Epithelium-mesenchyme interactions play a crucial. Original Article Control of -Cell Differentiation by the Pancreatic Mesenchyme

Size: px
Start display at page:

Download "Epithelium-mesenchyme interactions play a crucial. Original Article Control of -Cell Differentiation by the Pancreatic Mesenchyme"

Transcription

1 Original Article Control of -Cell Differentiation by the Pancreatic Mesenchyme Myriam Attali, 1 Volodymyr Stetsyuk, 1 Annie Basmaciogullari, 1 Virginie Aiello, 1 Maria A. Zanta-Boussif, 2 Bertrand Duvillie, 1 and Raphael Scharfmann 1 The importance of mesenchymal-epithelial interactions for normal development of the pancreas was recognized in the early 1960s, and mesenchymal signals have been shown to control the proliferation of early pancreatic progenitor cells. The mechanisms by which the mesenchyme coordinates cell proliferation and differentiation to produce the normal number of differentiated pancreatic cells are not fully understood. Here, we demonstrate that the mesenchyme positively controls the final number of -cells that develop from early pancreatic progenitor cells. In vitro, the number of -cells that developed from rat embryonic pancreatic epithelia was larger in cultures with mesenchyme than without mesenchyme. The effect of mesenchyme was not due to an increase in -cell proliferation but was due to increased proliferation of early pancreatic duodenal homeobox-1 (PDX1) positive progenitor cells, as confirmed by bromodeoxyuridine incorporation. Consequently, the window during which early PDX1 pancreatic progenitor cells differentiated into endocrine progenitor cells expressing Ngn3 was extended. Fibroblast growth factor 10 mimicked mesenchyme effects on proliferation of early PDX1 progenitor cells and induction of Ngn3 expression. Taken together, our results indicate that expansion of early PDX1 pancreatic progenitor cells represents a way to increase the final number of -cells developing from early embryonic pancreas. Diabetes 56: , 2007 Epithelium-mesenchyme interactions play a crucial role during organogenesis. They are mediated at least in part by soluble factors produced by the mesenchyme and acting on the epithelium (1). Evidence points to a crucial role for epithelialmesenchymal interactions in cell proliferation and differentiation during pancreatic development (2). However, the mechanisms by which the mesenchyme coordinates cell proliferation and differentiation to produce a From the 1 Faculty of Medicine, University Paris-Descartes, Institut National de la Santé et de la Recherche Médicale, INSERM E363 and U845, Necker Hospital, Paris, France; and 2 Genethon-Centre National de la Recherche Scientifique Unité Mixte de Recherche 8115, Evry, France. Address correspondence and reprint requests to Raphael Scharfmann, INSERM E363 and U845 Faculté Necker, 156 Rue de Vaugirard, Paris, France. scharfmann@necker.fr. Received for publication 15 September 2006 and accepted in revised form 22 January Published ahead of print at on 23 February DOI: /db BrdU, bromodeoxyuridine; FGF10, fibroblast growth factor 10; INSERM, Institut National de la Santé et de la Recherche Médicale; PDX1, pancreatic duodenal homeobox by the American Diabetes Association. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. normal number of differentiated pancreatic cells are not fully understood. The mature pancreas contains endocrine islets composed of cells producing hormones, such as insulin ( cells) and glucagon ( -cells), and exocrine tissue composed of acinar cells producing enzymes (e.g., carboxypeptidase-a) secreted into the intestine. The pancreas originates from the dorsal and ventral regions of the foregut endoderm. Recently, important findings have shed light on the processes controlling pancreatic endocrine cell development. Studies of genetically engineered mice identified a hierarchy of transcription factors regulating pancreas organogenesis and islet-cell differentiation (3 5). The endodermal region committed to a pancreatic fate first expresses transcription factor pancreatic duodenal homeobox-1 (Pdx1). Pdx1 is detected in mouse embryos on embryonic day 8.5 (E8.5) (E9 in rats) in early pancreatic progenitors. During adulthood, Pdx1 expression becomes largely confined to -cells, where it activates insulin gene transcription (6). Disruption of the Pdx1 gene in mice or human leads to pancreatic agenesis (7,8). These data indicate that Pdx1 is necessary for morphogenesis and differentiation of the pancreatic buds. Pdx1 is also an efficient marker of early pancreatic progenitor cells. Differentiation into endocrine and exocrine cells is the next step, and cell-tracing experiments have shown that both endocrine and exocrine cells derive from Pdx1-expressing progenitor cells (9,10). The transcription factor Neurogenin3 (Ngn3) is expressed in epithelial pancreatic progenitor cells before endocrine differentiation and is subsequently downregulated during differentiation (11). Ngn3-deficient mice lack pancreatic endocrine cells (12). Lineage-tracing experiments have demonstrated that Ngn3-expressing cells are islet progenitors (10). Thus, Ngn3 is a marker of choice for detecting the onset of pancreatic endocrine cell differentiation. Pancreas differentiation is controlled by permissive signals derived from adjacent mesodermal structures. Signals from the notochord and dorsal aorta control the first steps of pancreatic development (13,14). Signals from the mesenchyme, which condenses around the underlying committed endoderm, control the subsequent steps (2). Classic culture explant experiments highlighted the importance of the mesenchyme for exocrine-pancreas growth and differentiation (15,16). However, the role of the mesenchyme in regulating -cell mass remains unclear. For example, results based on loss-of-function of genes encoding growth factors expressed in the pancreatic mesenchyme, such as fibroblast growth factor 10 (FGF10), suggest that signals from the mesenchyme directly and positively control the final number of -cells (17). On the other hand, when 1248 DIABETES, VOL. 56, MAY 2007

2 M. ATTALI AND ASSOCIATES FIG. 1. In vitro development of rat embryonic pancreatic rudiments cultured at the air/liquid interface. E13.5 rat embryonic pancreatic epithelia were dissected with or without their mesenchyme and cultured for different periods. A J: Representative pictures after 0 (A and F), 1 (B and G), 3(C and H),5(D and I), and 7 (E and J) days of culture. In F J, the epithelium is circled. Scale bar 50 m. K: Representative real-time PCR analyses of Nkx6.2, Ngn3, Pax4, NeuroD, Pcsk1, Pcsk2, and Abcc8 mrna expression after 0, 1, 3, 5, or 7 days of culture. On each graph, gene expression is presented as a percentage of the highest sample. Each point represents the mean SE of three individual pools. FGF10 was overexpressed in the pancreas, -cell development was inhibited (18,19). Thus, the exact effect of mesenchymal signals on -cell development remains controversial, because studies of loss and gain of FGF10 function suggest opposite conclusions. Here, we further dissected the role of the pancreatic mesenchyme on -cell development. This required an in vitro model permissive for -cell development both with and without mesenchyme. Previously, we had shown that in collagen gel, -cells developed properly from embryonic DIABETES, VOL. 56, MAY

