Adult pancreatic acinar cells give rise to ducts but not endocrine cells in response to growth factor signaling

Size: px
Start display at page:

Download "Adult pancreatic acinar cells give rise to ducts but not endocrine cells in response to growth factor signaling"

Transcription

1 Access the Development most First recent posted version epress online at on online 9 June publication 2010 as /dev date 9 June 2010 AND STEM CELLS RESEARCH ARTICLE 2289 Development 137, (2010) doi: /dev Published by The Company of Biologists Ltd Adult pancreatic acinar cells give rise to ducts but not endocrine cells in response to growth factor signaling Stacy A. Blaine 1, Kevin C. Ray 1, Reginald Anunobi 1, Maureen A. Gannon 2,3,4, Mary K. Washington 5 and Anna L. Means 1,3, * SUMMARY Studies in both humans and rodents have found that insulin + cells appear within or near ducts of the adult pancreas, particularly following damage or disease, suggesting that these insulin + cells arise de novo from ductal epithelium. We have found that insulin + cells are continuous with duct cells in the epithelium that makes up the hyperplastic ducts of both chronic pancreatitis and pancreatic cancer in humans. Therefore, we tested the hypothesis that both hyperplastic ductal cells and their associated insulin + cells arise from the same cell of origin. Using a mouse model that develops insulin + cell-containing hyperplastic ducts in response to the growth factor TGF, we performed genetic lineage tracing experiments to determine which cells gave rise to both hyperplastic ductal cells and duct-associated insulin + cells. We found that hyperplastic ductal cells arose largely from acinar cells that changed their cell fate, or transdifferentiated, into ductal cells. However, insulin + cells adjacent to acinar-derived ductal cells arose from pre-existing insulin + cells, suggesting that islet endocrine cells can intercalate into hyperplastic ducts as they develop. We conclude that apparent pancreatic plasticity can result both from the ability of acinar cells to change fate and of endocrine cells to reorganize in association with duct structures. KEY WORDS: Beta cell neogenesis, Growth factor signaling, Transdifferentiation, Mouse 1 Department of Surgery, 2 Department of Medicine, 3 Department of Cell and Developmental Biology, 4 Department of Molecular Physiology and Biophysics, and 5 Department of Pathology, Vanderbilt University Medical Center, Nashville, TN , USA. *Author for correspondence (anna.means@vanderbilt.edu) Accepted 14 May 2010 INTRODUCTION Traditionally, terminally differentiated cells of mammals were thought to be immutable, that is, confined to their final differentiated state for the remainder of their lifespans. However, in cases of injury or disease, the cellular composition of organs has been noted to change. For example, in both chronic pancreatitis and pancreatic cancer, many of the enzymeproducing acinar cells in the affected region of the pancreas are absent and replaced by hyperplastic ductal epithelium (Kloppel, 1993; Kloppel and Maillet, 1998). In this and other similar cases, it has been unclear how much of the change in composition results from reprogramming of mature cells and how much from stimulation of progenitor cells residing either within the organ or in the bone marrow. Complicating the issue of origin, small clusters of endocrine cells, notably insulin-producing cells, are closely associated with the hyperplastic ducts that arise during chronic pancreatitis and pancreatic cancer (Chen et al., 1988; Esposito et al., 2007; Phillips et al., 2007). This association between duct and insulin-expressing (insulin + ) cell reflects the relationship between these tissues in normal embryonic development, when insulin-producing cells differentiate from progenitors within the duct-like epithelium of the developing pancreas. Thus, the presence of insulin + cells in hyperplastic ducts suggests that the adult pancreas is capable of recapitulating embryonic development for the production of new insulinproducing cells either from dedifferentiation of mature cells or from an unknown precursor. However, lineage tracing experiments that can follow the fate of different cell types have yielded conflicting results (Desai et al., 2007; Dor et al., 2004; Inada et al., 2008; Xu et al., 2008). The need to provide new sources of insulin-producing cells for diabetic patients has sparked considerable interest in finding ways to develop increased numbers of cells either in vivo or in vitro. Although considerable work has indicated that cells can replicate to increase cell mass (Dor et al., 2004; Nir et al., 2007; Teta et al., 2007), the ability of other cells to give rise to new cells has been controversial. Thus far, the most conclusive example of cell neogenesis, or cells arising from another cell type in the adult pancreas, comes from gene therapy experiments, in which three genes that regulate cell development were introduced into pancreatic acinar cells, effectively converting many acinar cells to insulin-producing cells (Zhou et al., 2008). The enzyme-producing acinar cells are the most abundant cells of the pancreas, making them an attractive source for cell replacement. We have shown previously that acinar cells can transdifferentiate into ductal cells in vitro in response to transforming growth factor (TGF ) and that this is dependent upon its receptor, the epidermal growth factor receptor (EGFR) (Means et al., 2005). However, without directed transcriptional reprogramming, it remains to be determined whether acinar cells have an innate ability to transdifferentiate into insulinproducing cells. We have now addressed this issue by doing direct lineage tracing to determine the cell of origin both for hyperplastic pancreatic ducts and for the insulin + cells that arise within them in vivo in response to TGF. We have found that acinar cells are capable of transdifferentiating into hyperplastic ductal cells, but the insulin + cells embedded within this ductal epithelium arise from pre-existing insulin + cells that become intercalated into the ductal epithelium. Thus, even though one cell type is physically associated with another cell type, their origins might be completely different.

2 2290 RESEARCH ARTICLE Development 137 (14) MATERIALS AND METHODS Animals All experiments were done with approval of the Vanderbilt Institutional Animal Care and Use Committee. Tissue-specific Cre transgenes were bred into mice carrying the R26R-EYFP [Gt(ROSA)26Sor tm1(eyfp)cos ] reporter (Srinivas et al., 2001) and the MT-TGF transgene (Sandgren et al., 1990) that contains the metallothionein promoter driving expression of TGF. The Cre transgene and the R26R-EYFP reporter allele were never derived from the same parent as this led to embryo-wide recombination in tissues throughout the mouse. Cre transgenes included elastase 500 -Cre, containing 500 bp of the elastase promoter that has been shown to direct acinar-specific expression (Kruse et al., 1993) and with growth hormone 3 untranslated sequences (Postic et al., 1999); Villin-Cre (el Marjou et al., 2004), a kind gift from Sylvie Robine; Pdx1 PB -CreERT (Zhang et al., 2005), containing the 1 kilobase islet-specific enhancer region from the Pdx1 promoter and the hsp68 minimal promoter driving expression of Cre fused to a mutated estrogen receptor hormone-binding domain that renders Cre inactive until tamoxifen administration; and RIP-CreERT (Dor et al., 2004) containing the rat insulin promoter driving expression of a similar tamoxifen-inducible CreERT protein. For these mice, tamoxifen or corn oil vehicle alone was administered at 4-6 weeks of age, in 3 doses over a 5-day time period, 2 mg/dose via intraperitoneal injection. One week following the final injections, mice were placed on water containing 25 mm ZnSO 4 to induce expression of the MT- TGF promoter. No ductal hyperplasia was observed until 2-3 months of Zn 2+ treatment. Following 6 months of ZnSO 4 treatment, mice were euthanized and analyzed for EYFP expression. Immunofluorescence Pancreatic samples were fixed overnight at 4 C in 4% paraformaldehyde. Shorter fixation times resulted in loss of EYFP, particularly in cytoplasm. Tissues were then washed, dehydrated through a series of ethanol and Histoclear (National Diagnostics, Atlanta, GA, USA) washes and then embedded in paraffin. Five micron sections were collected, dewaxed and heated either at 60 C overnight or at high pressure in a pressure cooker (Cuisinart, East Windsor, NJ, USA) for 15 minutes in 100 mm Tris, ph 10, cooled, washed in PBS, blocked in 5% donkey serum in PBS and then incubated with indicated antibodies to GFP (Invitrogen, Carlsbad, CA or Novus Biological, Littleton, CO, USA), insulin (Linco Research, St Charles, MO, USA), glucagon (Linco Research), pancreatic polypeptide (Linco Research), somatostatin (Santa Cruz Biotechnology, Santa Cruz, CA, USA), amylase (Santa Cruz Biotechnology), cytokeratin 19 [Developmental Studies Hybridoma Bank (DSHB), Iowa City, IA, USA], Ngn3 (DSHB) and laminin (Biogenex, San Ramon, CA, USA). Antibody staining was visualized with Cy2, Cy3 or Cy5 fluorophor-conjugated antirabbit, anti-guinea pig, anti-mouse or anti-rat antibodies (Jackson Immunoresearch Laboratories, West Grove, PA, USA) or biotinylated antirabbit (Vector Laboratories, Burlingame, CA, USA) followed by Cy3- conjugated avidin (Jackson Immunoresearch Laboratories). Nuclei were stained with Toto3 (Invitrogen). Images were captured on an LSM510 confocal microscope (Carl Zeiss Microimaging, Thornwood, NY, USA) at 1 m optic depth. Some sections were also labeled colorimetrically using the above primary antibodies and the Vectastain Elite ABC Kit (Vector Laboratories) and 3,3 -diaminobenzidine (Invitrogen) as the peroxidase substrate. Statistical methods Lineage tracing experiments were quantified from 2 Villin-Cre, 3 elastase 500 -Cre, 2 RIP-CreERT and 3 Pdx1 PB -CreERT mice. Results from Villin-Cre were combined with elastase 500 -Cre as these lines gave comparable results. For each mouse, insulin + cells within islets and insulin + cells in hyperplastic ducts were counted. Averages and s.e.m. were determined and compared by two-tailed t-tests. RESULTS Insulin + cells arise within ductal epithelium A number of studies have reported insulin-expressing (insulin + ) cell clusters associated with pancreatic ducts, particularly with the hyperplastic ducts that arise following damage or disease (Bonner- Weir et al., 1993; Chen et al., 1988; Esposito et al., 2007; Phillips et al., 2007). In order to determine if these insulin + cells are an integral part of the ductal epithelium, we first examined sections from human patients who had either chronic pancreatitis or pancreatic cancer. Whereas many insulin + cells were located immediately adjacent to hyperplastic ducts (data not shown), we also found individual insulin + cells that were continuous with the ductal epithelium (Fig. 1A,B) and thus did not reflect independent endocrine structures, but rather endocrine cells that were located within ductal epithelium. We also found that mouse models recapitulated this phenomenon. In early adenomatous lesions induced by pancreatic expression of the Kras G12D oncogene, insulin + cells were also found within ductal epithelium (Fig. 1C). Similarly, in mice overexpressing the growth factor TGF, a growth factor often overexpressed in human pancreatic cancer (Korc et al., 1992), insulin + cells were found within hyperplastic ductal epithelium (Fig. 1D). In both human and mouse models, many more insulin + cells were present immediately adjacent to ducts rather than being continuous with the cells of the ductal epithelium. In order to distinguish whether these closely associated cells were merely separate, juxtaposed structures or whether the insulin + cells might have delaminated out of ductal epithelium, we determined whether the single insulin + cells as well as clusters of insulin + cells associated with hyperplastic ducts were each part of the ductal epithelium. We double-labeled tissue sections from TGF overexpressing mice for insulin and laminin. Laminin is a basement membrane protein that delineates the outer edges of islets, normal pancreatic ducts and some hyperplastic ducts, effectively marking the boundaries around these structures. As expected, when insulin + cells were continuous with duct cells within ductal epithelium, these insulin + cells were contained within the same basement membrane as the surrounding ductal cells (Fig. 2A,B). Clusters of insulin + cells that appeared to be separate but closely juxtaposed to hyperplastic ducts were also contained within the basement membrane of the ducts (Fig. 2C,D). Notably, there was no laminin present separating ducts from insulin + cells, Fig. 1. Insulin-expressing cells arise within hyperplastic ductal epithelium in humans and mice. (A,B) Pancreatic sections were immunolabeled for insulin (brown) and nuclei were counterstained (blue) in samples from human chronic pancreatitis (A) and human lowgrade pancreatic intraepithelial neoplasia (PanIN) lesions associated with pancreatic ductal adenocarcinoma (B). (C) Mouse model of PanIN in which activated Kras G12D is expressed in pancreas. (D) Mouse overexpressing the growth factor TGF. Scale bar: 50 m.

