Many patients with chronic pancreatitis develop diabetes (chronic pancreatitis–related diabetes [CPRD]) through an undetermined mechanism. Here we used long-term partial pancreatic duct ligation (PDL) as a model to study CPRD. We found that long-term PDL induced significant β-cell dedifferentiation, followed by a time-dependent decrease in functional β-cell mass—all specifically in the ligated tail portion of the pancreas (PDL-tail). High levels of transforming growth factor β1 (TGFβ1) were detected in the PDL-tail and were mainly produced by M2 macrophages at the early stage and by activated myofibroblasts at the later stage. Loss of β-cell mass was then found to result from TGFβ1-triggered epithelial-mesenchymal transition (EMT) by β-cells, rather than resulting directly from β-cell apoptosis. Mechanistically, TGFβ1-treated β-cells activated expression of the EMT regulator gene Snail in a SMAD3/Stat3-dependent manner. Moreover, forced expression of forkhead box protein O1 (FoxO1), an antagonist for activated Stat3, specifically in β-cells ameliorated β-cell EMT and β-cell loss and prevented the onset of diabetes in mice undergoing PDL. Together, our data suggest that chronic pancreatitis may trigger TGFβ1-mediated β-cell EMT to lead to CPRD, which could substantially be prevented by sustained expression of FoxO1 in β-cells.

The prevalence of chronic pancreatitis is roughly 50 per 100,000 people worldwide (1). Chronic pancreatitis in the United States results in more than 122,000 outpatient visits and more than 56,000 hospitalizations each year (2). Many patients with chronic pancreatitis develop insulinopenia, glucose intolerance, insulin resistance, and eventually diabetes (2), largely as a result of the intimate proximity of the endocrine pancreas to the exocrine pancreas (3). Moreover, patients with chronic pancreatitis often develop a fibrotic pancreas with a reduced β-cell mass and have a 15- to 16-fold increased risk for pancreatic cancer (4). To date, the understanding of the development and pathogenesis of chronic pancreatitis–related diabetes (CPRD) is very limited. In addition, the mechanisms of β-cell loss in CPRD may in part be similar to those in type 2 diabetes (T2D) (5,6) and in cystic fibrosis (7). Thus, elucidation of the underlying mechanisms common to chronic pancreatitis, CPRD, and pancreatic cancer is critical.

Among animal models for acute and chronic pancreatitis (8), partial pancreatic duct ligation (PDL) has been used to generate chronic pancreatitis in mammals (9,10). Ligation of the pancreatic duct immediately at the beginning of the splenic or tail part of the pancreas blocks the drainage of ductal fluid from the distal pancreas, resulting in autodigestion of acinar cells and severe inflammation specifically in the ligated tail of the pancreas, although initially the islets are largely unaffected. Acinar cell death in the tail of the pancreas leads to the complete loss of acinar cells, without significant acinar cell regeneration (11). On the contrary, the nonligated part, or head, of the pancreas appears to be normal, thus offering an excellent internal control (12,13). We recently reported an inflammatory molecular signature in PDL, which induced β-cell proliferation in a transforming growth factor β (TGFβ) receptor signaling–dependent manner (1215). As a strong stimulant of epithelial-mesenchymal transition (EMT) in epithelial cells (1618), TGFβ1 is highly upregulated in the ligated tail of the pancreas after PDL (PDL-tail) (14). Thus we were prompted to evaluate the effects of PDL-induced TGFβ1 on the EMT of β-cells.

Forkhead box protein O1 (FoxO1) is a pivotal factor in orchestrating the response of β-cell mass and function to overnutrition and obesity (19) and to oxidative stress (2022). FoxO1 is predominantly expressed by β-cells in the adult pancreas. We and others have shown that FoxO1 nuclear translocation increases NeuroD1, MafA, and Nkx6.1 expression in β-cells, contributing to the maintenance of a functional differentiated phenotype to resist stress-induced dedifferentiation, dysfunction, and failure (2325). Nevertheless, a role for FoxO1 during the pathogenesis of CPRD is unknown.

Here we studied long-term PDL (12–24 weeks) as a model of chronic pancreatitis in humans. We further analyzed the molecular mechanisms underlying the gradual β-cell loss in this model, which mimics CPRD in humans.

Mouse Manipulation

All mouse experiments were approved by the Animal Research and Care Committee at the Children’s Hospital of Pittsburgh and the University of Pittsburgh Institutional Animal Care and Use Committee. BAC transgenic rat insulin promoter (RIP) Cre reporter (RIP-Cre) mice, MIP-GFP mice (green fluorescent protein reporter under the control of a mouse insulin promoter), and Rosa26CAG-mTmG (mTmG) mice have been described before (13). These mice and C57BL/6 mice were all purchased from The Jackson Laboratory (Bar Harbor, ME). TGFβ receptor II (TBR2) fx/fx mice were generous gifts from Professor Stefan Karlsson (University of Lund, Sweden) and have been described previously (12). All mice were 10-week-old males and had a C57BL/6 background. PDL was performed and validated as described elsewhere (1215). Intraductal viral infusion was performed as described previously (26). Adeno-associated virus (AAV) 8 viruses (titration of 1012 genome copy particles/mL in a 150-μL volume) were delivered via a catheter at a rate of 6 μL/min. BrdU-supplemented drinking water was given to mice 1 week before the analysis, as previously described (12).

Pancreatic Digestion, Islet Isolation, and FACS for β-Cells

Pancreas digestion, islet isolation, and β-cell purification from MIP-GFP mice have been described previously, taking advantage of the specific expression of green fluorescent protein on β-cells (1214,2729). Dissociated pancreatic cells were sorted based on α-smooth muscle actin (SMA) positivity after the cells were fixed in 4% formalin and then sequentially incubated with anti-α-SMA (Abcam, Cambridge, MA) and phycoerythrin-conjugated antirabbit antibody (BD Biosciences, San Jose, CA).

In Vitro Culture and Treatment of Mouse Islets

Mouse islets were cultured in Ham’s F10 medium (Life Technologies, St. Louis, MO) supplemented with 0.5% BSA (Sigma-Aldrich, St. Louis, MO), 2 mmol/L glutamine, 2 mmol/L calcium, and 5 mmol/L glucose at 37°C in 95% air and 5% CO2. After overnight culture, islets were either treated or not treated with 20 ng/mL TGFβ1 (with or without SMAD3 and Stat3 inhibitors) and harvested after 0, 0.5, 1, 3, 12, and 48 h. The SMAD3 inhibitor SIS3 (2 µmol/L) or Stat3 inhibitor cryptotanshinone (CTSN; 5 µmol/L) or control DMSO solution was added with the TGFβ1.

Western Blotting

Western blotting was performed as previously described (14,30). Primary antibodies for Western blotting are rabbit polyclonal anti-MafA (Abcam), anti-GAPDH, anti-Snail, anti-Slug, anti-Twist, anti-ZEB1, anti-ZEB2, anti-SMAD3, anti-pSMAD3, anti-Stat3, anti-pStat3, anti-NeuroD1, anti-Nkx6.1, anti-Pdx1, and anti–epithelial cadherin (E-cadherin), all from Cell Signaling Technologies, San Jose, CA). The secondary antibody was horseradish peroxidase–conjugated antirabbit (Dako, Carpinteria, CA).