3 MESENCHYME AND -CELL DIFFERENTIATION FIG. 2. -Cell development from epithelia grown with or without mesenchyme. A J: Immunohistological analysis of pancreatic epithelia grown in vitro without (A E) or with (F J) mesenchyme for 0 (A and F),1(B and G),3(C and H),5(D and I), and 7 days (E and J). -Cell development was evaluated using anti-insulin (red) staining. Nuclei were stained in blue with Hoechst. K: Quantification of the absolute surface areas occupied by insulin cells that developed with or without mesenchyme. Three rudiments were analyzed for each condition. Data are means SE. **P < Scale bar 50 m. pancreatic epithelium when cultured without mesenchyme (20), whereas this culture condition was poorly permissive for -cell development when the epithelium was cultured with mesenchyme (21). Here, we developed an in vitro model in which -cells can develop both with and without mesenchyme. Using this model, we showed that more -cells developed from epithelium cultured with mesenchyme than without mesenchyme. We next investigated whether mesenchyme increased -cell mass by activating -cell proliferation or by acting on earlier events. We found that -cell proliferation was not modified by mesenchymal signals, whereas the proliferation of early PDX1 progenitor cells was strongly increased in the presence of mesenchyme. Consequently, the window during which early PDX1 progenitors differentiate into endocrine progenitors expressing Ngn3 was extended and the amplitude of Ngn3 expression was increased. FGF10 mimicked the effects of the mesenchyme on proliferation of early PDX1 progenitor cells and on induction of Ngn3 expression. Thus, -cell development is enhanced by signals from the mesenchyme that positively control the proliferation of early embryonic pancreatic progenitor cells. RESEARCH DESIGN AND METHODS Pregnant Wistar rats were purchased from the Janvier breeding center (Centre d Elevage René Janvier, Le Genest, France). Pregnant female rats at 13.5 days of gestation were killed by CO 2 asphyxiation, according to the guidelines of the French Animal Care Committee. To study cell proliferation in vivo, pregnant rats at E13.5 and E18.5 were injected with bromodeoxyuridine (BrdU) (100 g/g body wt) (Sigma) and killed 1 h later. Dissection of pancreatic rudiments. The embryos were harvested on E13.5. The stomach, the pancreas, and a small portion of the intestine were dissected together; then, either the pancreatic primordium was dissected or the mesenchyme was separated from the pancreatic epithelium as described previously (21). Organ culture. Dorsal pancreatic rudiments with or without their mesenchyme were laid on Millicell culture plate inserts (Millipore) in 35-mm sterile Petri dishes containing 2 ml RPMI 1640 (Invitrogen) supplemented with penicillin (100 units/ml), streptomycin (100 g/ml), HEPES (10 mmol/l), L-glutamine (2 mmol/l), nonessential amino acids (1 ; Gibco), and 10% heat-inactivated calf serum (Hyclone). Under such culture conditions, the explants grew at the air/medium interface. Cultures were maintained at 37 C in humidified 95% air/5% CO 2. The medium was changed every other day. Recombinant human FGF10 (50 ng/ml; R&D Systems) was used in the presence of heparin (50 g/ml; Sigma). For cell proliferation, BrdU (10 mol/l) was added to the culture medium. Immunohistochemistry. Tissues were fixed in 10% formalin and processed for immunohistochemistry, as described previously (21,22). The following 1250 DIABETES, VOL. 56, MAY 2007

4 M. ATTALI AND ASSOCIATES FIG. 3. The mesenchyme does not activate -cell proliferation. A F: Pancreatic epithelia were grown for 3 (A and D),5(B and E), or 7 (C and F) days without (A C) or with (D F) mesenchyme and pulsed with BrdU during the last hour of culture. Insulin staining is in red and BrdU in green. G: Quantification of the number of -cells in S phase on days 3, 5, and 7, indicating that the mesenchyme did not activate -cell proliferation. H: For -cell proliferation in vivo, pregnant rats were injected with BrdU on E18.5 and killed 1 h later. Pancreases were immunostained for insulin (red) and BrdU (green). Scale bar 50 m. antibodies were used: mouse anti-human insulin (1/2,000; Sigma), rabbit anti-insulin (1/2,000; Diasorin), mouse anti-brdu (1/2; Amersham), and rabbit anti-pdx1 (1/1,000) (22). The fluorescent secondary antibodies were fluorescein isothiocyanate anti-rabbit and Texas-red anti-mouse antibodies (1/200; Jackson Immunoresearch) and Alexa-fluor anti-rabbit antibody (1/400; Biogenex). Photographs were taken using a fluorescence microscope (Leitz DMRB; Leica) and digitized using a C5810 cooled 3CCD camera (Hamamatsu, Middlesex, NJ). No signals were observed when the first antibodies were omitted. In situ hybridization. Tissues were fixed at 4 C in 4% paraformaldehyde in PBS and either included in paraffin or cryoprotected in 15% sucrose-pbs at 4 C overnight, embedded in 15% sucrose-7.5% gelatin in PBS, and frozen in isopentane. Paraffin sections (4 m thick) or cryosections (14 m thick) were prepared. A Ngn3 probe (726 bp) was used (23) and in situ hybridization was DIABETES, VOL. 56, MAY

5 MESENCHYME AND -CELL DIFFERENTIATION FIG. 4. The mesenchyme amplifies the early PDX1 cell pool. A B: Pancreatic epithelia were grown for 24 h without (A) or with (B and B ) mesenchyme and pulsed with BrdU during the last hour of culture. Immunohistochemistry was performed using anti-pdx1 (in green) and anti-brdu (in red) antibodies. Nuclei were stained in blue with Hoechst. B is an enlargement of B. C: Quantification of the number of early PDX1 cells in S phase after 24 h of culture. Data are means SE. **P < D and D : In vivo proliferation of early PDX1 progenitors. Pregnant rats were injected with BrdU on E13.5 and killed 1 h later. Immunostaining for PDX1 is in green and BrdU in red. D is an enlargement of D. Scale bar 50 m. done as previously described (24). No signal was obtained when a sense riboprobe was used. Quantification. To quantify the absolute numbers of insulin- and Ngn3- expressing cells, all sections of each pancreatic rudiment were digitized. On every image, the surfaces of insulin or Ngn3 stainings were quantified using IPLab Eval (version 3.2.4; Scanalytics), and the stained areas were summed as previously described (25). Three to four rudiments were analyzed per condition. To measure proliferation of either -cells or early progenitors expressing PDX1, we counted the frequency of BrdU nuclei among 1,000 insulin cells or 1,000 early PDX1 progenitors per rudiment. Three rudiments were analyzed per condition. To quantify the maintenance of early PDX1 progenitors, we counted the number of insulin cells among 1,000 PDX1 cells. The percentage of undifferentiated PDX1-expressing cells was then calculated. Three rudiments were analyzed per condition. Statistical significance was determined using Student s t test. RNA extraction and real-time PCR. Total RNA was extracted from pools of three to four embryonic pancreases (cultured with or without mesenchyme) using Rneasy microkit (Qiagen). The cdnas were generated using Superscript reagents (Invitrogen) according to the manufacturer s instructions. Real-time PCR was performed using a 7300 real-time PCR system (Applied Biosystems). Each reaction consist of a mix of Taqman universal PCR master mix (Applied Biosystems) with primers and Taqman-labeled probe specific for each gene (Applied Biosystems) and run after universal thermal cycling protocol (95 C for 10 min followed by 40 cycles of 95 C for 15 s and 65 C for 1 min). Control reactions in the absence of template were included in each assay. Results 1252 DIABETES, VOL. 56, MAY 2007

6 M. ATTALI AND ASSOCIATES FIG. 5. The pool of early PDX1 progenitor cells is maintained in the presence of mesenchyme. A I: Immunohistological analyses of E13.5 rat pancreatic epithelia cultured for 0 (A),1(B and F),3(C and G),5(D and H), or 7 (E and I) days without (B E) or with (F I) mesenchyme. Immunostaining for PDX1 is in green and insulin in red. J: Quantification of the proportion of PDX1 cells that remained negative for insulin. Three rudiments were analyzed for each condition. Data are means SE. *P < Scale bar 50 m. were normalized to the transcript encoding the housekeeping protein cyclophilin A. Each point represents the mean SE of three individual pools. PCR primer sequences are available on request. RESULTS Development of rat embryonic pancreatic epithelium cultured at the air/liquid interface with or without its mesenchyme. We developed a culture model that allowed -cell differentiation with and without mesenchyme. For this purpose, we dissected pancreatic epithelia at E13.5. At this stage, the pancreatic epithelium is mainly composed of early PDX1 progenitors, with few glucagon endocrine cells. We cultured pancreatic rudiments at the air/ liquid interface on filters floating on culture medium. Without mesenchyme, the epithelium acquired a spherical shape after 1 day in culture, and the size of the tissues did not change significantly during the 7-day culture period (Fig. 1A E). At the end of the 7 days, the epithelium appeared as a small dark sphere surrounded by a variable number of translucent buds (Fig. 1E). In the presence of mesenchyme, in contrast, the epithelium grew rapidly, spread into the mesenchyme, and developed lobules (Fig. 1F J). We next analyzed the in vitro pattern of expression of different pancreatic genes. We focused on Nkx6.2 expressed in vivo at early stages of development, its expression declining in late embryogenesis (26), Ngn3 and Pax4 known to be expressed in pancreatic endocrine progenitors, and NeuroD expressed both in endocrine progenitors and in mature -cells. We also followed the expression of three genes expressed in mature -cells: Pcsk1, Pcsk2, and Abcc8. As shown in Fig. 1K, Nkx6.2 was expressed during the first 3 days of culture, its expression decreasing thereafter. The expression of Ngn3 and Pax4 was turned on and next decreased both with and without mesenchyme, as expected for markers of pancreatic progenitors. The expression of NeuroD was rapidly activated and did not decrease with time, as expected for a gene expressed both in progenitors and mature -cells. Finally, the expression of Pcsk1, Pcsk2, and Abcc8 increased at late time points of in vitro development. Effect of the mesenchyme on the absolute number of -cells. We first compared -cell development from epithelia cultured with or without mesenchyme. In epithelia cultured with or without mesenchyme, the first insulinexpressing cells were detected after 3 days of culture (Fig. 2C and H), and their number increased thereafter (Fig. 2D, E, I, and J). We next compared -cell development in both conditions. As shown in Fig. 2K, both with and without DIABETES, VOL. 56, MAY