3 Lineage tracing ductal insulin + cells RESEARCH ARTICLE 2291 Fig. 2. Insulin + cell clusters are contained within ductal basement membrane. Sections from TGF -overexpressing pancreas were immunolabeled for insulin (green) and laminin (red) as a marker of the basement membrane, and counterstained to show nuclei (blue). (A) Several insulin + cells (arrow) that are continuous with the ductal epithelium are contained within a laminin-positive basement membrane. (B) Laminin staining alone from A. (C) A larger cluster of insulin + cells (arrows) is contained within a ductal basement membrane. (D) Laminin alone from C. Scale bar: 50 m. suggesting that the insulin + cells were an integral part of the ductal epithelium, possibly in the process of delaminating out of that epithelium as happens during embryonic development. For comparison, we also examined the formation of basement membrane as endocrine cells normally delaminated from ductal epithelium during late embryonic development. At embryonic day 17.5, laminin marked basement membrane around forming ducts and islets (see Fig. S1 in the supplementary material). However, at discreet points, no laminin was detected between the ducts and forming islets. At these points, laminin formed a continuous layer from the duct, extending out around the forming islet. This continuity of basement membrane from duct to delaminating endocrine cells is similar between embryonic tissue and TGF induced hyperplastic tissue. However, in the TGF -overexpressing mice, areas of contact between duct and islet clusters that were devoid of laminin were much broader than in embryonic pancreas. For example, approximately 12 endocrine cells were juxtaposed to ductal epithelium in Fig. 2C, whereas only 1-3 endocrine cells were immediately juxtaposed to duct cells in embryonic pancreas (see Fig. S1 in the supplementary material). As larger ductal endocrine clusters were observed in TGF -overexpressing mice, they tended to wrap around the duct, maintaining a close association rather than separating from the ductal epithelium. This morphology suggests that if endocrine cells are indeed delaminating from ductal epithelium, they remain within the original basement membrane structure for some period of time. Acinar cells transdifferentiate into duct cells but not endocrine cells Although the juxtaposition of single insulin + cells and insulin + cell clusters to ductal epithelium suggested that these endocrine cells might have arisen from the same progenitors as the hyperplastic duct cells, we used genetic lineage tracing to define the precise cell of origin for both the hyperplastic ducts and the insulin + cells associated with them. In order to label large numbers of specific cell types in vivo, we made use of the R26R-EYFP allele, which is transcriptionally silent until Cre recombinase removes a transcriptional stop cassette allowing expression of the EYFP protein (Srinivas et al., 2001). Once recombined, EYFP is expressed from the ubiquitously active Rosa26 locus, even if Cre is subsequently lost from the cell. Because expression of EYFP results from genomic recombination of the R26R-EYFP allele, it represents a heritable genetic trait that will be durably expressed throughout the life of the cell and also passed on to any progeny cells. Thus, tissue-specific activation of the R26R-EYFP allele provides an indelible marker of both Cre-expressing cells and of any cells that arise from those cells. We employed two Cre transgenes, Villin-Cre (el Marjou et al., 2004) and elastase 500 -Cre to determine the fate of acinar cells in response to TGF. We previously showed that Villin-Cre activity is specific to acinar cells in the pancreas, although it is also expressed in other cells of other organs (Means et al., 2005). We also used 500 basepairs of the elastase promoter (Kruse et al., 1993) to drive acinar expression of Cre in the elastase 500 -Cre transgene. However, as mice aged, we observed a small degree of Cre activity in endocrine cells, with approx. 5% of endocrine cells being labeled in addition to virtually all acinar cells in both Villin-Cre and elastase 500 -Cre mice. Endocrine recombination was lower in young mice (Means et al., 2005) (see Fig. S2 in the supplementary material) and probably resulted from rare misexpression of the transgenes during the 8- month timecourse of our experiments. However, we cannot rule out the possibility that there is a low rate of transdifferentiation of acinar cells into islet cells in normal pancreas. The Villin-Cre or elastase 500 -Cre transgenes along with the R26R-EYFP reporter were bred into mice expressing the MT- TGF transgene. Mice were placed on water containing 25 mm ZnSO 4 to induce expression from this metallothionein promoter and were examined 6 months later to determine if acinar cells had given rise to the hyperplastic ductal cells and/or duct-associated insulin + cells. Confirming previous work showing that acinar cells can transdifferentiate into ducts in vitro (Means et al., 2005), hyperplastic ductal cells carried the EYFP lineage label, indicating that acinar cells gave rise to the majority of hyperplastic ducts that developed in response to TGF expression (Fig. 3). However, with both acinar cell-specific Cre transgenes, there was no EYFP label present in the majority of insulin-producing cells found either within the hyperplastic ducts or in normal islet structures (Fig. 4). As discussed above and quantified below, both elastase 500 -Cre and Villin-Cre labeled 5.4±1.9% of normal islet cells and a similar 5.3±1.1% of duct-associated insulin + cells. When single insulin + cells were observed in hyperplastic ducts, they were continuous with EYFP + ductal cells. Thus, acinar cells were not the cells of origin for ductal insulin + cells even though they gave rise to duct cells immediately adjacent to these insulin + cells. These data indicate that although acinar cells can transdifferentiate into ductal cells, they do not seem to be capable of giving rise to insulinproducing cells in response to TGF. Duct-associated insulin + cells arise from pre-existing endocrine cells Although it was possible that either duct cells or an unknown progenitor cell might have given rise to duct-associated insulin + cells, we first sought to rule out the possibility that pre-existing islet cells could have intercalated into the epithelium of the hyperplastic ducts. The Pdx1 PB -CreERT transgene is expressed specifically in islets, principally in cells but also in 4-5% of glucagon-producing cells and somatostatin-producing cells (Zhang et al., 2005). However, Pdx1 PB -CreERT is only active following treatment with

4 2292 RESEARCH ARTICLE Development 137 (14) Fig. 3. Acinar cells contribute to hyperplastic ducts. In TGF -overexpressing mice, acini were labeled for expression of the EYFP protein via the Villin-Cre transgene and R26R-EYFP reporter. Mice were placed on Zn 2+ -containing water at 6 weeks of age and hyperplastic ducts were allowed to develop for 6 months. (A) Co-immunostaining demonstrated that EYFP (green) was expressed in amylase-positive acinar cells (red) as expected, but not all EYFP-positive cells co-expressed amylase, indicating that some previously labeled acinar cells were no longer acinar cells. (B) Amylase staining alone from A. (C) EYFP staining alone from A. (D) EYFP was also expressed in cytokeratin-19-postive (red) hyperplastic ducts, but not in cytokeratin- 19-positive ducts of normal morphology (arrowheads), indicating that hyperplastic ducts, but not normal ducts, arose from acinar-to-ductal transdifferentiation. (E) Cytokeratin 19 staining alone from D. (F) EYFP staining alone from D. Arrows, co-labeled cells; arrowheads, normal pancreatic duct; asterisks, non-specific staining of luminal duct contents. Scale bar: 50 m. tamoxifen, thus any endocrine cells that arise after tamoxifen has been cleared from the bloodstream will not be recombined or labeled with EYFP. Thus, we could investigate whether insulinproducing cells within ducts were derived from islet cells that were present before the onset of ductal hyperplasia. We also used a second islet-specific Cre transgene, RIP-CreERT (Dor et al., 2004) but found that, over the 8-month timecourse of these experiments, this transgene mediated recombination in more than half of all insulin + cells, even in the absence of tamoxifen (data not shown). Additionally, RIP-CreERT labeled a relatively large percentage of acinar cells. We examined two RIP-CreERT; R26R-EYFP mice, not overexpressing TGF, at 7-8 months of age (the average age of MT-TGF mice examined), and found that 3-5% of all acinar cells were labeled with EYFP (see Fig. S3 in the supplementary material). Therefore, we did not use the RIP-CreERT transgene for further lineage tracing experiments. Pdx1 PB -CreERT, conversely, did not show any labeling of acinar cells in 7- to 8-month-old mice (see Fig. S3 in the supplementary material) and labeled only 0.31±0.09% of endocrine cells with no tamoxifen over that same timecourse (n 3 mice, cells counted for each). We interbred mice carrying the islet-specific Pdx1 PB -CreERT transgene to mice carrying the R26R-EYFP reporter and the MT- TGF transgene. These mice were injected with three low doses of tamoxifen at 1 month of age to yield EYFP labeling in approximately 60-65% of islet cells. At this stage, MT-TGF mice have a morphologically normal pancreas, and require at least 2-3 months of zinc induction before hyperplastic ducts appear. This slow development precluded the inadvertent labeling of insulin + cells as they arose in hyperplastic ducts, so that only pre-existing insulin-producing cells would be labeled. As reported previously (Zhang et al., 2005), no labeling was seen in non-endocrine cells. One week after the final tamoxifen injection, mice were placed on water containing zinc to induce TGF expression. After 6 months of zinc induction, mice were analyzed for EYFP expression in different pancreatic cell types (Fig. 5). As discussed above, EYFP labeling was found only in islets in control mice not carrying the TGF transgene. However, in TGF -overexpressing mice, insulin + cells found either within or closely associated with the ductal epithelium also carried the EYFP label, indicating that these cells arose from the endocrine cells that were present at the time of tamoxifen injection. The number of ductal insulin + cells expressing EYFP reflected the percentage labeled in islets. That is, 64.2±1.2% of islet cells were labeled, whereas 63.1±3.4% of duct-associated insulin + cells were labeled. Supporting a cell origin for ductal insulin + cells, we did not detect any Ngn3 expression in MT-TGF mice nor did ductal insulin + cells co-express ductal markers such as cytokeratin 19 (Fig. 5) or bind DBA (data not shown). We also determined whether cells could act as multipotent progenitor cells and give rise to other cell types using Pdx1 PB -CreERT to label cells. No EYFP-labeling was found in duct cells, acinar cells or the majority of glucagon-expressing (glucagon + ) cells, both in islets and in hyperplastic ducts (Fig. 5; data not shown). A very small number of glucagon + cells containing the EYFP lineage label were seen in both islets and in ductal structures, Fig. 4. Acinar cells do not give rise to duct-associated insulin + cells. Acinar cells were labeled using the Villin-Cre transgene to activate EYFP expression. Following induction of hyperplastic ducts, sections were co-immunostained for EYFP (green) and insulin (red) and nuclei were counterstained in blue. (A,C) Although ductal cells immediately surrounding insulin + cells were lineage-labeled with EYFP expression, ductal insulin + cells (arrows) were not labeled. (B,D) EYFP only staining of A and C, respectively. Scale bar: 50 m.