Virus Production

AAV serotype 8 vectors were used to generate AAV-RIP-null and AAV-RIP-FoxO1 viruses, as described before (25).

Isolation of RNA and Quantitative RT-PCR

RNA extraction and quantitative RT-PCR have been described previously (12,13,27). The following primers all were purchased from Qiagen (Valencia, CA): CycloA (QT00247709), FoxO1 (QT00116186), Pdx1 (QT00102235), NeuroD1 (QT00251265), MafA (QT01037638), Nkx6.1 (QT00143318), TGFβ1 (QT00145250), vimentin (QT00159670), α-SMA (QT00140119), F4/80 (QT00099617), collagen I (QT00162204), E-cadherin (QT00121163), Snail (QT00240940), Slug (QT00098273), ZEB1 (QT00105385), ZEB2 (QT00148995), and fibronection (QT00135758). Quantitative RT-PCR values were normalized against CycloA, which proved to be stable across the samples. Fold changes from the control are shown in the figures.

Histology and Immunohistochemistry

All pancreas samples were fixed and cryoprotected in 30% sucrose overnight before freezing, as described before (12). Masson trichrome staining was performed using a Trichrome Stain (Masson) Kit (Sigma-Aldrich). The fluorescent membrane-targeted Tomato (mT) and membrane-targeted EGFP (mG) were detected by direct fluorescence. Primary antibodies for immunostaining were guinea pig polyclonal anti-insulin (Dako), rabbit polyclonal anti-F4/80 (Invitrogen, CA, Carlsbad), goat polyclonal antiamylase (Santa Cruz Biotechnology, Dallas, TX), rabbit polyclonal anti-FoxO1 (made by H.D.), goat polyclonal anti-E-cadherin (R&D Systems, Los Angeles, CA), rabbit polyclonal anti-α-SMA (Abcam), and rat polyclonal anti-CD45 (BD Biosciences). BrdU staining has been described before (12). Secondary antibodies for indirect fluorescent staining were Cy2, Cy3, or Cy5 conjugated rat-, rabbit- goat-, and guinea pig–specific antibodies (Jackson ImmunoResearch Labs, West Grove, PA). Nuclear staining was performed with Hoechst solution (BD Biosciences). β-Cell mass was quantified as described previously (12).

Data Analysis

All values are depicted as means ± standard errors. Five repeats were analyzed in each condition. All data were statistically analyzed using one-way ANOVA with a Bonferroni correction, followed by the Fisher exact test. Data were considered significant when P < 0.05.

Long-term PDL Is a Model of Chronic Pancreatitis

Recent studies of the effects of PDL on β-cell mass have primarily focused on the short-term (0–8 weeks). PDL can induce robust pancreatitis, as shown here in immunostaining for the pan-leukocyte marker CD45 (Fig. 1A) and the macrophage-specific marker F4/80 (Fig. 1B). Long-term PDL is a model of chronic pancreatitis. However, the effect of long-term PDL on β-cell mass has not been studied. Fasting blood glucose remained normal (Supplementary Fig. 1), presumably as a result of the head part of the pancreas remaining intact after PDL (PDL-head; Supplementary Fig. 2). Here, with gross imaging, we detected increasing exocrine tissue atrophy and fibrosis in the PDL-tail over the 12 to 24 weeks after PDL (Fig. 1C–F), which was confirmed by fibrosis quantified with Masson trichrome staining (Fig. 1G) and by quantification of transcript levels of fibrotic markers—collagen I, fibronectin, and vimentin—in the PDL-tail compared with the PDL-head (Fig. 1H). These findings are consistent with what has been seen in patients with chronic pancreatitis (2) and suggest that long-term PDL is a reasonable model of human chronic pancreatitis.

Figure 1

PDL is a model for both acute and chronic pancreatitis in humans. A and B: Immunostaining with CD45 (A) and F4/80 (B) in the PDL-tail 1 week after PDL (PDL1w). C–F: Gross images of the pancreas 1, 4 (PDL4w), 12 (PDL12w), and 24 (PDL24w) weeks after PDL. G: Quantification of pancreatic fibrosis in the PDL-tail and PDL-head based on the percentage of the area positively stained by Masson trichrome staining. H: Quantification of transcript levels of the fibrotic markers collagen I, fibronectin, and vimentin in the PDL-tail compared with the PDL-head through quantitative RT-PCR. Scale bars are 50 μm in A and B and 200 μm in C–F. Values were normalized to the sham-treated tail. *P < 0.05, PDL1w vs. sham; &P < 0.05, PDL4w vs. PDL1w; #P < 0.05, PDL12w vs. PDL4w (n = 5). HO, Hoechst nuclear staining; INS, insulin; NS, not significant.

Figure 1

PDL is a model for both acute and chronic pancreatitis in humans. A and B: Immunostaining with CD45 (A) and F4/80 (B) in the PDL-tail 1 week after PDL (PDL1w). C–F: Gross images of the pancreas 1, 4 (PDL4w), 12 (PDL12w), and 24 (PDL24w) weeks after PDL. G: Quantification of pancreatic fibrosis in the PDL-tail and PDL-head based on the percentage of the area positively stained by Masson trichrome staining. H: Quantification of transcript levels of the fibrotic markers collagen I, fibronectin, and vimentin in the PDL-tail compared with the PDL-head through quantitative RT-PCR. Scale bars are 50 μm in A and B and 200 μm in C–F. Values were normalized to the sham-treated tail. *P < 0.05, PDL1w vs. sham; &P < 0.05, PDL4w vs. PDL1w; #P < 0.05, PDL12w vs. PDL4w (n = 5). HO, Hoechst nuclear staining; INS, insulin; NS, not significant.

Close modal

Gradual β-Cell Loss Is Detected Over the Long-term After PDL

When we examined the morphology of the pancreas over time after PDL, we found that islets were easily visualized with the naked eye early after PDL as a result of elimination of acini, as shown grossly in Fig. 1C and D and in previous publications (12,13,31). Surprisingly, however, although acinar cells do not regenerate after PDL, islets were barely visible by 12 weeks after PDL (Fig. 1E) and were not grossly visible at all by 24 weeks after PDL (Fig. 1F). Further, insulin staining and β-cell mass quantification revealed a gradual β-cell loss, with a time-dependent decrease in β-cell mass, all specifically in the PDL-tail (Fig. 2A–K). Thus, β-cell loss occurs over the long-term after PDL.