7 MESENCHYME AND -CELL DIFFERENTIATION FIG. 6. Expression of Ngn3 in embryonic pancreatic epithelia cultured with or without mesenchyme. A K: E13.5 rat pancreatic epithelia cultured for0(a),1(b and G),3(C and H),5(D and I),7(E and J), or 9 (F and K) days without (B F) or with (G K) mesenchyme. Ngn3 expression was detected by in situ hybridization. L: Quantification of the absolute surface areas occupied by Ngn3 cells that developed over 0, 1, 3, 5, 7, or 9 days of culture with or without mesenchyme. Three to four rudiments were analyzed for each condition. Data are means SE. **P < Scale bar 50 m. mesenchyme, the number of -cells increased during the first 5 culture days. However, the number of -cells did not increase further from days 5 to 7 without mesenchyme but continued to increase with mesenchyme. As a result, a significantly larger number of -cells developed with than without mesenchyme. Individual -cell size did not vary in the different culture conditions (data not shown). When epithelium was dissected, directly reassociated with mesenchyme, and cultured, its development was similar to the one of whole pancreas (data not shown). The effect of the mesenchyme on the number of -cells that develop is not due to an increase in -cell proliferation. We compared BrdU incorporation in -cells developed without or with the mesenchyme. After 3 days of culture, -cells developed without or with mesenchyme did not incorporate BrdU (Fig. 3A and D). After 5 and 7 days of culture, some -cells developed without mesenchyme incorporated BrdU (Fig. 3B and C); this proliferation was even lower with mesenchyme (Fig. 3E G for quantification). This low proliferation of -cells resembles the one found in the rat embryonic pancreas at E18.5 (Fig. 3H). Thus, the increase in the number of -cells that developed with mesenchyme cannot be explained by an activation of -cell proliferation by the mesenchyme. The mesenchyme activates the proliferation of early pancreatic progenitors. We cultured epithelia for 1 day with or without mesenchyme and added BrdU during the last hour of culture. The percentage of PDX1 cells that incorporated BrdU was 5 2% without mesenchyme (Fig. 4A) and 39 7% with mesenchyme (Fig. 4B, B, and C for quantification). When epithelium was dissected, directly reassociated with mesenchyme, and cultured for 1 day, the proliferation of early PDX1 progenitors was similar to the one of whole pancreas (data not shown). A similar high level of PDX1 -cell proliferation in the presence of mesenchyme can be found in the E13.5 rat embryonic pancreas (Fig. 4D and D ). Thus, the mesenchyme is necessary for maintaining the proliferation of early pancreatic progenitors. The pool of early PDX1 progenitors is maintained in the presence of mesenchyme. To investigate the consequences of proliferation of early PDX1 pancreatic progenitors, we quantified the maintenance of these cells with or without mesenchyme. We cultured pancreatic epithelia with or without mesenchyme and quantified the decrease in the number of early PDX1 /insulin progenitors. At E13.5, the epithelium is composed of early PDX1 /insulin cells (Fig. 5A). In cultures without mesenchyme (Fig DIABETES, VOL. 56, MAY 2007

8 M. ATTALI AND ASSOCIATES FIG. 7. The mesenchyme does not activate the proliferation of Ngn3- expressing progenitor cells. Pancreatic epithelia were grown without (A and B) or with (C and D) mesenchyme for 3 (A and C) or5(b and D) days and pulsed with BrdU during the last hour of culture. Ngn3 expression was detected by in situ hybridization (in blue), whereas BrdU cells were shown in green. Scale bar 25 m. 5B E and J for quantification), the proportion of early PDX1 /insulin progenitors decreased rapidly, and after 3 and 5 days, 41 4 and 23 5% of PDX1 cells remained insulin negative, respectively. With mesenchyme (Fig. 5F I and J for quantification), the decrease in the proportion of early PDX1 /insulin progenitors was slower, and after 3 and 5 days, 76 4 and 51 11% of PDX1 cells remained insulin negative, respectively. By knowing the absolute area occupied by -cells and the percentage of undifferentiated Pdx1 cells, we deduced and compared the absolute number of undifferentiated Pdx1 cells. We found 4.02 and 4.37 times more PDX1 insulin cells with than without mesenchyme after 3 and 5 days of culture, respectively. These results indicate that, in addition to increasing progenitor cell proliferation, the mesenchyme maintains a pool of early PDX1 progenitors. The increase in the number of early PDX1 progenitors by the mesenchyme enhances the -cell pathway. We next monitored Ngn3 expression in epithelia grown with or without mesenchyme. At E13.5, very few cells expressed Ngn3 in rat pancreatic epithelium (Fig. 6A). After 1 day of culture without mesenchyme, Ngn3 expression increased dramatically, reaching a peak followed by a plateau until day 3 then by a sharp decrease on days 5 and 7 (Fig. 6B F). With mesenchyme (Fig. 6G K), the number of Ngn3 cells increased slightly at day 1 compared with epithelia cultured alone, but interestingly, in 3- and 5-dayold cultures, the number of Ngn3 cells was dramatically higher in epithelia cultured with than without mesenchyme (Fig. 6L). In addition, Ngn3 expression was prolonged with mesenchyme: Whereas Ngn3 cells were extremely scarce after 5 days and undetectable after 7 days without mesenchyme, they were present after 5 and 7 days of culture with mesenchyme. Compare Fig. 6D with I, and compare Fig. 6E with J. Quantifications are shown in Fig. 6L. Hypotheses to explain the increase in the number of Ngn3 cells in the presence of mesenchyme include increased differentiation of amplified PDX1 progenitors and proliferation of Ngn3 cells. To test the latter hypothesis, we measured BrdU incorporation by Ngn3 cells grown in vitro from epithelia cultured for 3 or 5 days with or without mesenchyme. Extremely rare Ngn3 cells incorporating BrdU could be found in epithelia that developed either with or without mesenchyme (Fig. 7). This poor in vitro proliferation of Ngn3 cells is reminiscent of the low level of Ngn3 -cell proliferation in the pancreas at E16.5 and E18.5 (data not shown) and could not explain the major increase in the number of Ngn3 cells in the presence of mesenchyme. Thus, our data strongly suggest that the mesenchyme may amplify Ngn3 expression by increasing the magnitude and duration of development of early PDX1 progenitors into Ngn3 progenitors. FGF10 mimics the effects of the mesenchyme on proliferation and differentiation of early PDX1 progenitors. In mice, Fgf10 is expressed in the mesenchyme adjacent to the early pancreatic epithelial buds and required for normal development of the pancreas (17). In rat, FGF10 is expressed in the pancreatic mesenchyme between E12.5 and E18 (27; data not shown). We thus tested whether the effects of the mesenchyme on early progenitor cell proliferation and differentiation could be mimicked by FGF10. Epithelia were cultured for 1 day with or without FGF10. As shown in Fig. 8A C, FGF10 treatment significantly increased the proliferation of PDX1 progenitors. To test the effect of FGF10 on progenitor cell differentiation, epithelia were cultured for 1, 3, or 5 days alone, with mesenchyme, or with FGF10. Without mesenchyme and FGF10, Ngn3 expression increased after 1 day of culture, reached a plateau at day 3, and decreased thereafter. On the other hand, in the presence of either mesenchyme or FGF10, the pattern of expression of Ngn3 was amplified and prolonged (Fig. 8D M). For example, whereas after 5 days of culture, extremely rare Ngn3 cells were detected in epithelia cultured alone (Fig. 8G), Ngn3 cells were abundant in epithelia cultured with FGF10 (Fig. 8M). Such effects of FGF10 on Ngn3 expression were further confirmed by real-time PCR (data not shown). Finally, we quantified the absolute number of -cells developed in epithelia cultured for 5 days in the absence or presence of FGF10. As shown in Fig. 8N P, FGF10 treatment significantly increased the absolute number of -cells that develop. Such effect of FGF10 was also confirmed by real-time PCR (data not shown). DISCUSSION Our results show that the mesenchyme positively regulates the final number of -cells developed from embryonic pancreas. This effect was not due to a direct action of the mesenchyme on the proliferation of either -cells or Ngn3 endocrine progenitors. Instead, the mesenchyme acted by increasing the proliferation of early PDX1 pancreatic progenitors and amplifying and prolonging the formation of Ngn3 endocrine progenitors. We used an in vitro model that allows -cell development from embryonic pancreatic epithelium cultured with and without mesenchyme (28). In earlier work, we showed that -cells developed from embryonic pancreatic epithelium cultured in collagen gel without mesenchyme (20), DIABETES, VOL. 56, MAY