5 Lineage tracing ductal insulin + cells RESEARCH ARTICLE 2293 Fig. 5. Ductal insulin + cells arise from pre-existing cells. cells were labeled via the Pdx1 PB -CreERT transgene with activity induced by tamoxifen injection. One week following the final tamoxifen injections, mice were given Zn 2+ to induce TGF expression and analyzed 6 months later for expression of EYFP in different cell types. (A-D) EYFP (green) was found in single insulin + cells (red; arrow) continuous with ductal epithelium (cytokeratin 19, blue), indicating that the insulin + cell arose from a pre-existing cell. Note that a second insulin + cell was not labeled by EYFP, reflecting the ~65% labeling efficiency for the Pdx1 PB -CreERT transgene. A is the merge of panels B-D. (E-H) A cluster of insulin + cells that is in contact with the ductal lumen was also labeled with EYFP, indicating that those cells arose from pre-existing cells. E is the merge of panels F-G. Arrows indicate locations that can be compared across rows of panels. Scale bar: 10 m. reflecting the small number of glucagon + cells normally labeled by the Pdx1 PB -CreERT transgene. Thus, pre-existing cells gave rise to ductal insulin + cells but did not give rise to other ductassociated cell types. When the EYFP-insulin double labeling efficiency is compared between mice labeled with acinar-specific Cres and mice labeled with an islet-specific Cre, it is clear that pre-existing endocrine cells, not acinar cells, gave rise to duct-associated insulin + cells (Fig. 6). Examining acinar-specific Cre activity alone with its lowefficiency labeling of endocrine cells leaves open the possibility that a small percentage of endocrine cells, in islets as well as in ducts, arose via acinar cell transdifferentiation. However, the nearly identical labeling of endocrine cells in islets and in ducts by Pdx1 PB -CreERT without any acinar labeling indicates that any acinar cell transdifferentation is a minor component at best. By combining analyses of different lineages, we conclude that acinar cells transdifferentiated into hyperplastic ductal epithelium and that pre-existing endocrine cells became intercalated into these structures. However, we could not rule out that insulin + cells were present normally in exocrine structures and maintained their exocrine association as ductal hyperplasia occurred. Insulin + cells have been reported in normal ducts of rats, with a frequency that decreases with age (Madden and Sarras, 1989). Therefore, we examined whether insulin + cells are present in normal pancreatic ducts as mice age. Examining random cross-sections of pancreas from 2-month-old normal mice, no insulin + cells were found within either ductal or acinar structures (n 3 mice, 3-4 cross-sections each). Because the main pancreatic duct has been proposed as a site of cell neogenesis (Sharma et al., 1999), we also specifically examined sections through the main pancreatic duct. Two-monthold mice did indeed contain 1-2 clusters each of insulin + cells continuous with ductal epithelium per approximately 1000 ductal cells of the main pancreatic duct or closely associated interlobular duct branches. Each cluster comprised 1-3 insulin + cells for an average of 0.32±0.13% of main duct cells (see Fig. S4 in the supplementary material; data not shown). However, in four 6- month-old mice examined, we found no insulin + cells within the main pancreatic duct, indicating that less than 0.1% of normal main duct cells are insulin +, which is comparable with the results from rat pancreas (Madden and Sarras, 1989). Thus, it appears that mice lose insulin + cells within their larger ducts as they age. Because we began Zn 2+ induction of TGF expression at approximately 6 weeks of age, it was possible that TGF merely expanded the population present in these large ducts of the pancreas. Therefore, we examined the occurrence and distribution of duct-associated insulin + cells in MT-TGF mice. We found that every cross-section of the pancreas, not just in main duct regions, contained insulin + cells within hyperplastic ducts, with an average of 12±1.3 ducts per cross-section containing insulin + cells (n 5 MT-TGF mice, 2-3 sections counted per mouse). Thus, it is unlikely that TGF overexpression expanded the number of insulin + cells normally present in ductal epithelium. Fig. 6. Pre-existing cells, not acinar cells, give rise to ductal insulin + cells in response to TGF signaling. Quantification of the results from Figs 4 and 5 compare the ability of acinar cells and islet cells to give rise to duct-associated insulin + cells. For acinar lineage tracing (left two bars), results from 3 elastase 500 -Cre and 2 Villin-Cre mice were averaged. Note that although these transgenes labeled virtually all acinar cells, they also labeled ~5% of islet (gray bar) cells by the time mice were 8 months old. Similarly, 5% of duct-associated (black bar) insulin + cells were labeled in these mice. For islet lineage tracing (right two bars), 3 Pdx1 PB -CreERT mice were used to label islet cells with tamoxifen injected one week before MT-TGF transgene induction and ~2 months before the appearance of hyperplastic ducts. At the tamoxifen dosage used, 64.2±1.2% of islet (gray bar) cells were labeled with the EYFP lineage reporter. Similarly, 63.1±3.4% of duct-associated (black bar) insulin + cells were labeled. Together, these data indicate that acinar cells do not contribute to ductal insulin + cells and that the majority of duct-associated insulin + cells arise from preexisting cells. Percentages were determined from 3 elastase 500 -Cre, 2 Villin-Cre and 3 Pdx1 PB -CreERT mice, with islet insulin + cells and ductal insulin + cells counted per mouse.

6 2294 RESEARCH ARTICLE Development 137 (14) DISCUSSION We have found that pancreatic acinar cells have a limited ability to give rise to other cell types within the adult mouse pancreas in vivo. Using rigorous genetic lineage tracing, we have demonstrated that pancreatic acinar cells have the ability to transdifferentiate into the hyperplastic ducts that develop upon overexpression of TGF. Although numerous insulin-expressing cells were part of this hyperplastic ductal epithelium, we have shown that these ductal insulin + cells originated from pre-existing cells and were not the result of cell neogenesis. Although acinar cells are capable of giving rise to ductal cells in response to TGF signaling both in vitro and in vivo, they were not the cell of origin for endocrine cells found associated with such ducts in vivo. Phylogenetic differences in cell location Although the majority of cells are clustered in pancreatic islets in mammals, other species show a variety of localizations of insulin-producing cells (for a review, see Gannon and Wright, 1999). Tunicates and cephalochordates such as amphioxus have individual insulin + cells located within gut epithelium, providing an evolutionary precedent for insulin + cells within epithelial sheets. However, vertebrates as distant as agnathans organize cells into distinct islet clusters. Thus, the ability of cells to emigrate out of epithelial sheets wherein they arise and cluster evolved even before development of a distinct exocrine pancreas. Acinar cells transdifferentiate into ductal cells In addition to demonstrating that ductal insulin + cells do not arise from acinar cells, this study demonstrates that cells do not give rise to hyperplastic ducts in response to TGF -overexpression. Rather, TGF induces transdifferentiation of acinar cells into hyperplastic ducts. In contrast to our studies, Strobel et al. (Strobel et al., 2007) found that acinar cells gave rise to only a small percentage of the hyperplastic ducts that arose in response to cerulein, a cholecystokinin analog that induces pancreatitis in mice. This difference might result from specific effects of TGF on acinar cells and/or because cerulein treatment results in widespread apoptosis of acinar cells. It is probable that both acinar cells and duct cells can give rise to hyperplastic ducts in the pancreas, with acinar cells predominating in our study owing to their overwhelming abundance. Gidekel Friedlander et al. (Gidekel Friedlander et al., 2009) performed lineage studies to demonstrate that activation of the Kras G12D oncogene via the insulin promoter could give rise to ductal adenomas, suggesting that islets could be a cell of origin for pancreatic ductal adenocarcinoma. However, this group used the RIP-CreERT transgene to activate Kras G12D expression. The authors described little or no labeling of acinar cells with this CreERT transgene. We have shown here that RIP-CreERT recombines 3-6 percent of acinar cells. The discrepancy in our studies might arise from the reporter gene used to assess recombination. Gidekel Friedlander et al. (Gidekel Friedlander et al., 2009) used the R26R reporter that expresses -galactosidase ( gal) upon Cre-mediated recombination (Soriano, 1999) rather than the EYFP reporter that we used (Srinivas et al., 2001). We have found that it is very difficult to preserve gal activity in adult acinar cells. In our hands, reliable gal detection in acinar cells requires transcardial perfusion of the fixative, rapid freezing and re-fixation of tissue sections on the slide. Even EYFP is less wellpreserved in acinar cytoplasm than in islet cytoplasm (see Fig. S3 in the supplementary material) but its small size allows EYFP to diffuse into the nucleus where it is more readily detected. Thus, the ability of Kras G12D to induce islet cells to give rise to ductal adenomas will have to await experiments with a more specific islet CreERT, such as the Pdx1 PB -CreERT used here. Origin of cells in the adult pancreas Various cell types have been postulated as possible sources for cell renewal in vivo. Many researchers have suggested that there are facultative progenitor cells residing within normal ductal epithelium that lead to islet cell regeneration. These studies have primarily relied on morphological analysis after the stimulation of pancreas-wide damage by either partial pancreatectomy (Ackermann Misfeldt et al., 2008; Bonner-Weir et al., 1993) or ductal ligation (Wang et al., 1995) that results in the presence of insulin + cells closely associated with ducts. The appearance of these insulin + cells has been interpreted as new islets budding from the ductal epithelium (Esposito et al., 2007; Gu and Sarvetnick, 1993; Phillips et al., 2007; Song et al., 1999). However, a recent report from Desai et al. (Desai et al., 2007) demonstrated that insulin + cells in islets observed following surgical removal of part of the pancreas did not result from acinar cell transdifferentiation, a result similar to ours using growth factor induction of ductal hyperplasia. Desai et al. (Desai et al., 2007) did not determine what the cell of origin was in their partial pancreatectomy model, nor did they examine duct-associated insulin + cells. In order to determine if duct cells could transdifferentiate into insulin-producing cells, Solar et al. (Solar et al., 2009) used the HNF1 promoter to drive CreERT in pancreatic ducts. They found that duct cells in adult mice did not contribute to insulin-producing cells even after pancreatic duct ligation, but they appeared to examine only endocrine cells in islet structures, not within ducts, and they did not identify what the origin was for these endocrine cells. Inada et al. (Inada et al., 2008) used the carbonic anhydrase II (CAII) promoter to drive CreERT expression in pancreatic ducts. After partial pancreatectomy, both insulin + and ductal cells were labeled with the Cre reporter, suggesting that ducts could give rise to new cells. However, this CAII-CreERT transgene was not characterized for its expression pattern or labeling efficiency, thus putting any conclusions on hold until those are determined. Other studies have also shown that duct-associated insulin + cells can arise in mouse pancreas, for example, in response to adenoviral-mediated overexpression of two EGFR ligands, HB- EGF and betacellulin (Kozawa et al., 2005; Tokui et al., 2006). The ability of both these ligands to activate EGFR, as does TGF used in the studies presented here, suggests that the increase in cells might have resulted from relocalization and/or expansion of preexisting cells. In addition to insulin + cells appearing in ducts, neurogenin 3 (Ngn3) has also been detected in pancreatic ducts in response to damage. Partial pancreatectomy that retains the main pancreatic duct in the tail of the pancreas while removing most other exocrine and endocrine tissue resulted in the appearance of Ngn3 + cells in ducts (Ackermann Misfeldt et al., 2008), whereas partial pancreatectomy completely removing the distal main duct did not result in Ngn3 expression (Lee et al., 2006). Xu et al. (Xu et al., 2008) demonstrated that partial pancreatic duct ligation, a method of constricting flow through the main pancreatic duct resulting in damage distal to the site of ligation, also resulted in the appearance of Ngn3-expressing cells. Ngn3 is a regulator of cell neogenesis during embryonic development, so its expression in adult pancreas might suggest that cell neogenesis could be occurring. In either of the above studies it is not known which cells gave rise to the Ngn3 + cells. No Ngn3 + cells were detected in MT-TGF mice.