Figure 2

Gradual β-cell loss is detected over the long-term after PDL. A–J: Representative images of immunohistochemistry for insulin (INS) by 3-3′-diaminobenzidine (A–E) and for INS (green) and amylase (AMY; red) by fluorescence (F–J). K: Quantification of β-cell mass in the PDL-tail at serial time points after PDL. L: PDL was performed in MIP-GFP mice and β-cells were isolated by flow cytometry based on green fluorescent protein at different time points after PDL in order to analyze the transcript levels of NeuroD1, Nkx6.1, Pdx1, and MafA using quantitative RT-PCR. Scale bars are 50 μm. Values were normalized to the sham-treated tail (n = 5). *P < 0.05, 1 week after PDL (PDL1w)/4 weeks after PDL (PDL4w) vs. sham; #P < 0.05, 12 weeks after PDL (PDL12w) vs. PDL1w/PDL4w; &P < 0.05, 24 weeks after PDL (PDL24w) vs. PDL12w. HO, Hoechst nuclear staining; NS, not significant.

Figure 2

Gradual β-cell loss is detected over the long-term after PDL. A–J: Representative images of immunohistochemistry for insulin (INS) by 3-3′-diaminobenzidine (A–E) and for INS (green) and amylase (AMY; red) by fluorescence (F–J). K: Quantification of β-cell mass in the PDL-tail at serial time points after PDL. L: PDL was performed in MIP-GFP mice and β-cells were isolated by flow cytometry based on green fluorescent protein at different time points after PDL in order to analyze the transcript levels of NeuroD1, Nkx6.1, Pdx1, and MafA using quantitative RT-PCR. Scale bars are 50 μm. Values were normalized to the sham-treated tail (n = 5). *P < 0.05, 1 week after PDL (PDL1w)/4 weeks after PDL (PDL4w) vs. sham; #P < 0.05, 12 weeks after PDL (PDL12w) vs. PDL1w/PDL4w; &P < 0.05, 24 weeks after PDL (PDL24w) vs. PDL12w. HO, Hoechst nuclear staining; NS, not significant.

Close modal

β-Cell Dedifferentiation Occurs Over the Long-term After PDL

Recent studies strongly suggest that protection of the differentiated phenotype of existing β-cells is critical to maintain a functional β-cell mass and thus to prevent T2D (23,24). In line with this concept, the β-cell–specific transcription factors Pdx1, NeuroD1, Nkx6.1, and MafA seem to be required for β-cells to be fully functional, whereas their loss correlates with β-cell dysfunction and β-cell loss (2325). As such, we performed PDL on MIP-GFP mice, and sorted β-cells via FACS at serial time points after PDL to asses Pdx1, NeuroD1, Nkx6.1, and MafA mRNA levels in β-cells by quantitative RT-PCR and Western blotting. We detected a gradual decrease in both mRNA and protein levels for these genes in β-cells (Fig. 2L and Supplementary Fig. 3), suggesting that β-cell dedifferentiation may occur in the tail of the pancreas over the long-term after PDL.

β-Cell Loss Over the Long-term After PDL Initially Results From β-Cell EMT Rather Than β-Cell Apoptosis

We then analyzed whether dedifferentiation of β-cells followed by β-cell apoptosis may be responsible for the progressive loss of β-cell mass. We performed both cleaved caspase 3 (Fig. 3A) and terminal deoxynucleotidyl TUNEL (Fig. 3B) staining to evaluate β-cell apoptosis. To our surprise, no significant increase in β-cell apoptosis was detected by either method at different time points after PDL (Fig. 3A and B), suggesting that β-cell loss over the long-term after PDL is not primarily attributable to β-cell apoptosis.

Figure 3

β-Cell loss over the long-term after PDL results from β-cell EMT rather than β-cell apoptosis. A and B: Quantification of the percentage of caspase3-positive (A) and TUNEL-positive (B) β-cells at different time points after PDL. C: PDL was performed in MIP-GFP mice and β-cells were sorted to analyze transcript levels for E-cadherin, ZEB1, ZEB2, Snail, and Slug using quantitative RT-PCR. D: PDL was performed in RIP-Cre;mTmG mice; a representative confocal image at 12 weeks after PDL is shown. Yellow arrows point to mG-positive/vimentin-positive/insulin (INS)-negative cells, also shown in high magnification. mT and insulin appear red; mG, green; and vimentin, blue. Values were normalized to the sham-treated tail (n = 5). Scale bar is 50 μm. #P < 0.05, 12 weeks after PDL (PDL12w) vs. 4 weeks after PDL (PDL4w). NS, nonsignificant; PDL1w, 1 week after PDL; PDL24w, 24 weeks after PDL.

Figure 3

β-Cell loss over the long-term after PDL results from β-cell EMT rather than β-cell apoptosis. A and B: Quantification of the percentage of caspase3-positive (A) and TUNEL-positive (B) β-cells at different time points after PDL. C: PDL was performed in MIP-GFP mice and β-cells were sorted to analyze transcript levels for E-cadherin, ZEB1, ZEB2, Snail, and Slug using quantitative RT-PCR. D: PDL was performed in RIP-Cre;mTmG mice; a representative confocal image at 12 weeks after PDL is shown. Yellow arrows point to mG-positive/vimentin-positive/insulin (INS)-negative cells, also shown in high magnification. mT and insulin appear red; mG, green; and vimentin, blue. Values were normalized to the sham-treated tail (n = 5). Scale bar is 50 μm. #P < 0.05, 12 weeks after PDL (PDL12w) vs. 4 weeks after PDL (PDL4w). NS, nonsignificant; PDL1w, 1 week after PDL; PDL24w, 24 weeks after PDL.

Close modal

We previously showed that large amounts of TGFβ1 were secreted by M2 macrophages in the PDL-tail, which led to SMAD7 expression and TGFβ receptor signaling in β-cells to coordinate a modest increase in β-cell proliferation. These actions occurred in conjunction with simultaneously activated epidermal growth factor receptor signaling (14). TGFβ1 is also a potent and well-defined trigger for EMT, a process by which epithelial cells lose their cell polarity and cell-cell adhesion and gain migratory and invasive properties to become mesenchyme-like cells (1618). Because we detected high TGFβ1 levels (14) and progressive fibrosis in the PDL-tail (Fig. 1A–G), we hypothesized that TGFβ1 may also trigger EMT of β-cells. Loss of E-cadherin is well established as a fundamental event in EMT, during which the transcription factors ZEB1, ZEB2, Snail, and Slug bind directly to E-box consensus sequences on the promoter region of E-cadherin and repress its expression (1618). Thus we analyzed expression levels of these EMT-related genes in purified β-cells from the tails of pancreases of MIP-GFP mice after PDL. We found that ZEB1 and Snail protein levels in β-cells increased with time, whereas E-cadherin levels decreased with time. In addition, ZEB2 and Slug mRNA (but not protein) increased with time (Fig. 3C and Supplementary Fig. 3). These results suggest that an EMT process may indeed occur in β-cells over time after PDL.

To further confirm this possibility, we performed a lineage tracing study using our previously published RIP-Cre;mTmG mouse model (13). These mice should express membrane-bound red fluorescence in all cells except insulin-positive β-cells and their progeny, whose floxed mT cassette is deleted, leading to constitutive expression of the mG cassette located just downstream. We noticed that fibroblasts tend to not express the mT well, and therefore many of the cells in a fibrotic pancreas do not fluoresce. In these mice, if any β-cells undergo dedifferentiation to become insulin-negative, they still keep mG expression and appear green. We detected quite a few mG-positive/vimentin-positive/insulin-negative cells 12 weeks after PDL; these cells likely represent β-cells that have undergone EMT (Fig. 3D). These data strongly suggest the occurrence of a β-cell EMT, which may be a critical contributor to β-cell loss over the long-term after PDL.

