic islet differentiation in mice
.
Diabetologia
54
,
2595
-
2605
.
Ye
,
Y. W.
,
Zhou
,
Y.
,
Yuan
,
L.
,
Wang
,
C. M.
,
Du
,
C. Y.
,
Zhou
,
X. Y.
,
Zheng
,
B. Q.
,
Cao
,
X.
,
Sun
,
M. H.
,
Fu
,
H.
, et al. 
(
2011
).
Fibroblast growth factor receptor 4 regulates proliferation and antiapoptosis during gastric cancer progression
.
Cancer
117
,
5304
-
5313
.
Zhang
,
H.
,
Ables
,
E. T.
,
Pope
,
C. F.
,
Washington
,
M. K.
,
Hipkens
,
S.
,
Means
,
A. L.
,
Path
,
G.
,
Seufert
,
J.
,
Costa
,
R. H.
,
Leiter
,
A. B.
, et al. 
(
2009
).
Multiple, temporal-specific roles for HNF6 in pancreatic endocrine and ductal differentiation
.
Mech. Dev.
126
,
958
-
973
.
Zhu
,
L.
,
Tran
,
T.
,
Rukstalis
,
J. M.
,
Sun
,
P.
,
Damsz
,
B.
and
Konieczny
,
S. F.
(
2004
).
Inhibition of Mist1 homodimer formation induces pancreatic acinar-to-ductal metaplasia
.
Mol. Cell. Biol.
24
,
2673
-
2681
.

Competing interests

The authors declare no competing or financial interests.

Supplementary information