9 MESENCHYME AND -CELL DIFFERENTIATION FIG. 8. FGF10 mimics the effects of the mesenchyme on proliferation and differentiation of early PDX1 progenitor cells. A--C: Pancreatic epithelia were grown without (A) or with (B) FGF10 for 1 day and pulsed with BrdU during the last hour of culture. Immunostaining for PDX1 is in green and BrdU in red. C: Quantification of the number of PDX1 cells in S phase. Data are means SE. **P < Scale bar 50 m. D M: E13.5 rat pancreatic epithelia cultured for 0 (D),1(E, H, and K),3(F, I, and L), or 5 (G, J, and M) days without (E G) or with (H J) mesenchyme or without mesenchyme but with FGF10 (K M). Ngn3 expression was detected by in situ hybridization (in blue). Scale bar 50 m. N P: In vitro effects of FGF10 on -cell development. Pancreatic epithelia were grown without (N) or with (O) FGF10 for 5 days. Insulin cells were revealed in red. Nuclei are stained in blue with Hoechst. P: Quantification of the absolute surface areas occupied by insulin cells that developed during 5 days without or with FGF10. Three rudiments were analyzed for each condition. Data are means SE. **P < Scale bar 50 m. whereas -cells developed poorly when cultured with mesenchyme (21). This inhibitory effect of mesenchyme was due to delayed Ngn3 induction and required a functional Notch pathway (24). A working hypothesis was that hypoxia occurred in collagen gel, activating the Notch pathway and inhibiting endocrine differentiation, a mechanism recently established for other cell types (29). Here, we cultured rat embryonic pancreatic epithelia at the air/liquid interface. Under these conditions, -cells developed in epithelia cultured with or without mesenchyme. -Cells never developed from mesenchyme cultured alone (data not shown). We then found that -cell development in this in vitro model mimicked in vivo development. For example, Ngn3 expression was turned on rapidly then turned off a few days later, in keeping with the reported in vivo pattern (11,30). Also similar to in vivo events, in vitro, Ngn3 induction was followed by the activation of other genes, such as Pax4 and NeuroD, and finally by the induction of markers of terminally differentiated -cells, such as Pcsk1 and Pcsk2, two endoproteolytic enzymes necessary for proinsulin processing, and Abcc8, a sulfonylurea receptor (31 33). It is also interest DIABETES, VOL. 56, MAY 2007

10 M. ATTALI AND ASSOCIATES ing to note that in vitro cell proliferation faithfully replicated in vivo events (17,26,34). Specifically, with mesenchyme, early PDX1 progenitors proliferated at a fast pace. When the cells entered the endocrine pathway and expressed Ngn3, proliferation decreased sharply. Finally, the first -cells that developed after 3 days of culture did not proliferate. This simple in vitro model in which proliferation and differentiation mimic in vivo events is extremely useful for elucidating the effects of the mesenchyme at each step of -cell development. The exact role for the mesenchyme in coordinating progenitor cell proliferation and differentiation is incompletely understood (15,16). Here, we demonstrate that the mesenchyme increases the proliferation of early PDX1 pancreatic progenitors, amplifying and prolonging the formation of Ngn3 endocrine progenitors and ultimately increasing -cell development, and FGF10 mimics the effect of the mesenchyme. However, FGF10 does not provide complete growth of the explants when compared with mesenchyme. We are currently testing whether in addition to FGF10, the mesenchyme generates additional signals that are important for pancreatic epithelial development. We previously demonstrated that FGF10 is produced by embryonic pancreatic mesenchymal cells and is required for the proliferation of early pancreatic progenitors (17). We suggested that failure of the progenitor pool to expand in Fgf10 mutants might lead to a reduced number of progenitors available for differentiation, ultimately resulting in an inadequate number of cells to produce a normal pancreas. However, the detrimental effects of FGF10 deletion hindered the interpretation of the role for FGF10 in later stages of development (17). However, in transgenic mice ectopically expressing FGF10 in the pancreatic epithelium, epithelial cell proliferation increased but pancreatic differentiation of all cell types was markedly decreased (18,19). Such a phenotype was also observed when E10 mouse pancreatic epithelium was cultured in matrigel in the presence of FGF10 (35). This inhibition of differentiation was unexpected, and the contradiction between the phenotype of FGF10-deficient mice and transgenic mice with ectopic FGF10 expression or E10 mouse pancreatic epithelium grown with FGF10 was unexplained. For example, the phenotype of FGF10-deficient mice suggested that FGF10 did not inhibit endocrine cell differentiation. In the absence of FGF10, there was no excessive expression of early markers for endocrine cells, such as Isl1 and Ngn3. Moreover, glucagon-expressing cells did not form prematurely or in excess (17). In contrast, endocrine cell differentiation was strongly inhibited in transgenic mice overexpressing FGF10 or in E10 mouse pancreatic epithelium grown with FGF10 (18,19,35). One hypothesis for explaining these apparent contradictions involves the timing of FGF10 expression in transgenic mice. In these mice, FGF10 expression is controlled by the Pdx1 promoter, which is expressed very early in the pancreas, before the mesenchyme condenses around the gut tube. At this early stage, FGF10 treatment of dorsal pancreatic endoderm explants was sufficient to increase the expression of Ptf1a, a transcription factor needed to complete pancreatic specification (36,37). Thus, one possibility is that early FGF10 misexpression in the pancreas modifies the pancreatic cell-differentiation program, a mechanism recently suggested to explain the pancreatic phenotype of mice that overexpress Wnt1 under the control of the Pdx1 promoter (38). These mice have pancreatic agenesis. Comparisons of their phenotype with that of mice deficient in -catenin suggest that Wnt1 overexpression at early development stages may reflect alternative roles for Wnt signaling, such as specifying the fate of the intestine (39). Difference in the stage of development of the explants could also explain the differences between our study and the one by Miralles et al. (35). We used E13.5 rat pancreas, whereas Miralles et al. used E10 mouse pancreas. Culture conditions could also explain the differences. Although, here, we grew explants on filters at the air/liquid interface, Miralles et al. performed cultures in matrigel that contains basement membrane proteins that are not fully characterized. Taken together, our data indicate that 1) the pancreatic mesenchyme increases the final -cell population by activating the proliferation of early PDX1 pancreatic progenitors; and 2) the effects of the mesenchyme on proliferation and differentiation can be mimicked by FGF10. We propose that early PDX1 pancreatic progenitors represent an expansion pool characterized by a significant proliferative potential that should be exploited for expansion with FGF10 and differentiation into -cells. Interestingly, Baetge and colleagues (40) recently directed differentiation of human embryonic stem cells into insulinproducing cells by mimicking embryonic development. However, as noted by the authors, the protocol must be further improved to produce therapeutic -cells. The information we provide here should be useful to define the exact timing during which FGF10 should be used to amplify pancreatic progenitors and thus increase the final number of -cells that will develop. ACKNOWLEDGMENTS M.A. received support from the French Ministry for Research and Technology and from the Association Pour la Recherche Médical. This work was supported by Juvenile Diabetes Research Foundation (JDRF Center for Cell Therapy in Europe), INSERM/Fondation pour la Recherche Médicale/JDRF (Grant 4DA03H), the National Institutes of Health Beta Cell Biology Consortium (DK ), the 6th European Union Framework Program ( -Cell Therapy Integrated Project), INSERM-JDRF Grant (AIP Cellules souches A03139MS), the French National Program of Research on Diabetes, and the Association Française des Diabetiques. REFERENCES 1. Hogan BL: Morphogenesis. Cell 96: , Pictet R, Rutter W: Development of the embryonic pancreas. In Handbook of Physiology. Vol. 1. Steiner DF, Freinkel N, Eds., Baltimore, MD, The Williams & Wilkins, 1972, p Edlund E: Transcribing pancreas. Diabetes 47: , Jensen J: Gene regulatory factors in pancreatic development. Dev Dyn 229: , Wilson ME, Scheel D, German MS: Gene expression cascades in pancreatic development. Mech Dev 120:65 80, Ohlsson H, Karlsson K, Edlund T: IPF1, a homeodomain-containing transactivator of the insulin gene. EMBO J 12: , Ahlgren U, Jonsson J, Edlund H: The morphogenesis of the pancreatic mesenchyme is uncoupled from that of the pancreatic epithelium in IPF1/PDX1-deficient mice. Development 122: , Offield M, Jetton T, Laborsky P, Ray M, Stein R, Magnuson M, Hogan B, Wright C: PDX-1 is required for pancreatic outgrowth and differentiation of the rostral duodenum. Development 122: , Herrera PL: Adult insulin- and glucagon-producing cells differentiate from two independent cell lineages. Development 127: , Gu G, Dubauskaite J, Melton DA: Direct evidence for the pancreatic lineage: NGN3 cells are islet progenitors and are distinct from duct progenitors. Development 129: , 2002 DIABETES, VOL. 56, MAY