TGFβ1 Is a Necessary Trigger of β-Cell EMT After PDL

We previously reported that PDL entails recruitment of M2 macrophages that secrete large amounts of TGFβ1 (14). However, macrophage infiltration in the PDL-tail peaked 1 week after PDL and then subsequently decreased, as shown previously (1214), and as shown here by F4/80 transcript enrichment in the PDL-tail (Fig. 4A). Because tissue fibrosis and β-cell EMT seem to occur over the long-term after PDL, we hypothesized that either TGFβ1 levels remain high over the long-term after PDL because the TGFβ1 comes from other sources, or non-TGFβ1 EMT triggers may be involved. We analyzed the transcript levels of TGFβ1 in the PDL-tail over time and found that they remained high, peaking ∼4 weeks after PDL, when F4/80 transcript levels were already significantly decreased (Fig. 4A). These data suggest that TGFβ1 may be the trigger of tissue fibrosis and β-cell EMT but that macrophages may not be the major source of TGFβ1 at these later time points after PDL (14). Indeed, previous studies have highlighted a specific cell population called myofibroblasts, also called pancreatic stellate cells (32), which, when activated, produce high levels of TGFβ1 to promote EMT and fibrosis (1618). Because α-SMA is a specific marker for myofibroblasts, we examined the transcript levels of α-SMA in the PDL-tail and found a significant and sustained increase in α-SMA by 4 weeks after PDL (Fig. 4A); this was further confirmed by immunostaining (Fig. 4B). Moreover, when we sorted α-SMA–negative and α-SMA–positive cells from the PDL-tail, we detected significantly higher levels of TGFβ1 in the α-SMA–negative population than in the α-SMA–positive population 1 week after PDL, but significantly higher levels of TGFβ1 in the α-SMA–positive population than in the α-SMA–negative population 4 weeks after PDL (Fig. 4C). This indicates that activated myofibroblasts may be the major source of TGFβ1 over the long-term after PDL and may play a more important role than macrophages in pancreatic tissue remodeling, fibrosis, and β-cell EMT.

Figure 4

TGFβ1 is secreted by M2 macrophages and activated myofibroblasts in the PDL-tail. A: Quantification of transcript levels of F4/80, TGFβ1, and α-SMA in the PDL-tail using quantitative RT-PCR. Values were normalized to the sham-treated tail (n = 5). B: Representative images of immunostaining for α-SMA in the PDL-tail. C: Quantitative RT-PCR for TGFβ1 in sorted α-SMA–negative and α-SMA–positive cells from the PDL-tail. Values were normalized to α-SMA–negative cells from the tail of the pancreas 1 week after PDL (PDL1w). Scale bar is 50 μm. *P < 0.05, PDL1w vs. sham; &P < 0.05, 4 weeks after PDL (PDL4w) vs. PDL1w; #P < 0.05, 12 weeks after PDL (PDL12w) vs. PDL4w. INS, insulin; PDL24w, 24 weeks after PDL.

Figure 4

TGFβ1 is secreted by M2 macrophages and activated myofibroblasts in the PDL-tail. A: Quantification of transcript levels of F4/80, TGFβ1, and α-SMA in the PDL-tail using quantitative RT-PCR. Values were normalized to the sham-treated tail (n = 5). B: Representative images of immunostaining for α-SMA in the PDL-tail. C: Quantitative RT-PCR for TGFβ1 in sorted α-SMA–negative and α-SMA–positive cells from the PDL-tail. Values were normalized to α-SMA–negative cells from the tail of the pancreas 1 week after PDL (PDL1w). Scale bar is 50 μm. *P < 0.05, PDL1w vs. sham; &P < 0.05, 4 weeks after PDL (PDL4w) vs. PDL1w; #P < 0.05, 12 weeks after PDL (PDL12w) vs. PDL4w. INS, insulin; PDL24w, 24 weeks after PDL.

Close modal

To confirm TGFβ1 as a necessary trigger for β-cell EMT in this model, we performed PDL on β-cell–specific TBR2-null mutant mice (RIP-Cre; TBR2fx/fx) (12,13) (Fig. 5A). While β-cell–specific suppression of TGFβ1-activated TGFβ receptor signaling did not alter pancreas fibrosis in PDL-tails (Fig. 5B), the gradual loss of β-cell mass over the long-term after PDL was significantly reduced (Fig. 5C and D). These data strongly suggest that TGFβ1 is a necessary trigger for β-cell EMT and β-cell loss over the long-term after PDL.

Figure 5

TGFβ1 is a necessary trigger of β-cell EMT in the PDL-tail. A: Schematic of a β-cell–specific TBR2-null mutant mouse. B: Quantification of fibrosis in the PDL-tail based on the area positively stained with Masson trichrome staining; RIP-Cre;TBR2fx/fx and control TBR2fx/fx mice were used. C and D: Quantification (C) and representative images (D) of β-cell mass in the PDL-tail using RIP-Cre;TBR2fx/fx and control TBR2fx/fx mice. Values were normalized to the sham-treated tail (n = 5). Scale bars are 50 μm. *P < 0.05. HO, Hoechst nuclear staining; INS, insulin; NS, nonsignificant; PDL1w, 1 week after PDL; PDL4w, 4 weeks after PDL; PDL12w, 12 weeks after PDL; PDL24w, 24 weeks after PDL.

Figure 5

TGFβ1 is a necessary trigger of β-cell EMT in the PDL-tail. A: Schematic of a β-cell–specific TBR2-null mutant mouse. B: Quantification of fibrosis in the PDL-tail based on the area positively stained with Masson trichrome staining; RIP-Cre;TBR2fx/fx and control TBR2fx/fx mice were used. C and D: Quantification (C) and representative images (D) of β-cell mass in the PDL-tail using RIP-Cre;TBR2fx/fx and control TBR2fx/fx mice. Values were normalized to the sham-treated tail (n = 5). Scale bars are 50 μm. *P < 0.05. HO, Hoechst nuclear staining; INS, insulin; NS, nonsignificant; PDL1w, 1 week after PDL; PDL4w, 4 weeks after PDL; PDL12w, 12 weeks after PDL; PDL24w, 24 weeks after PDL.