7 Lineage tracing ductal insulin + cells RESEARCH ARTICLE 2295 However, cellular changes in response to TGF occur slowly over many months so transient Ngn3 expression could have been missed. We chose this model because we could be sure that tamoxifen would be cleared from the bloodstream long before ductal insulin + cells were observed. However, we have also examined mice that overexpress a particular isoform of a related family member, soluble HB-EGF (shb-egf) (Ray et al., 2009). In these mice, ductal hyperplasia occurs beginning around 1-2 months of age and many ductal insulin + cells are observed by 2 months of age (data not shown). Even in these mice, no Ngn3 was detected. The discrepancies in Ngn3 detection throughout the literature might be owing to different inductive cues and/or differences in antibodies used for detection. In none of the studies were adult Ngn3 + cells lineage mapped to determine their cell of origin. Wang et al. (Wang et al., 2009) have demonstrated a low level of Ngn3 expression in adult islets and have demonstrated that its loss in adult cells affected islet function. Therefore, Ngn3 is not only a marker of endocrine precursors but might also be a marker of some adult cells. In the studies by Xu et al., adult Ngn3-expressing cells were not shown to give rise to cells in vivo, but could be induced to differentiate into insulin-producing cells ex vivo, suggesting that adult pancreatic cells are capable of generating new cells but the adult environment prohibits completion of this pathway. In support of this hypothesis, others have reported ex vivo expansion of insulin + cells from pancreatic cultures consisting primarily of acinar cells (Baeyens et al., 2005; Minami et al., 2005). Thus, it might be possible to derive ex vivo methods for increasing cell mass from adult pancreas for subsequent islet transplantation into diabetic patients. We have shown previously that acinar cells (Means et al., 2005) isolated from mature mouse pancreas could transdifferentiate into ductal cells ex vivo under the influence of exogenous TGF and that this process involved a dedifferentiation step. Here we show that this is recapitulated in vivo with acinar cells giving rise to hyperplastic ducts. The use of transgenes, allowing for both acinar cell- and islet cell-specific Cre recombinase expression coupled with the R26R-EYFP reporter, enabled us to determine the level of plasticity of adult pancreatic acinar cells in response to TGF in vivo. Our findings that pre-existing cells gave rise to ductassociated insulin + cells are in agreement with the recent results of others, that only cells can give rise to new cells in vivo. Dor et al. (Dor et al., 2004) used an elegant insulin-promoter-based approach to show that existing cells are the sole source of new cells in the adult mouse under normal conditions. Along the same lines, an innovative DNA analog-based lineage tracing method has been utilized to demonstrate that cell generation is the result of self duplication during both normal growth conditions and in cases of acute cell expansion, and does not involve specialized progenitors (Teta et al., 2007). Interestingly, multiple studies that demonstrated an increase in insulin-producing cells both in vivo and in vitro used EGFR ligands to stimulate this cell expansion (Baeyens et al., 2005; Minami et al., 2005; Rooman et al., 2000; Suarez-Pinzon et al., 2005a; Suarez- Pinzon et al., 2005b). Although our findings indicate that EGFR signaling does not lead to islet neogenesis in vivo, EGFR signaling may allow alterations in the cell environment, such as ductal localization, that allow increased proliferation or survival. It will be interesting to determine in future studies whether the ductal environment promotes the proliferation of functional cells. Such studies could aid our ability to increase endocrine function in diabetic patients that retained some cell mass. Acknowledgements We thank Steven Leach for collaborating to generate the elastase 500 -Cre mouse and for critically reading this manuscript. This work was supported by JDRF research grant (A.L.M.), and through the Vanderbilt Digestive Diseases Research Center (NIH P30DK058404, M.K.W.) and the use of the Vanderbilt Cell Imaging Shared Resource (NIH CA68485, DK20593, DK58404, HD15052, DK59637 and EY08126). Deposited in PMC for release after 12 months. Competing interests statement The authors declare no competing financial interests. Supplementary material Supplementary material for this article is available at References Ackermann Misfeldt, A., Costa, R. H. and Gannon, M. (2008). Beta-cell proliferation, but not neogenesis, following 60% partial pancreatectomy is impaired in the absence of FoxM1. Diabetes 57, Baeyens, L., De Breuck, S., Lardon, J., Mfopou, J. K., Rooman, I. and Bouwens, L. (2005). In vitro generation of insulin-producing beta cells from adult exocrine pancreatic cells. Diabetologia 48, Bonner-Weir, S., Baxter, L. A., Schuppin, G. T. and Smith, F. E. (1993). A second pathway for regeneration of adult exocrine and endocrine pancreas. A possible recapitulation of embryonic development. Diabetes 42, Chen, J., Baithun, S. I., Pollock, D. J. and Berry, C. L. (1988). Argyrophilic and hormone immunoreactive cells in normal and hyperplastic pancreatic ducts and exocrine pancreatic carcinoma. Virchows Arch. A Pathol. Anat. Histopathol. 413, Desai, B. M., Oliver-Krasinski, J., De Leon, D. D., Farzad, C., Hong, N., Leach, S. D. and Stoffers, D. A. (2007). Preexisting pancreatic acinar cells contribute to acinar cell, but not islet beta cell, regeneration. J. Clin. Invest. 117, Dor, Y., Brown, J., Martinez, O. I. and Melton, D. A. (2004). Adult pancreatic beta-cells are formed by self-duplication rather than stem-cell differentiation. Nature 429, el Marjou, F., Janssen, K. P., Chang, B. H., Li, M., Hindie, V., Chan, L., Louvard, D., Chambon, P., Metzger, D. and Robine, S. (2004). Tissue-specific and inducible Cre-mediated recombination in the gut epithelium. Genesis 39, Esposito, I., Seiler, C., Bergmann, F., Kleeff, J., Friess, H. and Schirmacher, P. (2007). Hypothetical progression model of pancreatic cancer with origin in the centroacinar-acinar compartment. Pancreas 35, Gannon, M. and Wright, C. V. (1999). Endodermal patterning and organogenesis. In Cell Lineage and Fate Determination (ed. S. A. Moody), pp San Diego, CA: Academic Press. Gidekel Friedlander, S. Y., Chu, G. C., Snyder, E. L., Girnius, N., Dibelius, G., Crowley, D., Vasile, E., DePinho, R. A. and Jacks, T. (2009). Contextdependent transformation of adult pancreatic cells by oncogenic K-Ras. Cancer Cell 16, Gu, D. and Sarvetnick, N. (1993). Epithelial cell proliferation and islet neogenesis in IFN-g transgenic mice. Development 118, Inada, A., Nienaber, C., Katsuta, H., Fujitani, Y., Levine, J., Morita, R., Sharma, A. and Bonner-Weir, S. (2008). Carbonic anhydrase II-positive pancreatic cells are progenitors for both endocrine and exocrine pancreas after birth. Proc. Natl. Acad. Sci. USA 105, Kloppel, G. (1993). Pathology of nonendocrine pancreatic tumors. In The Pancreas: Biology, Pathobiology, and Disease (ed. V. L. W. Go), pp New York: Raven Press. Kloppel, G. and Maillet, B. (1998). Pathology of chronic pancreatitis. In The Pancreas, vol. 1 (ed. H. G. Beger, A. L. Warshaw, M. W. Buchler, D. L. Carr- Locke, J. P. Neoptolemos, C. Russell and M. G. Sarr), pp London: Blackwell Science. Korc, M., Chandrasekar, B., Yamanaka, Y., Friess, H., Buchier, M. and Beger, H. G. (1992). Overexpression of the epidermal growth factor receptor in human pancreatic cancer is associated with concomitant increases in the levels of epidermal growth factor and transforming growth factor alpha. J. Clin. Invest. 90, Kozawa, J., Tokui, Y., Moriwaki, M., Li, M., Ohmoto, H., Yuan, M., Zhang, J., Iwahashi, H., Imagawa, A., Yamagata, K. et al. (2005). Regenerative and therapeutic effects of heparin-binding epidermal growth factor-like growth factor on diabetes by gene transduction through retrograde pancreatic duct injection of adenovirus vector. Pancreas 31, Kruse, F., Rose, S. D., Swift, G. H., Hammer, R. E. and MacDonald, R. J. (1993). An endocrine-specific element is an integral component of an exocrinespecific pancreatic enhancer. Genes Dev. 7, Lee, C. S., De Leon, D. D., Kaestner, K. H. and Stoffers, D. A. (2006). Regeneration of pancreatic islets after partial pancreatectomy in mice does not involve the reactivation of neurogenin-3. Diabetes 55,