11 MESENCHYME AND -CELL DIFFERENTIATION 11. Apelqvist A, Li H, Sommer L, Beatus P, Anderson DJ, Honjo T, Hrabe de Angelis M, Lendahl U, Edlund H: Notch signalling controls pancreatic cell differentiation. Nature 400: , Gradwohl G, Dierich A, LeMeur M, Guillemot F: Neurogenin3 is required for the development of the four endocrine cell lineages of the pancreas. Proc Natl Acad Sci U S A 97: , Kim S, Hebrok M, Melton D: Notochord to endoderm signaling is required for pancreas development. Development 124: , Lammert E, Cleaver O, Melton D: Induction of pancreatic differentiation by signals from blood vessels. Science 294: , Golosow N, Grobstein C: Epitheliomesenchymal interaction in pancreatic morphogenesis. Dev Biol 4: , Wessels N, Cohen J: Early pancreas organogenesis: morphogenesis, tissue interactions, and mass effects. Dev Biol 15: , Bhushan A, Itoh N, Kato S, Thiery J, Czernichow P, Bellusci S, Scharfmann R: Fgf10 is essential for maintaining the proliferative capacity of epithelial progenitor cells during early pancreatic organogenesis. Development 128: , Hart A, Papadopoulou S, Edlund H: Fgf10 maintains notch activation, stimulates proliferation, and blocks differentiation of pancreatic epithelial cells. Dev Dyn 228: , Norgaard GA, Jensen JN, Jensen J: FGF10 signaling maintains the pancreatic progenitor cell state revealing a novel role of Notch in organ development. Dev Biol 264: , Miralles F, Serup P, Cluzeaud F, Vandewalle A, Czernichow P, Scharfmann R: Characterization of beta cells developed in vitro from rat embryonic pancreatic epithelium. Dev Dyn 214: , Miralles F, Czernichow P, Scharfmann R: Follistatin regulates the relative proportions of endocrine versus exocrine tissue during pancreatic development. Development 125: , Duvillie B, Attali M, Aiello V, Quemeneur E, Scharfmann R: Label-retaining cells in the rat pancreas: location and differentiation potential in vitro. Diabetes 52: , Ravassard P, Chatail F, Mallet J, Icard-Liepkalns C: Relax, a novel rat bhlh transcriptional regulator transiently expressed in the ventricular proliferating zone of the developing central nervous system. J Neurosci Res 48: , Duvillie B, Attali M, Bounacer A, Ravassard P, Basmaciogullari A, Scharfmann R: The mesenchyme controls the timing of pancreatic -cell differentiation. Diabetes 55: , Cras-Meneur C, Elghazi L, Czernichow P, Scharfmann R: Epidermal growth factor increases undifferentiated pancreatic embryonic cells in vitro: a balance between proliferation and differentiation. Diabetes 50: , Henseleit KD, Nelson SB, Kuhlbrodt K, Hennings JC, Ericson J, Sander M: NKX6 transcription factor activity is required for alpha- and beta-cell development in the pancreas. Development 132: , Miralles F, Czernichow P, Ozaki K, Itoh N, Scharfmann R: Signaling through fibroblast growth factor receptor 2b plays a key role in the development of the exocrine pancreas. Proc Natl Acad Sci U S A 96: , Gittes G, Galante P, Hanahan D, Rutter W, Debas H: Lineage specific morphogenesis in the developing pancreas: role of mesenchymal factors. Development 122: , Gustafsson MV, Zheng X, Pereira T, Gradin K, Jin S, Lundkvist J, Ruas JL, Poellinger L, Lendahl U, Bondesson M: Hypoxia requires notch signaling to maintain the undifferentiated cell state. Dev Cell 9: , Jensen J, Heller RS, Funder-Nielsen T, Pedersen EE, Lindsell C, Weinmaster G, Madsen OD, Serup P: Independent development of pancreatic - and -cells from neurogenin3-expressing precursors: a role for the notch pathway in repression of premature differentiation. Diabetes 49: , Sosa-Pineda B, Chowdhury K, Torres M, Oliver G, Gruss P: The Pax4 gene is essential for differentiation of insulin-producing cells in the mammalian pancreas. Nature 386: , Naya F, Huang H-P, Qiu Y, Mutoh H, DeMayo F, Leiter A, Tsai M-J: Diabetes, defective pancreatic morphogenesis, and abnormal enteroendocrine differentiation in BETA2/NeuroD-deficient mice. Genes Dev 11: , Kim SK, Hebrok M: Intercellular signals regulating pancreas development and function. Genes Dev 15: , Ahlgren U, Pfaff S, Jessel T, Edlund T, Edlund H: Independent requirement for ISL1 in formation of pancreatic mesenchyme and islet cells. Nature 385: , Miralles F, Lamotte L, Couton D, Joshi RL: Interplay between FGF10 and Notch signalling is required for the self-renewal of pancreatic progenitors. Int J Dev Biol 50:17 26, Kawaguchi Y, Cooper B, Gannon M, Ray M, MacDonald RJ, Wright CV: The role of the transcriptional regulator Ptf1a in converting intestinal to pancreatic progenitors. Nat Genet 32: , Jacquemin P, Yoshitomi H, Kashima Y, Rousseau GG, Lemaigre FP, Zaret KS: An endothelial-mesenchymal relay pathway regulates early phases of pancreas development. Dev Biol 290: , Heller RS, Dichmann DS, Jensen J, Miller C, Wong G, Madsen OD, Serup P: Expression patterns of Wnts, Frizzleds, sfrps, and misexpression in transgenic mice suggesting a role for Wnts in pancreas and foregut pattern formation. Dev Dyn 225: , Murtaugh LC, Law AC, Dor Y, Melton DA: Beta-catenin is essential for pancreatic acinar but not islet development. Development 132: , D Amour KA, Bang AG, Eliazer S, Kelly OG, Agulnick AD, Smart NG, Moorman MA, Kroon E, Carpenter MK, Baetge EE: Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nat Biotechnol 24: , DIABETES, VOL. 56, MAY 2007

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

Fgf10 is essential for maintaining the proliferative capacity of epithelial

Fgf10 is essential for maintaining the proliferative capacity of epithelial Development 128, 5109-5117 (2001) Printed in Great Britain The Company of Biologists Limited 2001 DEV6542 5109 Fgf10 is essential for maintaining the proliferative capacity of epithelial progenitor cells

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

Homeobox gene Nkx6.1 lies downstream of Nkx2.2 in the major pathway of β-cell formation in the pancreas

Homeobox gene Nkx6.1 lies downstream of Nkx2.2 in the major pathway of β-cell formation in the pancreas Development 127, 5533-5540 (2000) Printed in Great Britain The Company of Biologists Limited 2000 DEV6501 5533 Homeobox gene Nkx6.1 lies downstream of Nkx2.2 in the major pathway of β-cell formation in