Close modal

TGFβ1 Triggers EMT of β-Cells Through a SMAD3/Stat3 Signaling Cascade

To understand the molecular basis underlying TGFβ1-activated β-cell EMT, we treated cultured primary mouse islets with 20 ng/mL TGFβ1 and harvested the cells at 0, 0.5, 1, 3, 12, 24, and 48 h after TGFβ1 administration. Western blotting was performed to examine the EMT regulatory proteins Twist, Snail, Slug, ZEB1, and ZEB2; among these, Snail was the only one found to be strongly activated (Fig. 6A). Moreover, Snail was activated as early as 1 h after TGFβ1 treatment, and Snail expression seemed to be sustained (Fig. 6A). In addition, Snail mRNA was activated in a pattern similar to that of the protein (Fig. 6B), suggesting that Snail may be activated primarily at the transcription level. Thus we focused on the regulation of Snail by TGFβ signaling. SMAD3 is a key factor in mediating signal transduction of activated TGFβ receptors. Activated TGFβ/SMAD3 has been shown to induce phosphorylation and activation of Stat3 to promote the transcription of Snail (33). TGFβ1-treated mouse islets were cotreated with either a specific SMAD3 phosphorylation inhibitor (SIS3) or a specific Stat3 phosphorylation (Tyr705) inhibitor (CTSN) in order to suppress phosphorylation-associated activation of SMAD3 and Stat3, respectively. Islets were analyzed 12 h after treatment. We found that TGFβ1 induced phosphorylation of SMAD3 and Stat3 and activation of Snail (Fig. 6C–F). Suppression of SMAD3 phosphorylation by SIS3 prevented both TGFβ1-induced phosphorylation of Stat3 and TGFβ1-induced activation of Snail, but suppression of Stat3 phosphorylation by CTSN prevented only TGFβ1-induced activation of Snail, without affecting TGFβ1-induced phosphorylation of SMAD3 (Fig. 6C–F). Thus TGFβ1 may trigger EMT of β-cells through a SMAD3/Stat3 signaling cascade, and SMAD3 phosphorylation occurs upstream of Stat3 phosphorylation, consistent with a previous report (33).

Figure 6

TGFβ1 triggers EMT of β-cells through a SMAD3/Stat3 signaling cascade. A and B: Cultured primary mouse islets were treated with 20 ng/mL TGFβ1 and harvested at 0, 0.5, 1, 3, 12, 24, and 48 h after TGFβ1 administration. A: Representative Western blotting for the EMT regulatory proteins Twist, Snail, Slug, ZEB1, and ZEB2. B: Results of quantitative RT-PCR for Snail. C–F: Cultured primary mouse islets were treated with 20 ng/mL TGFβ1 alone or with 2 µmol/L SIS3 and 5 µmol/L CTSN and harvested 12 h after TGFβ1 administration. C: Representative Western blotting for SMAD3, pSMAD3, Stat3, pStat3, and Snail. D–F: Quantification of the pSMAD3-to-SMAD3 ratio (D), the pStat3-to-Stat3 ratio (E), and the Snail-to-GAPDH ratio (F). *P < 0.05. N = 5 mice for each experimental group. NS, nonsignificant; und, undetected.

Figure 6

TGFβ1 triggers EMT of β-cells through a SMAD3/Stat3 signaling cascade. A and B: Cultured primary mouse islets were treated with 20 ng/mL TGFβ1 and harvested at 0, 0.5, 1, 3, 12, 24, and 48 h after TGFβ1 administration. A: Representative Western blotting for the EMT regulatory proteins Twist, Snail, Slug, ZEB1, and ZEB2. B: Results of quantitative RT-PCR for Snail. C–F: Cultured primary mouse islets were treated with 20 ng/mL TGFβ1 alone or with 2 µmol/L SIS3 and 5 µmol/L CTSN and harvested 12 h after TGFβ1 administration. C: Representative Western blotting for SMAD3, pSMAD3, Stat3, pStat3, and Snail. D–F: Quantification of the pSMAD3-to-SMAD3 ratio (D), the pStat3-to-Stat3 ratio (E), and the Snail-to-GAPDH ratio (F). *P < 0.05. N = 5 mice for each experimental group. NS, nonsignificant; und, undetected.

Close modal

FoxO1 Prevents EMT of β-Cells Over the Long-term After PDL

FoxO1 is a transcription factor that plays a key protective role against β-cell failure during stress (2325). Previous reports have shown that FoxO1 and Stat3 antagonize each other in the regulation of T-cell function (34,35) and leptin signaling transduction (36,37). We were thus prompted to evaluate the role of FoxO1 during the process of β-cell EMT in this CPRD model, because the inflammatory environment in the PDL-tail presumably induces significant local cell stress. We performed immunostaining for FoxO1 and, compared with the PDL-head and the sham-operated control, found enhanced expression and nuclear localization of FoxO1 in β-cells in the PDL-tail 4 weeks after PDL, suggesting that FoxO1 may be activated during β-cell stress after PDL (Fig. 7A). However, FoxO1 was absent from β-cells 12 weeks after PDL (Fig. 7A). To document a protective role for FoxO1 against EMT of β-cells, we forcibly expressed FoxO1 specifically in β-cells using an intraductal viral infusion technique recently developed in our laboratory (26) (Fig. 7B). Here, forced expression of FoxO1 in β-cells by intraductal infusion of AAV carrying recombinant FoxO1 under RIP in mice undergoing PDL prevented FoxO1 loss in islets after PDL (Fig. 7C) and significantly ameliorated β-cell loss over time (Fig. 7D). Moreover, forced expression of FoxO1 by AAV-mediated gene transfer in purified β-cells from tails of pancreases 12 weeks after PDL inhibited activation of EMT genes and increased E-cadherin (Fig. 7E). Together, these data suggest that FoxO1 may prevent β-cell EMT over the long-term after PDL (Fig. 7F).

Figure 7

FoxO1 seems to play a pivotal role in preventing β-cell EMT after PDL. A: Representative images of immunostaining for FoxO1 in the PDL-tail 4 weeks (PDL4w) and 12 weeks (PDL12w) after PDL. B: Schematic of the experiment. AAV carrying either RIP-FoxO1 or RIP-null as a control was intraductally infused immediately before PDL, and β-cell mass was then analyzed at 12 and 24 weeks (PDL24w) after PDL. C: FoxO1 mRNA in mouse islets. Values were normalized to the sham-treated tail at week 0. D: Analysis of β-cell mass showed that expression of FoxO1 specifically in β-cells, forced by intraductal viral infusion, significantly reduced β-cell loss over the long-term after PDL. E: β-Cells were isolated from the PDL-tail of MIP-GFP mice at PDL12w and placed in culture. These β-cells were transduced with either AAV-RIP-FoxO1 or AAV-RIP-null as a control. After infection (48 h), the β-cells were analyzed for expression of E-cadherin, Snail, Slug, ZEB1, and ZEB2. Values were normalized to the AAV-RIP-null treated control. F: Proposed signaling cascade in CPRD. High levels of TGFβ1 in the inflamed pancreas are produced by M2 macrophages early and by activated myofibroblasts at later stages. TGFβ1-activated TGFβ receptor (TGFβR) signaling in β-cells, in which phosphorylation and activation of SMAD3 induce phosphorylation and activation of Stat3 to activate transcription of Snail, leads to EMT in β-cells, resulting in β-cell dysfunction and loss (CPRD). FoxO1 and Stat3 antagonize one another, so early activation of Stat3 overcomes FoxO1 suppression, leading to β-cell dysfunction. However, strengthening FoxO1 expression in β-cells suppresses Stat3 signaling and thereby suppresses β-cell EMT. Scale bars are 50 μm. *P < 0.05. HO, Hoechst nuclear staining; INS, insulin; NS, nonsignificant. N = 5 mice for each experimental group.