8 2296 RESEARCH ARTICLE Development 137 (14) Madden, M. E. and Sarras, M. P., Jr (1989). The pancreatic ductal system of the rat: cell diversity, ultrastructure, and innervation. Pancreas 4, Means, A. L., Meszoely, I. M., Suzuki, K., Miyamoto, Y., Rustgi, A. K., Coffey, R. J., Jr, Wright, C. V., Stoffers, D. A. and Leach, S. D. (2005). Pancreatic epithelial plasticity mediated by acinar cell transdifferentiation and generation of nestin-positive intermediates. Development 132, Minami, K., Okuno, M., Miyawaki, K., Okumachi, A., Ishizaki, K., Oyama, K., Kawaguchi, M., Ishizuka, N., Iwanaga, T. and Seino, S. (2005). Lineage tracing and characterization of insulin-secreting cells generated from adult pancreatic acinar cells. Proc. Natl. Acad. Sci. USA 102, Nir, T., Melton, D. A. and Dor, Y. (2007). Recovery from diabetes in mice by beta cell regeneration. J. Clin. Invest. 117, Phillips, J. M., O Reilly, L., Bland, C., Foulis, A. K. and Cooke, A. (2007). Patients with chronic pancreatitis have islet progenitor cells in their ducts, but reversal of overt diabetes in NOD mice by anti-cd3 shows no evidence for islet regeneration. Diabetes 56, Postic, C., Shiota, M., Niswender, K. D., Jetton, T. L., Chen, Y., Moates, J. M., Shelton, K. D., Lindner, J., Cherrington, A. D. and Magnuson, M. A. (1999). Dual roles for glucokinase in glucose homeostasis as determined by liver and pancreatic beta cell-specific gene knock-outs using Cre recombinase. J. Biol. Chem. 274, Ray, K. C., Blaine, S. A., Washington, M. K., Braun, A. H., Singh, A. B., Harris, R. C., Harding, P. A., Coffey, R. J. and Means, A. L. (2009). Transmembrane and soluble isoforms of heparin-binding epidermal growth factor-like growth factor regulate distinct processes in the pancreas. Gastroenterology 137, Rooman, I., Heremans, Y., Heimberg, H. and Bouwens, L. (2000). Modulation of rat pancreatic acinoductal transdifferentiation and expression of PDX-1 in vitro. Diabetologia 43, Sandgren, E. P., Luetteke, N. C., Palmiter, R. D., Brinster, R. L. and Lee, D. C. (1990). Overexpression of TGF alpha in transgenic mice: induction of epithelial hyperplasia, pancreatic metaplasia, and carcinoma of the breast. Cell 61, Sharma, A., Zangen, D. H., Reitz, P., Taneja, M., Lissauer, M. E., Miller, C. P., Weir, G. C., Habener, J. F. and Bonner-Weir, S. (1999). The homeodomain protein IDX-1 increases after an early burst of proliferation during pancreatic regeneration. Diabetes 48, Solar, M., Cardalda, C., Houbracken, I., Martin, 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, Song, S. Y., Gannon, M., Washington, M. K., Scoggins, C. R., Meszoely, I. M., Goldenring, J. R., Marino, C. R., Sandgren, E. P., Coffey, R. J., Jr, Wright, C. V. et al. (1999). Expansion of Pdx1-expressing pancreatic epithelium and islet neogenesis in transgenic mice overexpressing transforming growth factor alpha. Gastroenterology 117, Soriano, P. (1999). Generalized lacz expression with the ROSA26 Cre reporter strain. Nat. Genet. 21, Srinivas, S., Watanabe, T., Lin, C. S., William, C. M., Tanabe, Y., Jessell, T. M. and Costantini, F. (2001). Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus. BMC Dev. Biol. 1, 4. Strobel, O., Dor, Y., Alsina, J., Stirman, A., Lauwers, G., Trainor, A., Castillo, C. F., Warshaw, A. L. and Thayer, S. P. (2007). In vivo lineage tracing defines the role of acinar-to-ductal transdifferentiation in inflammatory ductal metaplasia. Gastroenterology 133, Suarez-Pinzon, W. L., Lakey, J. R., Brand, S. J. and Rabinovitch, A. (2005a). Combination therapy with epidermal growth factor and gastrin induces neogenesis of human islet {beta}-cells from pancreatic duct cells and an increase in functional {beta}-cell mass. J. Clin. Endocrinol. Metab. 90, Suarez-Pinzon, W. L., Yan, Y., Power, R., Brand, S. J. and Rabinovitch, A. (2005b). Combination therapy with epidermal growth factor and gastrin increases beta-cell mass and reverses hyperglycemia in diabetic NOD mice. Diabetes 54, Teta, M., Rankin, M. M., Long, S. Y., Stein, G. M. and Kushner, J. A. (2007). Growth and regeneration of adult beta cells does not involve specialized progenitors. Dev. Cell 12, Tokui, Y., Kozawa, J., Yamagata, K., Zhang, J., Ohmoto, H., Tochino, Y., Okita, K., Iwahashi, H., Namba, M., Shimomura, I. et al. (2006). Neogenesis and proliferation of beta-cells induced by human betacellulin gene transduction via retrograde pancreatic duct injection of an adenovirus vector. Biochem. Biophys. Res. Commun. 350, Wang, R. N., Kloppel, G. and Bouwens, L. (1995). Duct- to islet-cell differentiation and islet growth in the pancreas of duct-ligated adult rats. Diabetologia 38, Wang, S., Jensen, J. N., Seymour, P. A., Hsu, W., Dor, Y., Sander, M., Magnuson, M. A., Serup, P. and Gu, G. (2009). Sustained Neurog3 expression in hormone-expressing islet cells is required for endocrine maturation and function. Proc. Natl. Acad. Sci. USA 106, Xu, X., D Hoker, J., Stange, G., Bonne, S., De Leu, N., Xiao, X., Van de Casteele, M., Mellitzer, G., Ling, Z., Pipeleers, D. et al. (2008). Beta cells can be generated from endogenous progenitors in injured adult mouse pancreas. Cell 132, Zhang, H., Fujitani, Y., Wright, C. V. and Gannon, M. (2005). Efficient recombination in pancreatic islets by a tamoxifen-inducible Cre-recombinase. Genesis 42, Zhou, Q., Brown, J., Kanarek, A., Rajagopal, J. and Melton, D. A. (2008). In vivo reprogramming of adult pancreatic exocrine cells to beta-cells. Nature 455,

Analysis of the plasticity of duct cells in the embryonic and adult pancreas. Myriam Solar Hospital Clinic de Barcelona Barcelona 2009

Analysis of the plasticity of duct cells in the embryonic and adult pancreas. Myriam Solar Hospital Clinic de Barcelona Barcelona 2009 Analysis of the plasticity of duct cells in the embryonic and adult pancreas Myriam Solar Hospital Clinic de Barcelona Barcelona 2009 Major periods of formation of new β-cells: 1- Embryogenesis (secondary

More information

Impact of Sox9 Dosage and Hes1-mediated Notch Signaling in Controlling the Plasticity of Adult Pancreatic Duct Cells in Mice

Impact of Sox9 Dosage and Hes1-mediated Notch Signaling in Controlling the Plasticity of Adult Pancreatic Duct Cells in Mice Impact of Sox9 Dosage and Hes1-mediated Notch Signaling in Controlling the Plasticity of Adult Pancreatic Duct Cells in Mice Shinichi Hosokawa 1,3,Kenichiro Furuyama 1,3, Masashi Horiguchi 1,3,Yoshiki

More information

SUPPLEMENTARY DATA. Supplementary Table 2. Antibodies used for Immunofluoresence. Supplementary Table 3. Real-time PCR primer sequences.

SUPPLEMENTARY DATA. Supplementary Table 2. Antibodies used for Immunofluoresence. Supplementary Table 3. Real-time PCR primer sequences. Supplementary Table 2. Antibodies used for Immunofluoresence. Antibody Dilution Source Goat anti-pdx1 1:100 R&D Systems Rabbit anti-hnf6 1:100 Santa Cruz Biotechnology Mouse anti-nkx6.1 1:200 Developmental

More information

Acinar cells in the neonatal pancreas grow by self-duplication and not by neogenesis from duct cells Houbracken, Isabelle; Bouwens, Luc

Acinar cells in the neonatal pancreas grow by self-duplication and not by neogenesis from duct cells Houbracken, Isabelle; Bouwens, Luc Vrije Universiteit Brussel Acinar cells in the neonatal pancreas grow by self-duplication and not by neogenesis from duct cells Houbracken, Isabelle; Bouwens, Luc Published in: Scientific Reports DOI:

More information

Preexisting pancreatic acinar cells contribute to acinar cell, but not islet β cell, regeneration

Preexisting pancreatic acinar cells contribute to acinar cell, but not islet β cell, regeneration Related Commentary, page 859 Research article Preexisting pancreatic acinar cells contribute to acinar cell, but not islet β cell, regeneration Biva M. Desai, 1 Jennifer Oliver-Krasinski, 1 Diva D. De

More information

Quantification of early stage lesions for loss of p53 should be shown in the main figures.

Quantification of early stage lesions for loss of p53 should be shown in the main figures. Reviewer #1 (Remarks to the Author): Expert in prostate cancer The manuscript "Clonal dynamics following p53 loss of heterozygosity in Kras-driven cancers" uses a number of novel genetically engineered

More information

Primary Explant Cultures of Adult and Embryonic Pancreas

Primary Explant Cultures of Adult and Embryonic Pancreas Primary Explant Cultures of Pancreas 259 17 Primary Explant Cultures of Adult and Embryonic Pancreas Farzad Esni, Yoshiharu Miyamoto, Steven D. Leach, and Bidyut Ghosh Summary The developmental plasticity

More information

Persistent expression of PDX-1 in the pancreas causes acinar-to-ductal metaplasia through Stat3 activation

Persistent expression of PDX-1 in the pancreas causes acinar-to-ductal metaplasia through Stat3 activation RESEARCH COMMUNICATION Persistent expression of PDX-1 in the pancreas causes acinar-to-ductal metaplasia through Stat3 activation Takeshi Miyatsuka, 1,6 Hideaki Kaneto, 1 Toshihiko Shiraiwa, 1 Taka-aki

More information

Cell Therapy for Diabetes: Generating Functional Islets from Human Exocrine Tissue

Cell Therapy for Diabetes: Generating Functional Islets from Human Exocrine Tissue Cell Therapy for Diabetes: Generating Functional Islets from Human Exocrine Tissue Kevin Docherty University of Aberdeen ELRIG Drug Discovery 2016 ACC Liverpool 14 th October 2016 Autoimmune destruction

More information

For almost a century (for historical review, see 1)

For almost a century (for historical review, see 1) PERSPECTIVES IN DIABETES -Cell Growth and Regeneration: Replication Is Only Part of the Story Susan Bonner-Weir, Wan-Chun Li, Limor Ouziel-Yahalom, Lili Guo, Gordon C. Weir, and Arun Sharma I am accordingly

More information

Article. Reference. Progenitor cell domains in the developing and adult pancreas. KOPP, Janel L, et al.

Article. Reference. Progenitor cell domains in the developing and adult pancreas. KOPP, Janel L, et al. Article Progenitor cell domains in the developing and adult pancreas KOPP, Janel L, et al. Abstract Unlike organs with defined stem cell compartments, such as the intestine, the pancreas has limited capacity

More information

Mice( Dissertation_ 全文 ) https://doi.org/ /doctor.k19

Mice( Dissertation_ 全文 ) https://doi.org/ /doctor.k19 Impact of Sox9 Dosage and Hes1-medi TitleControlling the Plasticity of Adult Mice( Dissertation_ 全文 ) Author(s) Hosokawa, Shinichi Citation Kyoto University ( 京都大学 ) Issue Date 2015-07-23 URL https://doi.org/10.14989/doctor.k19

More information

DEVELOPMENT. Shelley B. Nelson, Ashleigh E. Schaffer and Maike Sander*

DEVELOPMENT. Shelley B. Nelson, Ashleigh E. Schaffer and Maike Sander* RESEARCH ARTICLE 2491 Development 134, 2491-2500 (2007) doi:10.1242/dev.002691 The transcription factors Nkx6.1 and Nkx6.2 possess equivalent activities in promoting beta-cell fate specification in Pdx1

More information

Preferential reduction of β cells derived from Pax6 MafB pathway in MafB deficient mice

Preferential reduction of β cells derived from Pax6 MafB pathway in MafB deficient mice Available online at www.sciencedirect.com Developmental Biology 314 (2008) 443 456 www.elsevier.com/developmentalbiology Genomes & Developmental Control Preferential reduction of β cells derived from Pax6

More information

Generating Mouse Models of Pancreatic Cancer

Generating Mouse Models of Pancreatic Cancer Generating Mouse Models of Pancreatic Cancer Aom Isbell http://www2.massgeneral.org/cancerresourceroom/types/gi/index.asp Spring/Summer 1, 2012 Alexandros Tzatsos, MD PhD Bardeesy Lab: Goals and Objectives

More information

Identification of Sox9-Dependent Acinar-to-Ductal Reprogramming as the Principal Mechanism for Initiation of Pancreatic Ductal Adenocarcinoma

Identification of Sox9-Dependent Acinar-to-Ductal Reprogramming as the Principal Mechanism for Initiation of Pancreatic Ductal Adenocarcinoma Article Identification of Sox9-Dependent Acinar-to-Ductal Reprogramming as the Principal Mechanism for Initiation of Pancreatic Ductal Adenocarcinoma Janel L. Kopp, 1,6 Guido von Figura, 2,6 Erin Mayes,