More information

Defining the cell lineages of the islets of langerhans using transgenic mice

Defining the cell lineages of the islets of langerhans using transgenic mice Int. J. Dev. Biol. 46: 97-103 (2002) Defining the cell lineages of the islets of langerhans using transgenic mice PEDRO L. HERRERA* Department of Morphology, University of Geneva Medical School. 1 Rue

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

Pancreatic endocrine and exocrine cells are thought

Pancreatic endocrine and exocrine cells are thought Changes in the Expression of Transcription Factors in Pancreatic AR42J Cells During Differentiation Into Insulin-Producing Cells You-Qing Zhang, Hirosato Mashima, and Itaru Kojima Pancreatic AR42J cells

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

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

Pancreas and beta-cell development: from the actual to the possible

Pancreas and beta-cell development: from the actual to the possible REVIEW 427 Development 134, 427-438 (2007) doi:10.1242/dev.02770 Pancreas and beta-cell development: from the actual to the possible L. Charles Murtaugh The development of insulin-producing pancreatic

More information

ROLE OF EPIDERMAL AND FIBROBLAST GROWTH FACTORS IN PANCREATIC DEVELOPMENT

ROLE OF EPIDERMAL AND FIBROBLAST GROWTH FACTORS IN PANCREATIC DEVELOPMENT Helsinki University Biomedical Dissertations No. 33 ROLE OF EPIDERMAL AND FIBROBLAST GROWTH FACTORS IN PANCREATIC DEVELOPMENT Mari-Anne Pulkkinen Program for Developmental and Reproductive Biology Biomedicum

More information

NKX6 transcription factor activity is required for α- and β-cell development in the pancreas

NKX6 transcription factor activity is required for α- and β-cell development in the pancreas Research article and disease 3139 NKX6 transcription factor activity is required for α- and β-cell development in the pancreas Korinna D. Henseleit 1,2, Shelley B. Nelson 1, *, Kirsten Kuhlbrodt 2, *,

More information

Functional Analysis of Ipf1/Pdx1, MFng and Id during Pancreatic Growth and Differentiation. Per Svensson

Functional Analysis of Ipf1/Pdx1, MFng and Id during Pancreatic Growth and Differentiation. Per Svensson Functional Analysis of Ipf1/Pdx1, MFng and Id during Pancreatic Growth and Differentiation Per Svensson Umeå Centre for Molecular Medicine Umeå University, Umeå Sweden 2008 Front cover: In situ hybridisation

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

Shh-dependent differentiation of intestinal tissue from embryonic pancreas by activin A

Shh-dependent differentiation of intestinal tissue from embryonic pancreas by activin A Research Article 2077 Shh-dependent differentiation of intestinal tissue from embryonic pancreas by activin A Jonathan M. van Eyll, Christophe E. Pierreux, Frédéric P. Lemaigre and Guy G. Rousseau* Hormone

More information

Multifaceted pancreatic mesenchymal control of epithelial lineage selection

Multifaceted pancreatic mesenchymal control of epithelial lineage selection Developmental Biology 269 (2004) 252 263 www.elsevier.com/locate/ydbio Multifaceted pancreatic mesenchymal control of epithelial lineage selection Zhixing Li, 1 Pradip Manna, 1 Hiroyuki Kobayashi, 1 Troy

More information

ARTICLE. A. Soggia & K. Flosseau & P. Ravassard & G. Szinnai & R. Scharfmann & G. Guillemain

ARTICLE. A. Soggia & K. Flosseau & P. Ravassard & G. Szinnai & R. Scharfmann & G. Guillemain Diabetologia (2012) 55:2713 2722 DOI 10.1007/s00125-012-2623-0 ARTICLE Activation of the transcription factor carbohydrate-responsive element-binding protein by glucose leads to increased pancreatic beta

More information

Dynamics of embryonic pancreas development using real-time imaging

Dynamics of embryonic pancreas development using real-time imaging Developmental Biology 306 (2007) 82 93 www.elsevier.com/locate/ydbio Dynamics of embryonic pancreas development using real-time imaging Sapna Puri, Matthias Hebrok Diabetes Center, Department of Medicine,

More information

Hedgehog signaling regulates expansion of pancreatic epithelial cells

Hedgehog signaling regulates expansion of pancreatic epithelial cells Developmental Biology 280 (2005) 111 121 www.elsevier.com/locate/ydbio Hedgehog signaling regulates expansion of pancreatic epithelial cells Hiroshi Kawahira, David W. Scheel, Stuart B. Smith, Michael

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

Genes controlling pancreas ontogeny. CLAIRE BONAL and PEDRO L. HERRERA*

Genes controlling pancreas ontogeny. CLAIRE BONAL and PEDRO L. HERRERA* Int. J. Dev. Biol. 52: 823-835 (2008) doi: 10.1387/ijdb.072444cb THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY www.intjdevbiol.com Genes controlling pancreas ontogeny CLAIRE BONAL and PEDRO L. HERRERA*

More information

Expression of neurogenin3 reveals an islet cell precursor population in the pancreas

Expression of neurogenin3 reveals an islet cell precursor population in the pancreas Development 127, 3533-3542 (2000) Printed in Great Britain The Company of Biologists Limited 2000 DEV6458 3533 Expression of neurogenin3 reveals an islet cell precursor population in the pancreas Valérie

More information

Combined activities of hedgehog signaling inhibitors regulate pancreas development

Combined activities of hedgehog signaling inhibitors regulate pancreas development Research article 4871 Combined activities of hedgehog signaling inhibitors regulate pancreas development Hiroshi Kawahira 1, Nancy H. Ma 1, Emmanouhl S. Tzanakakis 1, Andrew P. McMahon 2, Pao-Tien Chuang

More information

-Catenin is essential for pancreatic acinar but not islet development

-Catenin is essential for pancreatic acinar but not islet development Research article 4663 -Catenin is essential for pancreatic acinar but not islet development L. Charles Murtaugh*, Anica C. Law, Yuval Dor and Douglas A. Melton Department of Molecular and Cellular Biology

More information

Developmental Biology

Developmental Biology Developmental Biology 318 (2008) 224 235 Contents lists available at ScienceDirect Developmental Biology journal homepage: www.elsevier.com/developmentalbiology Analysis of mpygo2 mutant mice suggests

More information

Sonic hedgehog Is Required Early in Pancreatic Islet Development

Sonic hedgehog Is Required Early in Pancreatic Islet Development Developmental Biology 244, 75 84 (2002) doi:10.1006/dbio.2002.0573, available online at http://www.idealibrary.com on Sonic hedgehog Is Required Early in Pancreatic Islet Development Philip J. diiorio,*,1,2

More information

Hepatogenesis I Liver development

Hepatogenesis I Liver development Hepatogenesis I Liver development HB 308 George Yeoh Room 2.59 MCS Building yeoh@cyllene.uwa.edu.au Topics Early liver development Tissue interaction - role of morphogens and cytokines Liver enriched transcription

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

A bipotential precursor population for pancreas and liver within the embryonic endoderm

A bipotential precursor population for pancreas and liver within the embryonic endoderm Development 128, 871-881 (2001) Printed in Great Britain The Company of Biologists Limited 2001 DEV3286 871 A bipotential precursor population for pancreas and liver within the embryonic endoderm Gail

More information

Studying pancreas development and diabetes using human pluripotent stem cells

Studying pancreas development and diabetes using human pluripotent stem cells Editorial Studying pancreas development and diabetes using human pluripotent stem cells David W. Scoville, Anton M. Jetten Cell Biology Group, Immunity, Inflammation, and Disease Laboratory, National Institute

More information

Stem Cell Reports Repor t

Stem Cell Reports Repor t Stem Cell Reports Repor t Endothelial Cells Control Pancreatic Cell Fate at Defined Stages through EGFL7 Signaling Der-I Kao, 1 Lauretta A. Lacko, 2 Bi-Sen Ding, 3 Chen Huang, 4 Kathleen Phung, 1 Guoqiang

More information

Mice lacking the homeodomain transcription factor Nkx2.2 have diabetes due to arrested differentiation of pancreatic β cells

Mice lacking the homeodomain transcription factor Nkx2.2 have diabetes due to arrested differentiation of pancreatic β cells Development 125, 2213-2221 (1998) Printed in Great Britain The Company of Biologists Limited 1998 DEV6347 2213 Mice lacking the homeodomain transcription factor Nkx2.2 have diabetes due to arrested differentiation