Figure 7

FoxO1 seems to play a pivotal role in preventing β-cell EMT after PDL. A: Representative images of immunostaining for FoxO1 in the PDL-tail 4 weeks (PDL4w) and 12 weeks (PDL12w) after PDL. B: Schematic of the experiment. AAV carrying either RIP-FoxO1 or RIP-null as a control was intraductally infused immediately before PDL, and β-cell mass was then analyzed at 12 and 24 weeks (PDL24w) after PDL. C: FoxO1 mRNA in mouse islets. Values were normalized to the sham-treated tail at week 0. D: Analysis of β-cell mass showed that expression of FoxO1 specifically in β-cells, forced by intraductal viral infusion, significantly reduced β-cell loss over the long-term after PDL. E: β-Cells were isolated from the PDL-tail of MIP-GFP mice at PDL12w and placed in culture. These β-cells were transduced with either AAV-RIP-FoxO1 or AAV-RIP-null as a control. After infection (48 h), the β-cells were analyzed for expression of E-cadherin, Snail, Slug, ZEB1, and ZEB2. Values were normalized to the AAV-RIP-null treated control. F: Proposed signaling cascade in CPRD. High levels of TGFβ1 in the inflamed pancreas are produced by M2 macrophages early and by activated myofibroblasts at later stages. TGFβ1-activated TGFβ receptor (TGFβR) signaling in β-cells, in which phosphorylation and activation of SMAD3 induce phosphorylation and activation of Stat3 to activate transcription of Snail, leads to EMT in β-cells, resulting in β-cell dysfunction and loss (CPRD). FoxO1 and Stat3 antagonize one another, so early activation of Stat3 overcomes FoxO1 suppression, leading to β-cell dysfunction. However, strengthening FoxO1 expression in β-cells suppresses Stat3 signaling and thereby suppresses β-cell EMT. Scale bars are 50 μm. *P < 0.05. HO, Hoechst nuclear staining; INS, insulin; NS, nonsignificant. N = 5 mice for each experimental group.

Close modal

Chronic pancreatitis is highly prevalent (1,2) and predisposes patients to a high risk of developing diabetes (CPRD) (5). The current treatment of chronic pancreatitis includes surgical removal of part or all of the pancreas, with the potential to isolate and transplant the islets back into the patient. Many patients already have diabetes, however, and therefore are not candidates for this islet salvage. Also, islet isolation from a pancreas with chronic pancreatitis characterized by severe tissue fibrosis is difficult and inefficient. Moreover, transplanted islets are situated in the liver, where they may gradually lose function (1,2). Therefore a better understanding of the molecular mechanisms underlying the development and pathogenesis of CPRD may lead to novel, more effective therapeutic strategies.

The development of CPRD may largely result from the proximity of the endocrine pancreas to the exocrine pancreas. When the exocrine pancreas is severely inflamed and fibrotic, the inflammatory milieu results in the presence of many cytokines and factors that negatively affect the neighboring islet cells in a paracrine way. In addition, the vasculature is typically severely disrupted. These alterations may significantly affect the biology and homeostasis of islet β-cells.

Complete PDL and partial PDL have been used for decades as models for acute and chronic pancreatitis in humans (9,10). Compared with complete PDL, the partial PDL model has several advantages. The nonligated head of the pancreas (PDL-head) is minimally affected and thus can be used as an internal control for the PDL-tail. Also, the islets and acinar cells in the head of the pancreas are adequate to meet physiological needs for glucose homeostasis and secretion of pancreatic digestive enzymes, respectively. The disadvantage of the partial PDL model is that the intact head of the pancreas prevents changes in blood glucose at the late stage of CPRD. Since 2008, PDL over the short-term—traditionally regarded as a model of acute pancreatitis—has been extensively investigated as a model of β-cell regeneration. Although controversy still exists, most of these short-term studies have shown that little or no β-cell neogenesis occurs, but β-cell proliferation increases modestly, which contributes minimally to β-cell mass (13,31,3840).

Compared with other chronic pancreatitis models (e.g., serial cerulein injections, serial l-arginine injections, alcohol feeding, and genetic modifications), PDL confers several advantages. First, PDL requires only one treatment, which substantially reduces stress to the animals and reduces experimental variability. Second, PDL induces robust pancreatitis through ductal occlusion, which may mimic the pathogenesis of pancreatitis in many humans. Third, acute pancreatitis by PDL progresses to chronic pancreatitis relatively quickly, which not only significantly shortens the experimental period but also allows easier detection of changes in the parameters of interest (911,4143).

In this study we show that long-term PDL becomes a chronic pancreatitis model, and the gradual loss of β-cell mass after PDL mimics the similar phenomenon in patients with CPRD. We found that β-cell dedifferentiation followed by β-EMT occurs in the PDL-tail, resulting in a progressive loss of functional β-cell mass. In the lineage tracing experiment, no vimentin/insulin double-positive cells were ever detected. However, vimentin-positive β-cell progeny cells were broadly detected by 12 weeks after PDL, suggesting widespread β-cell EMT. Of note, EMT β-cells may then undergo cell death, since the fold-increase in EMT-associated genes was not dramatic in purified β-cells at serial time points after PDL.

Using a loss-of-function experiment in which TGFβ receptor signaling was blocked (TBR2-null mutant mice), we confirmed that TGFβ receptor signaling was necessary for β-cell EMT in the PDL-tail. We also showed evidence that TGFβ1 ligand is initially derived predominantly from M2 macrophages soon after PDL, but it comes from activated myofibroblasts in the later stages after PDL. Myofibroblasts can be activated by TGFβ1, but as a result they begin to produce and secrete TGFβ1 themselves, as a positive feedback loop for TGFβ1 production (1618). Early after PDL, the recruited macrophages may act as activators of myofibroblasts through their production and secretion of TGFβ1 (14). Furthermore, high TGFβ1 has been detected in the pancreas in patients with acute (44) and chronic pancreatitis (45). Thus a critical role for TGFβ1 in β-cell EMT over the long-term after PDL seems a likely mechanism of CPRD pathogenesis. Mechanistically, we showed that TGFβ1 activated TGFβ receptor signaling in β-cells to induce phosphorylation and activation of SMAD3, which subsequently induced activation of Stat3, possibly through competitive binding to protein inhibitor of activated Stat3 (33).

FoxO1 and Stat3 are known to antagonize one another in several cell types, but this has not been studied in β-cells. However, recent studies have demonstrated a critical role for Stat3 in pancreatic cell homeostasis (46,47). Here, early activation of Stat3 overcomes FoxO1 suppression, leading to β-cell dysfunction. Strengthening FoxO1 expression in β-cells suppresses Stat3 signaling and thereby suppresses β-cell EMT. In our mouse model, we further showed that FoxO1 overexpression is sufficient to prevent β-cell EMT, and the late loss of FoxO1 seemed to portend the completion of β-cell EMT.