More information

Lineage tracing and characterization of insulin-secreting cells generated from adult pancreatic acinar cells

Lineage tracing and characterization of insulin-secreting cells generated from adult pancreatic acinar cells Lineage tracing and characterization of insulin-secreting cells generated from adult pancreatic acinar cells Kohtaro Minami*, Masaaki Okuno*, Kazumasa Miyawaki*, Akinori Okumachi*, Katsuhiko Ishizaki,

More information

a) Primary cultures derived from the pancreas of an 11-week-old Pdx1-Cre; K-MADM-p53

a) Primary cultures derived from the pancreas of an 11-week-old Pdx1-Cre; K-MADM-p53 1 2 3 4 5 6 7 8 9 10 Supplementary Figure 1. Induction of p53 LOH by MADM. a) Primary cultures derived from the pancreas of an 11-week-old Pdx1-Cre; K-MADM-p53 mouse revealed increased p53 KO/KO (green,

More information

Pharmacological induction of pancreatic islet cell transdifferentiation: relevance to type I diabetes

Pharmacological induction of pancreatic islet cell transdifferentiation: relevance to type I diabetes OPEN Citation: (2014) 5, e; doi:10.1038/cddis.2014.311 & 2014 Macmillan Publishers Limited All rights reserved 2041-4889/14 www.nature.com/cddis Pharmacological induction of pancreatic islet cell transdifferentiation:

More information

CURRICULUM VITAE. Anna Lois Means, PhD

CURRICULUM VITAE. Anna Lois Means, PhD CURRICULUM VITAE Anna Lois Means, PhD PERSONAL DATA Work address: Anna L. Means Division of Surgical Oncology 10445 MRB IV 2213 Garland Ave. Vanderbilt University Medical Center Nashville, TN 37232-0443

More information

The differentiation potential of stem cells and precursors

The differentiation potential of stem cells and precursors STEM CELLS AND DEVELOPMENT Volume 22, Number 15, 2013 Ó Mary Ann Liebert, Inc. DOI: 10.1089/scd.2013.0027 ORIGINAL RESEARCH REPORT The Adult Mammalian Pancreas Contains Separate Precursors of Pancreatic

More information

NIH Public Access Author Manuscript Diabetologia. Author manuscript; available in PMC 2014 February 01.

NIH Public Access Author Manuscript Diabetologia. Author manuscript; available in PMC 2014 February 01. NIH Public Access Author Manuscript Published in final edited form as: Diabetologia. 2013 February ; 56(2): 231 233. doi:10.1007/s00125-012-2788-6. Lipotoxicity impairs incretin signalling V. Poitout 1,2

More information

PANCREATIC BETA CELLS PRODUCE AND SECRETE

PANCREATIC BETA CELLS PRODUCE AND SECRETE 15 March, 2018 PANCREATIC BETA CELLS PRODUCE AND SECRETE Document Filetype: PDF 374.06 KB 0 PANCREATIC BETA CELLS PRODUCE AND SECRETE Among the oldest and cheapest drugs for diabetes are the drugs that

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION doi:10.1038/nature10188 Supplementary Figure 1. Embryonic epicardial genes are down-regulated from midgestation stages and barely detectable post-natally. Real time qrt-pcr revealed a significant down-regulation

More information

Supplementary Figure 1

Supplementary Figure 1 Supplementary Figure 1 Control Pancreatitis Supplementary Figure 2 A Panc Liver SI Spleen H 2 O B EZH2 fl/fl C EZH2 fl/fl 37bp EZH2 ERK2 D E 5 EZH2 fl/fl Fasting Glucose (mg/dl) 2 18 16 14 12 1 8 6 4 2

More information

Direct evidence for the pancreatic lineage: NGN3+ cells are islet progenitors and are distinct from duct progenitors

Direct evidence for the pancreatic lineage: NGN3+ cells are islet progenitors and are distinct from duct progenitors Development 129, 2447-2457 (2002) Printed in Great Britain The Company of Biologists Limited 2002 DEV3582 2447 Direct evidence for the pancreatic lineage: NGN3+ cells are islet progenitors and are distinct

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION DOI: 10.1038/ncb2697 Figure S1 Cytokeratin 5 is a specific marker for basal and intermediate cells in all mouse prostate lobes. (a) Immunofluorescence staining showing co-localization of YFP with p63 in

More information

Figure legends Supplemental Fig.1. Glucose-induced insulin secretion and insulin content of islets. Supplemental Fig. 2.

Figure legends Supplemental Fig.1. Glucose-induced insulin secretion and insulin content of islets. Supplemental Fig. 2. Figure legends Supplemental Fig.. Glucose-induced insulin secretion and insulin content of islets. Insulin secretory responses to.,., and. mm glucose (A) (n = 7-), and the insulin content in the islets

More information

Induction of mouse pancreatic ductal differentiation, an in vitro assay

Induction of mouse pancreatic ductal differentiation, an in vitro assay In Vitro Cell.Dev.Biol. Animal DOI 10.1007/s11626-012-9555-3 Induction of mouse pancreatic ductal differentiation, an in vitro assay Julie A. Rhodes & Angela Criscimanna & Farzad Esni Received: 5 July

More information

Development of the Pancreas and Response to Disease

Development of the Pancreas and Response to Disease Chapter 1 Development of the Pancreas and Response to Disease The pancreas develops from the primitive gut, which is derived from endoderm. An interdependent series of signals is necessary to form the

More information

Recent data indicate that stem cells exist in many

Recent data indicate that stem cells exist in many Label-Retaining Cells in the Rat Pancreas Location and Differentiation Potential in Vitro Bertrand Duvillié, 1 Myriam Attali, 1 Virginie Aiello, 1 Eric Quemeneur, 2 and Raphael Scharfmann 1 Islets of Langerhans

More information

Review Article Development and Regeneration in the Endocrine Pancreas

Review Article Development and Regeneration in the Endocrine Pancreas International Scholarly Research Network ISRN Endocrinology Volume 2012, Article ID 640956, 12 pages doi:10.5402/2012/640956 Review Article Development and Regeneration in the Endocrine Pancreas Ahmed

More information

Supplemental Figure 1. Intracranial transduction of a modified ptomo lentiviral vector in the mouse

Supplemental Figure 1. Intracranial transduction of a modified ptomo lentiviral vector in the mouse Supplemental figure legends Supplemental Figure 1. Intracranial transduction of a modified ptomo lentiviral vector in the mouse hippocampus targets GFAP-positive but not NeuN-positive cells. (A) Stereotaxic

More information

Material and Methods Production and analysis of -gal+ clones Immunostaining Optical projection tomography Statistical analysis

Material and Methods Production and analysis of -gal+ clones Immunostaining Optical projection tomography Statistical analysis Material and Methods Production and analysis of β-gal+ clones The α-cardiac actin nlaacz/+ and Cx40 egfp mouse lines used in this study were genotyped as previously reported 1, 2. Double transgenic knock-in

More information

Islets of Langerhans consist mostly of insulin-producing β cells. They will appear to be densely labeled

Islets of Langerhans consist mostly of insulin-producing β cells. They will appear to be densely labeled Are adult pancreatic beta cells formed by self-duplication or stem cell differentiation? Introduction Researchers have long been interested in how tissues produce and maintain the correct number of cells

More information

Chapter V.2. Islet Neogenesis: A Possible Pathway for Beta-Cell Replenishment

Chapter V.2. Islet Neogenesis: A Possible Pathway for Beta-Cell Replenishment Reprint from The Review of DIABETIC STUDIES Vol 9 No 4 2012 Special Issue Immunology and Treatment of T1D The Review of DIABETIC STUDIES Chapter V.2 Islet Neogenesis: A Possible Pathway for Beta-Cell Replenishment

More information

Ductal pancreatic cancer modeling and drug screening using human pluripotent stem cell and patient-derived tumor organoids

Ductal pancreatic cancer modeling and drug screening using human pluripotent stem cell and patient-derived tumor organoids Ductal pancreatic cancer modeling and drug screening using human pluripotent stem cell and patient-derived tumor organoids Ling Huang 1, Audrey Holtzinger 1,2,11, Ishaan Jagan 1,11, Michael BeGora 1, Ines

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION b 350 300 250 200 150 100 50 0 E0 E10 E50 E0 E10 E50 E0 E10 E50 E0 E10 E50 Number of organoids per well 350 300 250 200 150 100 50 0 R0 R50 R100 R500 1st 2nd 3rd Noggin 100 ng/ml Noggin 10 ng/ml Noggin

More information

β cell replication is the primary mechanism for maintaining postnatal β cell mass

β cell replication is the primary mechanism for maintaining postnatal β cell mass Research article β cell replication is the primary mechanism for maintaining postnatal β cell mass Senta Georgia and Anil Bhushan Department of Biochemistry and Molecular Biology, University of Southern

More information

Origin of exocrine pancreatic cells from nestin-positive precursors in developing mouse pancreas

Origin of exocrine pancreatic cells from nestin-positive precursors in developing mouse pancreas Mechanisms of Development 121 (2004) 15 25 www.elsevier.com/locate/modo Origin of exocrine pancreatic cells from nestin-positive precursors in developing mouse pancreas Farzad Esni a, Doris A. Stoffers

More information

Stem cells: units of development and regeneration. Fernando D. Camargo Ph.D. Whitehead Fellow Whitehead Institute for Biomedical Research.

Stem cells: units of development and regeneration. Fernando D. Camargo Ph.D. Whitehead Fellow Whitehead Institute for Biomedical Research. Stem cells: units of development and regeneration Fernando D. Camargo Ph.D. Whitehead Fellow Whitehead Institute for Biomedical Research Concepts 1. Embryonic vs. adult stem cells 2. Hematopoietic stem

More information

Pancreas-specific deletion of mouse Gata4 and Gata6 causes pancreatic agenesis

Pancreas-specific deletion of mouse Gata4 and Gata6 causes pancreatic agenesis Research article Related Commentary, page 3469 Pancreas-specific deletion of mouse Gata4 and Gata6 causes pancreatic agenesis Shouhong Xuan, 1 Matthew J. Borok, 1 Kimberly J. Decker, 2 Michele A. Battle,

More information

(A) PCR primers (arrows) designed to distinguish wild type (P1+P2), targeted (P1+P2) and excised (P1+P3)14-

(A) PCR primers (arrows) designed to distinguish wild type (P1+P2), targeted (P1+P2) and excised (P1+P3)14- 1 Supplemental Figure Legends Figure S1. Mammary tumors of ErbB2 KI mice with 14-3-3σ ablation have elevated ErbB2 transcript levels and cell proliferation (A) PCR primers (arrows) designed to distinguish

More information

The pancreas mainly consists of exocrine tissue, which

The pancreas mainly consists of exocrine tissue, which GASTROENTEROLOGY 2011;141:731 741 Lineage Tracing Evidence for Transdifferentiation of Acinar to Duct Cells and Plasticity of Human Pancreas ISABELLE HOUBRACKEN,* EVELIEN DE WAELE,* JESSY LARDON,* ZHIDONG

More information

hexahistidine tagged GRP78 devoid of the KDEL motif (GRP78-His) on SDS-PAGE. This

hexahistidine tagged GRP78 devoid of the KDEL motif (GRP78-His) on SDS-PAGE. This SUPPLEMENTAL FIGURE LEGEND Fig. S1. Generation and characterization of. (A) Coomassie staining of soluble hexahistidine tagged GRP78 devoid of the KDEL motif (GRP78-His) on SDS-PAGE. This protein was expressed