More information

Reciprocal endoderm-mesoderm interactions mediated by fgf24 and fgf10 govern pancreas development

Reciprocal endoderm-mesoderm interactions mediated by fgf24 and fgf10 govern pancreas development RESEARCH ARTICLE 4011 Development 134, 4011-4021 (2007) doi:10.1242/dev.007823 Reciprocal endoderm-mesoderm interactions mediated by fgf24 and fgf10 govern pancreas development Isabelle Manfroid 1, *,

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

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

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

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

Edited by Kathryn V. Anderson, Sloan Kettering Institute, New York, NY, and approved December 20, 2004 (received for review August 6, 2004)

Edited by Kathryn V. Anderson, Sloan Kettering Institute, New York, NY, and approved December 20, 2004 (received for review August 6, 2004) Lack of TCF2 vhnf1 in mice leads to pancreas agenesis C. Haumaitre*, E. Barbacci*, M. Jenny, M. O. Ott*, G. Gradwohl, and S. Cereghini* *Biologie du Développement, Unité Mixte de Recherche 7622, Centre

More information

Development of the Pancreas and Neonatal Diabetes

Development of the Pancreas and Neonatal Diabetes Development of the Pancreas and Neonatal Diabetes Endocrine Development Vol. 12 Series Editor P. Mullis Bern 1st ESPE Advanced Seminar in Developmental Endocrinology, Paris, May 10 11, 2007 Development

More information

The pancreas originates from the dorsal and ventral

The pancreas originates from the dorsal and ventral Role for VPAC2 Receptor Mediated Signals in Pancreas Development Latif Rachdi, 1 Jean-Claude Marie, 2 and Raphael Scharfmann 1 Mature pancreatic cells develop from progenitors that proliferate and differentiate

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

CHAPTER 6 SUMMARIZING DISCUSSION

CHAPTER 6 SUMMARIZING DISCUSSION CHAPTER 6 SUMMARIZING DISCUSSION More than 20 years ago the founding member of the Wnt gene family, Wnt-1/Int1, was discovered as a proto-oncogene activated in mammary gland tumors by the mouse mammary

More information

Developmental Biology

Developmental Biology Developmental Biology 352 (2011) 267 277 Contents lists available at ScienceDirect Developmental Biology journal homepage: www.elsevier.com/developmentalbiology Growth-limiting role of endothelial cells

More information

Glucagon Is Required for Early Insulin-Positive Differentiation in the Developing Mouse Pancreas

Glucagon Is Required for Early Insulin-Positive Differentiation in the Developing Mouse Pancreas Glucagon Is Required for Early Insulin-Positive Differentiation in the Developing Mouse Pancreas Krishna Prasadan, Erica Daume, Barry Preuett, Troy Spilde, Amina Bhatia, Hiroyuki Kobayashi, Mark Hembree,

More information

Pancreas development is promoted by cyclopamine, a Hedgehog signaling inhibitor

Pancreas development is promoted by cyclopamine, a Hedgehog signaling inhibitor Proc. Natl. Acad. Sci. USA Vol. 95, pp. 13036 13041, October 1998 Developmental Biology Pancreas development is promoted by cyclopamine, a Hedgehog signaling inhibitor SEUNG K. KIM* AND DOUGLAS A. MELTON

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

ARTICLE. Patricia Vázquez & Ana M. Robles & Flora de Pablo & Catalina Hernández-Sánchez

ARTICLE. Patricia Vázquez & Ana M. Robles & Flora de Pablo & Catalina Hernández-Sánchez DOI 10.1007/s00125-014-3341-6 ARTICLE Non-neural tyrosine hydroxylase, via modulation of endocrine pancreatic precursors, is required for normal development of beta cells in the mouse pancreas Patricia

More information

HOX6 FUNCTION IS NECESSARY FOR ENDOCRINE PANCREAS DEVELOPMENT IN VIVO AND IN VITRO. Brian Matthew Larsen

HOX6 FUNCTION IS NECESSARY FOR ENDOCRINE PANCREAS DEVELOPMENT IN VIVO AND IN VITRO. Brian Matthew Larsen HOX6 FUNCTION IS NECESSARY FOR ENDOCRINE PANCREAS DEVELOPMENT IN VIVO AND IN VITRO by Brian Matthew Larsen A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of

More information

MOLECULAR AND CELLULAR BIOLOGY, June 2000, p Vol. 20, No. 12. Copyright 2000, American Society for Microbiology. All Rights Reserved.

MOLECULAR AND CELLULAR BIOLOGY, June 2000, p Vol. 20, No. 12. Copyright 2000, American Society for Microbiology. All Rights Reserved. MOLECULAR AND CELLULAR BIOLOGY, June 2000, p. 4445 4454 Vol. 20, No. 12 0270-7306/00/$04.00 0 Copyright 2000, American Society for Microbiology. All Rights Reserved. Transcription Factor Hepatocyte Nuclear

More information

The role of decellularized matrix in directing differentiation of pancreatic progenitor cells in pancreatic endocrine cell fate

The role of decellularized matrix in directing differentiation of pancreatic progenitor cells in pancreatic endocrine cell fate The role of decellularized matrix in directing differentiation of pancreatic progenitor cells in pancreatic endocrine cell fate SUBMITTED TO THE FACULTY OF THE GRADUATE SCHOOL OF THE UNIVERSITY OF MINNESOTA

More information

Development of the human pancreas from foregut to endocrine commitment

Development of the human pancreas from foregut to endocrine commitment Page 1 of 35 Development of the human pancreas from foregut to endocrine commitment [Short running title: Early human pancreas development] Rachel E. Jennings 1,2, Andrew A. Berry 1, Rebecca Kirkwood-Wilson

More information

The window period of NEUROGENIN3 during human gestation

The window period of NEUROGENIN3 during human gestation The window period of NEUROGENIN3 during human gestation Rachel J Salisbury, Jennifer Blaylock, Andrew A Berry, Rachel E Jennings, Ronald De Krijger, Karen Piper Hanley, and Neil A Hanley QUERY SHEET This

More information

Conditional inactivation of Pax6 in the pancreas causes early onset of diabetes

Conditional inactivation of Pax6 in the pancreas causes early onset of diabetes Developmental Biology 269 (2004) 479 488 www.elsevier.com/locate/ydbio Conditional inactivation of Pax6 in the pancreas causes early onset of diabetes Ruth Ashery-Padan, a, * Xunlei Zhou, b Till Marquardt,

More information

Ectopic pancreas formation in Hes1-knockout mice reveals plasticity of endodermal progenitors of the gut, bile duct, and pancreas

Ectopic pancreas formation in Hes1-knockout mice reveals plasticity of endodermal progenitors of the gut, bile duct, and pancreas Research article Ectopic pancreas formation in Hes1-knockout mice reveals plasticity of endodermal progenitors of the gut, bile duct, and pancreas Akihisa Fukuda, 1,2,3 Yoshiya Kawaguchi, 1,4 Kenichiro

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

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

Solution Key EXAM 3 (4 / 29 / 13)

Solution Key EXAM 3 (4 / 29 / 13) Solution Key- 7.013 EXAM 3 (4 / 29 / 13) Question 1 (20 points) Diabetes occurs either due to a reduced insulin hormone production by the pancreatic β cells or the resistance of target cells to insulin.