The relatively delayed onset of β-cell EMT in vivo, compared with in vitro, possibly results from the in vivo environment of the islets that might protect against EMT through cell-cell communication. In summary, we present here a mouse model for CPRD. To the best of our knowledge, this study is the first to show β-cell EMT in an experimental setting of CPRD. Because β-cell EMT seems to underlie the process of β-cell loss, prevention of β-cell EMT may be an effective treatment for CPRD.

Funding. This work was supported by the startup from the Department of Surgery of Children’s Hospital of Pittsburgh (to X.X.), and by the National Institutes of Health/National Institute of Diabetes and Digestive and Kidney Diseases (grant nos. R01DK111460 and R01DK112836 to G.K.G.).

Duality of Interest. No conflicts of interest relevant to this article were reported.

Author Contributions. X.X. conceived and designed the study. X.X., S.F., T.Z., C.C., Q.S., R.Z., S.P., J.F., Y.M., P.G., C.S., K.P., and N.G. acquired data. X.X., S.Z.H., H.D., and G.K.G. analyzed and interpreted the data. X.X. wrote the manuscript. All authors revised the article and approved the final version to be published. X.X. and G.K.G. are the guarantors of this work and, as such, had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.

Prior Presentation. Part of this study was presented orally (no. 2015-A-1495-Diabetes) at the 75th Scientific Sessions of the American Diabetes Association, Boston, MA, 5–9 June 2015.