More information

Ahtiainen et al., http :// /cgi /content /full /jcb /DC1

Ahtiainen et al., http ://  /cgi /content /full /jcb /DC1 Supplemental material JCB Ahtiainen et al., http ://www.jcb.org /cgi /content /full /jcb.201512074 /DC1 THE JOURNAL OF CELL BIOLOGY Figure S1. Distinct distribution of different cell cycle phases in the

More information

Pleiotropic Roles of PDX-1 in the Pancreas. Hideaki Kaneto, Takeshi Miyatsuka, Dan Kawamori and Taka-aki Matsuoka

Pleiotropic Roles of PDX-1 in the Pancreas. Hideaki Kaneto, Takeshi Miyatsuka, Dan Kawamori and Taka-aki Matsuoka REVIEW Pleiotropic Roles of PDX-1 in the Pancreas Hideaki Kaneto, Takeshi Miyatsuka, Dan Kawamori and Taka-aki Matsuoka Department of Internal Medicine and Therapeutics (A8), Osaka University Graduate

More information

Animal Tissue Culture SQG 3242 Biology of Cultured Cells. Dr. Siti Pauliena Mohd Bohari

Animal Tissue Culture SQG 3242 Biology of Cultured Cells. Dr. Siti Pauliena Mohd Bohari Animal Tissue Culture SQG 3242 Biology of Cultured Cells Dr. Siti Pauliena Mohd Bohari The Culture Environment Changes of Cell s microenvironment needed that favor the spreading, migration, and proliferation

More information

Sox9: A Master Regulator of the Pancreatic Program. Philip A. Seymour

Sox9: A Master Regulator of the Pancreatic Program. Philip A. Seymour Reprint from The Review of DIABETIC STUDIES Vol 11 No 1 2014 Special Edition Stem Cells and Pancreas Regeneration The Review of DIABETIC STUDIES Sox9: A Master Regulator of the Pancreatic Program Philip

More information

Supplemental Information. Otic Mesenchyme Cells Regulate. Spiral Ganglion Axon Fasciculation. through a Pou3f4/EphA4 Signaling Pathway

Supplemental Information. Otic Mesenchyme Cells Regulate. Spiral Ganglion Axon Fasciculation. through a Pou3f4/EphA4 Signaling Pathway Neuron, Volume 73 Supplemental Information Otic Mesenchyme Cells Regulate Spiral Ganglion Axon Fasciculation through a Pou3f4/EphA4 Signaling Pathway Thomas M. Coate, Steven Raft, Xiumei Zhao, Aimee K.

More information

Supplementary Table 1. The primers used for quantitative RT-PCR. Gene name Forward (5 > 3 ) Reverse (5 > 3 )

Supplementary Table 1. The primers used for quantitative RT-PCR. Gene name Forward (5 > 3 ) Reverse (5 > 3 ) 770 771 Supplementary Table 1. The primers used for quantitative RT-PCR. Gene name Forward (5 > 3 ) Reverse (5 > 3 ) Human CXCL1 GCGCCCAAACCGAAGTCATA ATGGGGGATGCAGGATTGAG PF4 CCCCACTGCCCAACTGATAG TTCTTGTACAGCGGGGCTTG

More information

α-cell role in β-cell generation and regeneration

α-cell role in β-cell generation and regeneration Islets ISSN: 1938-2014 (Print) 1938-2022 (Online) Journal homepage: http://www.tandfonline.com/loi/kisl20 α-cell role in β-cell generation and regeneration Joel F. Habener & Violeta Stanojevic To cite

More information

Nature Neuroscience: doi: /nn Supplementary Figure 1. MADM labeling of thalamic clones.

Nature Neuroscience: doi: /nn Supplementary Figure 1. MADM labeling of thalamic clones. Supplementary Figure 1 MADM labeling of thalamic clones. (a) Confocal images of an E12 Nestin-CreERT2;Ai9-tdTomato brain treated with TM at E10 and stained for BLBP (green), a radial glial progenitor-specific

More information

Pancreatic intraepithelial

Pancreatic intraepithelial Pancreatic intraepithelial neoplasia (PanIN) Markéta Hermanová St. Anne s University Hospital Brno Faculty of Medicine, Masaryk University Precursor lesions of invasive pancreatic cancer Pancreatic intraepithelial

More information

Stem Cells and The Endometrium. Director, Division of Reproductive Endocrinology and infertility

Stem Cells and The Endometrium. Director, Division of Reproductive Endocrinology and infertility Stem Cells and The Endometrium Hugh S. Taylor, M.D. Director, Division of Reproductive Endocrinology and infertility Nothing to disclose Stem Cells Cells that are capable of both self-renewal and have

More information

Genetic Control of -Cell Mass Homeostasis

Genetic Control of -Cell Mass Homeostasis The Open Endocrinology Journal, 2010, 4, 11-24 11 Genetic Control of -Cell Mass Homeostasis Open Access Mangala M. Soundarapandian 1, Maria L. Nieves 1, Roxane Pasquier 1, Ulrika Bergström 1, Mark A. Atkinson

More information

MPB333:Molecular Endocrinology of Obesity and Diabetes

MPB333:Molecular Endocrinology of Obesity and Diabetes MPB333:Molecular Endocrinology of Obesity and Diabetes The Use of Stem Cells as a Cure for Type 1 Diabetes January 15, 2010 Trish Labosky 9415C MRBIV trish.labosky@vanderbilt.edu In theory. 2. ~Easy and

More information

Control of Glucose Metabolism

Control of Glucose Metabolism Glucose Metabolism Control of Glucose Metabolism The pancreas is both an exocrine and endocrine gland. It secretes digestive enzymes into the duodenum (exocrine) and 3 specific hormones into the bloodstream

More information

Supplemental Information. b Cell Aging Markers Have Heterogeneous. Distribution and Are Induced by Insulin Resistance

Supplemental Information. b Cell Aging Markers Have Heterogeneous. Distribution and Are Induced by Insulin Resistance Cell Metabolism, Volume 25 Supplemental Information b Cell Aging Markers Have Heterogeneous Distribution and Are Induced by Insulin Resistance Cristina Aguayo-Mazzucato, Mark van Haaren, Magdalena Mruk,

More information

Research, University of California San Francisco, San Francisco, CA , USA *senior authors

Research, University of California San Francisco, San Francisco, CA , USA *senior authors REPROGRAMMING OF HUMAN PANCREATIC EXOCRINE CELLS TO BETA-LIKE CELLS Marie Lemper 1, Gunter Leuckx 1, Yves Heremans 1, Michael S. German 2, Harry Heimberg 1, Luc Bouwens 1,* and Luc Baeyens 1,2,* 1 Diabetes

More information

Biochemical and Biophysical Research Communications

Biochemical and Biophysical Research Communications Biochemical and Biophysical Research Communications 399 (2010) 440 445 Contents lists available at ScienceDirect Biochemical and Biophysical Research Communications journal homepage: www.elsevier.com/locate/ybbrc

More information

Supplementary Materials. for Garmy-Susini, et al, Integrin 4 1 signaling is required for lymphangiogenesis and tumor metastasis

Supplementary Materials. for Garmy-Susini, et al, Integrin 4 1 signaling is required for lymphangiogenesis and tumor metastasis Supplementary Materials for Garmy-Susini, et al, Integrin 4 1 signaling is required for lymphangiogenesis and tumor metastasis 1 Supplementary Figure Legends Supplementary Figure 1: Integrin expression

More information

Fig. S1. Generation of pancreas-specific Stard13 PA-deleted mice. (A) Schematic representation of the targeted Stard13 flox allele, indicating the

Fig. S1. Generation of pancreas-specific Stard13 PA-deleted mice. (A) Schematic representation of the targeted Stard13 flox allele, indicating the Fig. S1. Generation of pancreas-specific Stard13 PA-deleted mice. (A) Schematic representation of the targeted Stard13 flox allele, indicating the loxp sites (in pink) flanking exon 5 of the gene, FRT-flanked-Hygromicin,

More information

Diabetic pdx1-mutant zebrafish show conserved responses to nutrient overload and anti-glycemic treatment

Diabetic pdx1-mutant zebrafish show conserved responses to nutrient overload and anti-glycemic treatment Supplementary Information Diabetic pdx1-mutant zebrafish show conserved responses to nutrient overload and anti-glycemic treatment Robin A. Kimmel, Stefan Dobler, Nicole Schmitner, Tanja Walsen, Julia

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION SUPPLEMENTARY INFORMATION doi:1.138/nature11463 %Sox17(+) 9 8 7 6 5 4 3 2 1 %Sox17(+) #Sox17(+) d2 d4 d6 d8 d1 d12 d14 d18 25 2 15 1 5 Number of Sox17(+) cells X 1 Supplementary Figure 1: Expression of

More information

Successful transplantation of human pancreatic

Successful transplantation of human pancreatic Original Article Combination Therapy With Epidermal Growth Factor and Gastrin Increases -Cell Mass and Reverses Hyperglycemia in Diabetic NOD Mice Wilma L. Suarez-Pinzon, 1 Yanhua Yan, 2 Robert Power,

More information

Ultrastructural and immunohistochemical analysis of the 8-20 week human fetal pancreas

Ultrastructural and immunohistochemical analysis of the 8-20 week human fetal pancreas Islets 6:4, e982949; November 1, 2014; 2014 Published with license by Taylor & Francis Group, LLC RESEARCH PAPER Ultrastructural and immunohistochemical analysis of the 8-20 week human fetal pancreas Matthew

More information

Supplemental Figure 1

Supplemental Figure 1 Supplemental Figure 1 A S100A4: SFLGKRTDEAAFQKLMSNLDSNRDNEVDFQEYCVFLSCIAMMCNEFFEGFPDK Overlap: SF G DE KLM LD N D VDFQEY VFL I M N FF G PD S100A2: SFVGEKVDEEGLKKLMGSLDENSDQQVDFQEYAVFLALITVMCNDFFQGCPDR

More information

GATA4 and GATA6 control mouse pancreas organogenesis

GATA4 and GATA6 control mouse pancreas organogenesis GATA4 and GATA6 control mouse pancreas organogenesis Manuel Carrasco,, Francisco Martín, Anabel Rojas J Clin Invest. 2012;122(10):3504-3515. https://doi.org/10.1172/jci63240. Research Article Development

More information

Origin and Development of the Precursor Lesions in Experimental Pancreatic Cancer in Rats

Origin and Development of the Precursor Lesions in Experimental Pancreatic Cancer in Rats 0023-6837/03/8306-853$03.00/0 LABORATORY INVESTIGATION Vol. 83, No. 6, p. 853, 2003 Copyright 2003 by The United States and Canadian Academy of Pathology, Inc. Printed in U.S.A. Origin and Development

More information

Controlled induction of human pancreatic progenitors produces functional beta-like cells in vitro.