1.
Yadav
D
,
Timmons
L
,
Benson
JT
,
Dierkhising
RA
,
Chari
ST
.
Incidence, prevalence, and survival of chronic pancreatitis: a population-based study
.
Am J Gastroenterol
2011
;
106
:
2192
2199
[PubMed]
2.
DiMagno
MJ
,
DiMagno
EP
.
Chronic pancreatitis
.
Curr Opin Gastroenterol
2013
;
29
:
531
536
[PubMed]
3.
Pasca di Magliano
M
,
Forsmark
C
,
Freedman
S
, et al
.
Advances in acute and chronic pancreatitis: from development to inflammation and repair
.
Gastroenterology
2013
;
144
:
e1
e4
[PubMed]
4.
Lowenfels
AB
,
Maisonneuve
P
,
Cavallini
G
, et al.;
International Pancreatitis Study Group
.
Pancreatitis and the risk of pancreatic cancer
.
N Engl J Med
1993
;
328
:
1433
1437
[PubMed]
5.
Pan
J
,
Xin
L
,
Wang
D
, et al
.
Risk factors for diabetes mellitus in chronic pancreatitis: a cohort of 2,011 patients
.
Medicine (Baltimore)
2016
;
95
:
e3251
[PubMed]
6.
Brodovicz
KG
,
Kou
TD
,
Alexander
CM
, et al
.
Impact of diabetes duration and chronic pancreatitis on the association between type 2 diabetes and pancreatic cancer risk
.
Diabetes Obes Metab
2012
;
14
:
1123
1128
[PubMed]
7.
Ode
KL
,
Moran
A
.
New insights into cystic fibrosis-related diabetes in children
.
Lancet Diabetes Endocrinol
2013
;
1
:
52
58
[PubMed]
8.
Lerch
MM
,
Gorelick
FS
.
Models of acute and chronic pancreatitis
.
Gastroenterology
2013
;
144
:
1180
1193
[PubMed]
9.
Tanaka
T
,
Ichiba
Y
,
Fujii
Y
,
Itoh
H
,
Kodama
O
,
Dohi
K
.
New canine model of chronic pancreatitis due to chronic ischemia with incomplete pancreatic duct obstruction
.
Digestion
1988
;
41
:
149
155
[PubMed]
10.
Boerma
D
,
Straatsburg
IH
,
Offerhaus
GJ
,
Gouma
DJ
,
van Gulik
TM
.
Experimental model of obstructive, chronic pancreatitis in pigs
.
Dig Surg
2003
;
20
:
520
526
[PubMed]
11.
Yamaguchi
Y
,
Matsuno
K
,
Goto
M
,
Ogawa
M
.
In situ kinetics of acinar, duct, and inflammatory cells in duct ligation-induced pancreatitis in rats
.
Gastroenterology
1993
;
104
:
1498
1506
[PubMed]
12.
Xiao
X
,
Wiersch
J
,
El-Gohary
Y
, et al
.
TGFβ receptor signaling is essential for inflammation-induced but not β-cell workload-induced β-cell proliferation
.
Diabetes
2013
;
62
:
1217
1226
[PubMed]
13.
Xiao
X
,
Chen
Z
,
Shiota
C
, et al
.
No evidence for β cell neogenesis in murine adult pancreas
.
J Clin Invest
2013
;
123
:
2207
2217
[PubMed]
14.
Xiao
X
,
Gaffar
I
,
Guo
P
, et al
.
M2 macrophages promote beta-cell proliferation by up-regulation of SMAD7
.
Proc Natl Acad Sci U S A
2014
;
111
:
E1211
E1220
[PubMed]
15.
Xiao
X
,
Guo
P
,
Shiota
C
, et al
.
Neurogenin3 activation is not sufficient to direct duct-to-beta cell transdifferentiation in the adult pancreas
.
J Biol Chem
2013
;
288
:
25297
25308
[PubMed]
16.
Chaudhury
A
,
Hussey
GS
,
Ray
PS
,
Jin
G
,
Fox
PL
,
Howe
PH
.
TGF-beta-mediated phosphorylation of hnRNP E1 induces EMT via transcript-selective translational induction of Dab2 and ILEI
.
Nat Cell Biol
2010
;
12
:
286
293
[PubMed]
17.
Park
SH
,
Jung
EH
,
Kim
GY
,
Kim
BC
,
Lim
JH
,
Woo
CH
.
Itch E3 ubiquitin ligase positively regulates TGF-β signaling to EMT via Smad7 ubiquitination
.
Mol Cells
2015
;
38
:
20
25
[PubMed]
18.
Song
J
.
EMT or apoptosis: a decision for TGF-beta
.
Cell Res
2007
;
17
:
289
290
[PubMed]
19.
Zhang
T
,
Kim
DH
,
Xiao
X
, et al
.
FoxO1 plays an important role in regulating β-cell compensation for insulin resistance in male mice
.
Endocrinology
2016
;
157
:
1055
1070
[PubMed]
20.
Kitamura
T
,
Ido Kitamura
Y
.
Role of FoxO proteins in pancreatic beta cells
.
Endocr J
2007
;
54
:
507
515
[PubMed]
21.
Mezza
T
,
Shirakawa
J
,
Martinez
R
,
Hu
J
,
Giaccari
A
,
Kulkarni
RN
.
Nuclear export of FoxO1 is associated with ERK signaling in β-cells lacking insulin receptors
.
J Biol Chem
2016
;
291
:
21485
21495
[PubMed]
22.
Gannon
M
.
BuMP-ing up insulin secretion by pancreatic beta cells
.
Cell Metab
2007
;
5
:
157
159
[PubMed]
23.
Guo
S
,
Dai
C
,
Guo
M
, et al
.
Inactivation of specific β cell transcription factors in type 2 diabetes
.
J Clin Invest
2013
;
123
:
3305
3316
[PubMed]
24.
Talchai
C
,
Xuan
S
,
Lin
HV
,
Sussel
L
,
Accili
D
.
Pancreatic β cell dedifferentiation as a mechanism of diabetic β cell failure
.
Cell
2012
;
150
:
1223
1234
[PubMed]
25.
Xiao
X
,
Chen
C
,
Guo
P
, et al
.
Forkhead box protein 1 (FoxO1) inhibits accelerated β cell aging in pancreas-specific SMAD7 mutant mice
.
J Biol Chem
2017
;
292
:
3456
3465
[PubMed]
26.
Xiao
X
,
Guo
P
,
Prasadan
K
, et al
.
Pancreatic cell tracing, lineage tagging and targeted genetic manipulations in multiple cell types using pancreatic ductal infusion of adeno-associated viral vectors and/or cell-tagging dyes
.
Nat Protoc
2014
;
9
:
2719
2724
[PubMed]
27.
Xiao
X
,
Guo
P
,
Chen
Z
, et al
.
Hypoglycemia reduces vascular endothelial growth factor A production by pancreatic beta cells as a regulator of beta cell mass
.
J Biol Chem
2013
;
288
:
8636
8646
[PubMed]
28.
Xiao
X
,
Prasadan
K
,
Guo
P
, et al
.
Pancreatic duct cells as a source of VEGF in mice
.
Diabetologia
2014
;
57
:
991
1000
[PubMed]
29.
Song
Z
,
Fusco
J
,
Zimmerman
R
, et al
.
Epidermal growth factor receptor signaling regulates β cell proliferation in adult mice
.
J Biol Chem
2016
;
291
:
22630
22637
[PubMed]
30.
Xiao
X
,
Fischbach
S
,
Song
Z
, et al
.
Transient suppression of TGFβ receptor signaling facilitates human islet transplantation
.
Endocrinology
2016
;
157
:
1348
1356
[PubMed]
31.
Pan
FC
,
Bankaitis
ED
,
Boyer
D
, et al
.
Spatiotemporal patterns of multipotentiality in Ptf1a-expressing cells during pancreas organogenesis and injury-induced facultative restoration
.
Development
2013
;
140
:
751
764
[PubMed]
32.
Bhatia
V
,
Cao
Y
,
Ko
TC
,
Falzon
M
.
Parathyroid hormone-related protein interacts with the transforming growth factor-β/bone morphogenetic protein-2/gremlin signaling pathway to regulate proinflammatory and profibrotic mediators in pancreatic acinar and stellate cells
.
Pancreas
2016
;
45
:
659
670
[PubMed]
33.
Saitoh
M
,
Endo
K
,
Furuya
S
, et al
.
STAT3 integrates cooperative Ras and TGF-β signals that induce Snail expression
.
Oncogene
2016
;
35
:
1049
1057
[PubMed]
34.
Oh
HM
,
Yu
CR
,
Dambuza
I
,
Marrero
B
,
Egwuagu
CE
.
STAT3 protein interacts with class O Forkhead transcription factors in the cytoplasm and regulates nuclear/cytoplasmic localization of FoxO1 and FoxO3a proteins in CD4(+) T cells
.
J Biol Chem
2012
;
287
:
30436
30443
[PubMed]
35.
Hsu
P
,
Santner-Nanan
B
,
Hu
M
, et al
.
IL-10 potentiates differentiation of human induced regulatory T cells via STAT3 and Foxo1
.
J Immunol
2015
;
195
:
3665
3674
[PubMed]
36.
Ma
W
,
Fuentes
G
,
Shi
X
,
Verma
C
,
Radda
GK
,
Han
W
.
FoxO1 negatively regulates leptin-induced POMC transcription through its direct interaction with STAT3
.
Biochem J
2015
;
466
:
291
298
[PubMed]
37.
Yang
G
,
Lim
CY
,
Li
C
, et al
.
FoxO1 inhibits leptin regulation of pro-opiomelanocortin promoter activity by blocking STAT3 interaction with specificity protein 1
.
J Biol Chem
2009
;
284
:
3719
3727
[PubMed]
38.
Rankin
MM
,
Wilbur
CJ
,
Rak
K
,
Shields
EJ
,
Granger
A
,
Kushner
JA
.
β-Cells are not generated in pancreatic duct ligation-induced injury in adult mice
.
Diabetes
2013
;
62
:
1634
1645
[PubMed]
39.
Kopp
JL
,
Dubois
CL
,
Schaffer
AE
, et al
.
Sox9+ ductal cells are multipotent progenitors throughout development but do not produce new endocrine cells in the normal or injured adult pancreas
.
Development
2011
;
138
:
653
665
[PubMed]
40.
Cavelti-Weder
C
,
Shtessel
M
,
Reuss
JE
, et al
.
Pancreatic duct ligation after almost complete β-cell loss: exocrine regeneration but no evidence of β-cell regeneration
.
Endocrinology
2013
;
154
:
4493
4502
[PubMed]
41.
Nepomnyashchikh
LM
,
Lushnikova
EL
,
Viskunov
VG
,
Protsenko
SI
,
Molodykh
OP
,
Koldysheva
EV
.
Ultrastructure of acinar cell injuries in experimental acute pancreatitis created by common bile duct ligation
.
Bull Exp Biol Med
2011
;
150
:
747
752
[PubMed]
42.
Meyerholz
DK
,
Williard
DE
,
Grittmann
AM
,
Samuel
I
.
Murine pancreatic duct ligation induces stress kinase activation, acute pancreatitis, and acute lung injury
.
Am J Surg
2008
;
196
:
675
682
[PubMed]
43.
Kishi
S
,
Takeyama
Y
,
Ueda
T
, et al
.
Pancreatic duct obstruction itself induces expression of alpha smooth muscle actin in pancreatic stellate cells
.
J Surg Res
2003
;
114
:
6
14
[PubMed]
44.
Friess
H
,
Lu
Z
,
Riesle
E
, et al
.
Enhanced expression of TGF-betas and their receptors in human acute pancreatitis
.
Ann Surg
1998
;
227
:
95
104
[PubMed]
45.
van Laethem
JL
,
Deviere
J
,
Resibois
A
, et al
.
Localization of transforming growth factor beta 1 and its latent binding protein in human chronic pancreatitis
.
Gastroenterology
1995
;
108
:
1873
1881
[PubMed]
46.
Valdez
IA
,
Dirice
E
,
Gupta
MK
,
Shirakawa
J
,
Teo
AKK
,
Kulkarni
RN
.
Proinflammatory cytokines induce endocrine differentiation in pancreatic ductal cells via STAT3-dependent NGN3 activation
.
Cell Rep
2016
;
15
:
460
470
[PubMed]
47.
Gorogawa
S
,
Fujitani
Y
,
Kaneto
H
, et al
.
Insulin secretory defects and impaired islet architecture in pancreatic beta-cell-specific STAT3 knockout mice
.
Biochem Biophys Res Commun
2004
;
319
:
1159
1170
[PubMed]
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at http://www.diabetesjournals.org/content/license.

Supplementary data