Controlled induction of human pancreatic progenitors produces functional beta-like cells in vitro. Manuscript EMBO-2015-91058 Controlled induction of human pancreatic progenitors produces functional beta-like cells in vitro. Holger A Russ, Audrey V Parent, Jennifer J Ringler, Thomas G Hennings, Gopika

More information

Meeting Report. From December 8 to 11, 2012 at Atlanta, GA, U.S.A

Meeting Report. From December 8 to 11, 2012 at Atlanta, GA, U.S.A Meeting Report Affiliation Department of Transfusion Medicine and Cell Therapy Name Hisayuki Yao Name of the meeting Period and venue Type of your presentation Title of your presentation The 54 th Annual

More information

Rgs16 and Rgs8 in embryonic endocrine pancreas and mouse models of diabetes

Rgs16 and Rgs8 in embryonic endocrine pancreas and mouse models of diabetes First posted online on 8 July 2010 as 10.1242/dmm.003210 Access the most recent version at http://dmm.biologists.org/lookup/doi/10.1242/dmm.003210 Disease Models & Mechanisms 3, 000-000 (2010) doi:10.1242/dmm.003210

More information

Supplementary Figure 1: GFAP positive nerves in patients with adenocarcinoma of

Supplementary Figure 1: GFAP positive nerves in patients with adenocarcinoma of SUPPLEMENTARY FIGURES AND MOVIE LEGENDS Supplementary Figure 1: GFAP positive nerves in patients with adenocarcinoma of the pancreas. (A) Images of nerves stained for GFAP (green), S100 (red) and DAPI

More information

Supporting Information

Supporting Information Supporting Information Guney et al. 1.173/pnas.117218 SI Materials and Methods PCR and Genotyping. Genotyping of CTGF e2coin/e2coin mice was performed by PCR on N isolated from ear punches using the following

More information

Gastric PDX-1 expression in pancreatic metaplasia and endocrine cell hyperplasia in atrophic corpus gastritis

Gastric PDX-1 expression in pancreatic metaplasia and endocrine cell hyperplasia in atrophic corpus gastritis & 2004 USCAP, Inc All rights reserved 0893-3952/04 $25.00 www.modernpathology.org Gastric PDX-1 expression in pancreatic metaplasia and endocrine cell hyperplasia in atrophic corpus gastritis Maike Buettner

More information

Journal Club WS 2012/13 Stefanie Nickl

Journal Club WS 2012/13 Stefanie Nickl Journal Club WS 2012/13 Stefanie Nickl Background Mesenchymal Stem Cells First isolation from bone marrow 30 ys ago Isolation from: spleen, heart, skeletal muscle, synovium, amniotic fluid, dental pulp,

More information

Title Diabetes Caused by Elastase-Cre-Med in Mice Kodama, Sota; Nakano, Yasuhiro; Hir Author(s) Kenichiro; Horiguchi, Masashi; Kuha Toshihiko; Kawaguchi, Michiya; Gann Christopher V. E.; Uemoto, Shinji;

More information

Pancreatic neurogenin 3-expressing cells are unipotent islet precursors

Pancreatic neurogenin 3-expressing cells are unipotent islet precursors RESEARCH ARTICLE 3567 Development 136, 3567-3574 (2009) doi:10.1242/dev.039214 Pancreatic neurogenin 3-expressing cells are unipotent islet precursors Renaud Desgraz and Pedro L. Herrera* Pancreatic islet

More information

Models of Regeneration of Beta Cells in Vivo Ioana Ilie 1*, MD,PhD, Razvan Ilie 2, MD

Models of Regeneration of Beta Cells in Vivo Ioana Ilie 1*, MD,PhD, Razvan Ilie 2, MD Models of Regeneration of Beta Cells in Vivo Ioana Ilie 1*, MD,PhD, Razvan Ilie 2, MD Abstract - Due to the widespread increase in the number of subjects suffering from diabetes, our understanding of the

More information

Enterprise Interest Nothing to declare

Enterprise Interest Nothing to declare Enterprise Interest Nothing to declare Update of mixed tumours of the GI tract, the pancreas and the liver Introduction to the concept of mixed tumours and clinical implication Jean-Yves SCOAZEC Surgical

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION DOI: 10.1038/ncb2610 Figure S1 FSMCs derived from MSLN CLN transgenic mice express smooth muscle-specific proteins. Beta-galactosidase is ubiquitously expressed within cultured FSMCs derived from MSLN

More information

EGF receptor in pancreatic β-cell mass regulation

EGF receptor in pancreatic β-cell mass regulation 280 Biochemical Society Transactions (2008) Volume 36, part 3 EGF receptor in pancreatic β-cell mass regulation Päivi Miettinen, Päivi Ormio, Elina Hakonen, Meenal Banerjee and Timo Otonkoski 1 Hospital

More information

Supplementary Figure 1: Hsp60 / IEC mice are embryonically lethal (A) Light microscopic pictures show mouse embryos at developmental stage E12.

Supplementary Figure 1: Hsp60 / IEC mice are embryonically lethal (A) Light microscopic pictures show mouse embryos at developmental stage E12. Supplementary Figure 1: Hsp60 / IEC mice are embryonically lethal (A) Light microscopic pictures show mouse embryos at developmental stage E12.5 and E13.5 prepared from uteri of dams and subsequently genotyped.

More information

Journal Club. 03/04/2012 Lama Nazzal

Journal Club. 03/04/2012 Lama Nazzal Journal Club 03/04/2012 Lama Nazzal NOTCH and the kidneys Is an evolutionarily conserved cell cell communication mechanism. Is a regulator of cell specification, differentiation, and tissue patterning.

More information

NEW IHC A n t i b o d i e s

NEW IHC A n t i b o d i e s NEW IHC Antibodies TABLE OF CONTENTS NEW IHC ANTIBODIES from Cell Marque CITED1 (5H6).... 1 Claudin 7 (5D10F3).... 1 GATA1 (4F5).... 1 Transgelin (2A10C2).... 1 NEW IHC ANTIBODIES using RabMAb Technology

More information

Supplemental Data. Wnt/β-Catenin Signaling in Mesenchymal Progenitors. Controls Osteoblast and Chondrocyte

Supplemental Data. Wnt/β-Catenin Signaling in Mesenchymal Progenitors. Controls Osteoblast and Chondrocyte Supplemental Data Wnt/β-Catenin Signaling in Mesenchymal Progenitors Controls Osteoblast and Chondrocyte Differentiation during Vertebrate Skeletogenesis Timothy F. Day, Xizhi Guo, Lisa Garrett-Beal, and

More information

Pancreas Fox Chapter 18 part 2 (also Chapter 19.3 & 19.4)

Pancreas Fox Chapter 18 part 2 (also Chapter 19.3 & 19.4) Vert Phys PCB3743 Pancreas Fox Chapter 18 part 2 (also Chapter 19.3 & 19.4) T. Houpt, Ph.D. Anatomy of Digestive System Peristalsis Stomach and Acid Secretion Liver and Bile Secretion Pancreas and pancreatic

More information

Supplementary Table 1. Primer sequences for conventional RT-PCR on mouse islets

Supplementary Table 1. Primer sequences for conventional RT-PCR on mouse islets Supplementary Table 1. Primer sequences for conventional RT-PCR on mouse islets Gene 5 Forward 3 5 Reverse 3.T. Product (bp) ( C) mnox1 GTTCTTGGGCTGCCTTGG GCTGGGGCGGCGG 60 300 mnoxa1 GCTTTGCCGCGTGC GGTTCGGGTCCTTTGTGC

More information

Notch signaling in pancreatic cancer: oncogene or tumor suppressor?

Notch signaling in pancreatic cancer: oncogene or tumor suppressor? Review Notch signaling in pancreatic cancer: oncogene or tumor suppressor? Jacqueline L. Avila 1 and Joseph L. Kissil 2 1 Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia,

More information

Supplementary Table 1. Antibodies and dilutions used in the immunohistochemical study

Supplementary Table 1. Antibodies and dilutions used in the immunohistochemical study Supplementary Table 1. Antibodies and dilutions used in the immunohistochemical study Antigen Species Clone Source Dilution Insulin Guinea pig - Dako, Carpinteria, 1:200 Glucagon Rabbit - Dako, Carpinteria,

More information

THE ROLE OF CONNECTIVE TISSUE GROWTH FACTOR IN ISLET. Michelle Guney. Dissertation. Submitted to the Faculty of the

THE ROLE OF CONNECTIVE TISSUE GROWTH FACTOR IN ISLET. Michelle Guney. Dissertation. Submitted to the Faculty of the THE ROLE OF CONNECTIVE TISSUE GROWTH FACTOR IN ISLET MORPHOGENESIS AND β CELL PROLIFERATION By Michelle Guney Dissertation Submitted to the Faculty of the Graduate School of Vanderbilt University In partial

More information

Week 3 The Pancreas: Pancreatic ph buffering:

Week 3 The Pancreas: Pancreatic ph buffering: Week 3 The Pancreas: A gland with both endocrine (secretion of substances into the bloodstream) & exocrine (secretion of substances to the outside of the body or another surface within the body) functions

More information

Endocrine Pancreas Development and Regeneration: Noncanonical Ideas From Neural Stem Cell Biology

Endocrine Pancreas Development and Regeneration: Noncanonical Ideas From Neural Stem Cell Biology 314 Diabetes Volume 65, February 2016 Jimmy Masjkur, 1 Steven W. Poser, 1 Polyxeni Nikolakopoulou, 1 George Chrousos, 2 Ronald D. McKay, 3 Stefan R. Bornstein, 1 Peter M. Jones, 4 and Andreas Androutsellis-Theotokis

More information

SUPPLEMENTARY FIGURES

SUPPLEMENTARY FIGURES SUPPLEMENTARY FIGURES 1 Supplementary Figure 1, Adult hippocampal QNPs and TAPs uniformly express REST a-b) Confocal images of adult hippocampal mouse sections showing GFAP (green), Sox2 (red), and REST

More information

Zhu et al, page 1. Supplementary Figures

Zhu et al, page 1. Supplementary Figures Zhu et al, page 1 Supplementary Figures Supplementary Figure 1: Visual behavior and avoidance behavioral response in EPM trials. (a) Measures of visual behavior that performed the light avoidance behavior

More information

Supporting Online Material for

Supporting Online Material for www.sciencemag.org/cgi/content/full/1171320/dc1 Supporting Online Material for A Frazzled/DCC-Dependent Transcriptional Switch Regulates Midline Axon Guidance Long Yang, David S. Garbe, Greg J. Bashaw*

More information

Mouse Models of K-RAS- and B-RAFinduced

Mouse Models of K-RAS- and B-RAFinduced Mouse Models of K-RAS- and B-RAFinduced Cancer Martin O. Bergö, Professor Sahlgrenska Cancer Center University of Gothenburg www.gu.se Why models? Why mice? Critical to understanding pathogenesis and identifying

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION In the format provided by the authors and unedited. 2 3 4 DOI: 10.1038/NMAT4893 EGFR and HER2 activate rigidity sensing only on rigid matrices Mayur Saxena 1,*, Shuaimin Liu 2,*, Bo Yang 3, Cynthia Hajal

More information

Glucose Homeostasis. Liver. Glucose. Muscle, Fat. Pancreatic Islet. Glucose utilization. Glucose production, storage Insulin Glucagon

Glucose Homeostasis. Liver. Glucose. Muscle, Fat. Pancreatic Islet. Glucose utilization. Glucose production, storage Insulin Glucagon Glucose Homeostasis Liver Glucose Glucose utilization Glucose production, storage Insulin Glucagon Muscle, Fat Pancreatic Islet Classification of Diabetes Type 1 diabetes Type 2 diabetes Other types of

More information