More information

Development of the Liver and Pancreas

Development of the Liver and Pancreas Development of the Liver and Pancreas Professor Alfred Cuschieri Department of Anatomy University of Malta Three glandular buds arise from the distal end of the foregut during the fourth week Day 22 -The

More information

The Epithelial-Mesenchymal Interaction Plays a Role in the Maintenance of the Stem Cell Niche of Mouse Incisors via Fgf10 and Fgf9 Signaling

The Epithelial-Mesenchymal Interaction Plays a Role in the Maintenance of the Stem Cell Niche of Mouse Incisors via Fgf10 and Fgf9 Signaling The Open Biotechnology Journal, 2008, 2, 111-115 111 The Epithelial-Mesenchymal Interaction Plays a Role in the Maintenance of the Stem Cell Niche of Mouse Incisors via Fgf10 and Fgf9 Signaling Tamaki

More information

Signals from the neural crest regulate beta-cell mass in the pancreas

Signals from the neural crest regulate beta-cell mass in the pancreas RESEARCH ARTICLE 2151 Development 135, 2151-2160 (2008) doi:10.1242/dev.015859 Signals from the neural crest regulate beta-cell mass in the pancreas Nada Nekrep 1, Juehu Wang 1, Takeshi Miyatsuka 1 and

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

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION DOI: 10.1038/ncb3200 Supplementary Figure 1 Expression analysis of stomach markers in gutlike structure. (a) Differentiation scheme of gut-like structure formation from embryonic stem cells. (b) RT-PCR

More information

Supplemental Experimental Procedures

Supplemental Experimental Procedures Cell Stem Cell, Volume 2 Supplemental Data A Temporal Switch from Notch to Wnt Signaling in Muscle Stem Cells Is Necessary for Normal Adult Myogenesis Andrew S. Brack, Irina M. Conboy, Michael J. Conboy,

More information

EMBO REPORT SUPPLEMENTARY SECTION. Quantitation of mitotic cells after perturbation of Notch signalling.

EMBO REPORT SUPPLEMENTARY SECTION. Quantitation of mitotic cells after perturbation of Notch signalling. EMBO REPORT SUPPLEMENTARY SECTION Quantitation of mitotic cells after perturbation of Notch signalling. Notch activation suppresses the cell cycle indistinguishably both within and outside the neural plate

More information

Translational Embryology: Using Embryonic Principles to Generate Pancreatic Endocrine Cells From Embryonic Stem Cells

Translational Embryology: Using Embryonic Principles to Generate Pancreatic Endocrine Cells From Embryonic Stem Cells DEVELOPMENTAL DYNAMICS 236:3218 3227, 2007 SPECIAL ISSUE REVIEWS A PEER REVIEWED FORUM Translational Embryology: Using Embryonic Principles to Generate Pancreatic Endocrine Cells From Embryonic Stem Cells

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

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

Pancreas Organogenesis: From Bud to Plexus to Gland

Pancreas Organogenesis: From Bud to Plexus to Gland a DEVELOPMENTAL DYNAMICS 240:530 565, 2011 SPECIAL ISSUE REVIEWS A PEER REVIEWED FORUM Pancreas Organogenesis: From Bud to Plexus to Gland Fong Cheng Pan and Chris Wright* Pancreas oganogenesis comprises

More information

MODULATION OF β-cell INTRINSIC AND EXTRINSIC CHARACTERISTICS BY CTGF TO PROMOTE β-cell MASS REGENERATION. Kimberly G. Riley.

MODULATION OF β-cell INTRINSIC AND EXTRINSIC CHARACTERISTICS BY CTGF TO PROMOTE β-cell MASS REGENERATION. Kimberly G. Riley. MODULATION OF β-cell INTRINSIC AND EXTRINSIC CHARACTERISTICS BY CTGF TO PROMOTE β-cell MASS REGENERATION By Kimberly G. Riley Dissertation Submitted to the Faculty of the Graduate School of Vanderbilt

More information

Vascular function and sphingosine-1-phosphate regulate development of the dorsal pancreatic mesenchyme

Vascular function and sphingosine-1-phosphate regulate development of the dorsal pancreatic mesenchyme Research article 1085 Vascular function and sphingosine-1-phosphate regulate development of the dorsal pancreatic mesenchyme Josefina Edsbagge 1, *, Jenny K. Johansson 1, *,, Farzad Esni 1,, Yang Luo 2,

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

Transgenic Expression of FGF8 and FGF10 Induces Transdifferentiation of Pancreatic Islet Cells into Hepatocytes and Exocrine Cells

Transgenic Expression of FGF8 and FGF10 Induces Transdifferentiation of Pancreatic Islet Cells into Hepatocytes and Exocrine Cells Biochemical and Biophysical Research Communications 292, 138 143 (2002) doi:10.1006/bbrc.2002.6601, available online at http://www.idealibrary.com on Transgenic Expression of FGF8 and FGF10 Induces Transdifferentiation

More information

Branching morphogenesis of the lung: new molecular insights into an old problem

Branching morphogenesis of the lung: new molecular insights into an old problem 86 Review TRENDS in Cell Biology Vol.13 No.2 February 2003 Branching morphogenesis of the lung: new molecular insights into an old problem Pao-Tien Chuang 1 and Andrew P. McMahon 2 1 Cardiovascular Research

More information

LUP. Lund University Publications. Institutional Repository of Lund University

LUP. Lund University Publications. Institutional Repository of Lund University LUP Lund University Publications Institutional Repository of Lund University This is an author produced version of a paper published in Developmental Biology. This paper has been peer-reviewed but does

More information

Supplementary Fig. 1 Blocking shh function at the protein level confirms its role as a guidance cue for postcommissural axons.

Supplementary Fig. 1 Blocking shh function at the protein level confirms its role as a guidance cue for postcommissural axons. Supplementary Fig. 1 Blocking shh function at the protein level confirms its role as a guidance cue for postcommissural axons. As an alternative method to demonstrate the role of shh as a guidance cue

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

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

Endocrine pancreas development is altered in foetuses from rats previously showing intra-uterine growth retardation in response to malnutrition

Endocrine pancreas development is altered in foetuses from rats previously showing intra-uterine growth retardation in response to malnutrition Diabetologia 2002) 45: 394±401 Ó Springer-Verlag 2002 Endocrine pancreas development is altered in foetuses from rats previously showing intra-uterine growth retardation in response to malnutrition B.

More information

UNIVERSITÀ DEGLI STUDI DI NAPOLI FEDERICO II Dipartimento di Medicina Veterinaria e Produzioni Animali

UNIVERSITÀ DEGLI STUDI DI NAPOLI FEDERICO II Dipartimento di Medicina Veterinaria e Produzioni Animali UNIVERSITÀ DEGLI STUDI DI NAPOLI FEDERICO II Dipartimento di Medicina Veterinaria e Produzioni Animali Dottorato di ricerca in Organismi modello nella ricerca biomedica e veterinaria XXV Ciclo IDENTIFICATION

More information

Supplementary Figure 1. EC-specific Deletion of Snail1 Does Not Affect EC Apoptosis. (a,b) Cryo-sections of WT (a) and Snail1 LOF (b) embryos at

Supplementary Figure 1. EC-specific Deletion of Snail1 Does Not Affect EC Apoptosis. (a,b) Cryo-sections of WT (a) and Snail1 LOF (b) embryos at Supplementary Figure 1. EC-specific Deletion of Snail1 Does Not Affect EC Apoptosis. (a,b) Cryo-sections of WT (a) and Snail1 LOF (b) embryos at E10.5 were double-stained for TUNEL (red) and PECAM-1 (green).

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

Neuroepithelial Cells and Neural Differentiation

Neuroepithelial Cells and Neural Differentiation Neuroepithelial Cells and Neural Differentiation Neurulation The cells of the neural tube are NEUROEPITHELIAL CELLS Neural crest cells migrate out of neural tube Neuroepithelial cells are embryonic stem

More information

Neurodevelopment II Structure Formation. Reading: BCP Chapter 23

Neurodevelopment II Structure Formation. Reading: BCP Chapter 23 Neurodevelopment II Structure Formation Reading: BCP Chapter 23 Phases of Development Ovum + Sperm = Zygote Cell division (multiplication) Neurogenesis Induction of the neural plate Neural proliferation

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

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

Genotype analysis by Southern blots of nine independent recombinated ES cell clones by

Genotype analysis by Southern blots of nine independent recombinated ES cell clones by Supplemental Figure 1 Selected ES cell clones show a correctly recombined conditional Ngn3 allele Genotype analysis by Southern blots of nine independent recombinated ES cell clones by hybridization with

More information

A Cxcl12-Cxcr4 Chemokine Signaling Pathway Defines

A Cxcl12-Cxcr4 Chemokine Signaling Pathway Defines Supplemental Data A Cxcl12-Cxcr4 Chemokine Signaling Pathway Defines the Initial Trajectory of Mammalian Motor Axons Ivo Lieberam, Dritan Agalliu, Takashi Nagasawa, Johan Ericson, and Thomas M. Jessell

More information

Dr. Heba Kalbouneh. Dr. Heba Kalbouneh. Dr. Heba Kalbouneh

Dr. Heba Kalbouneh. Dr. Heba Kalbouneh. Dr. Heba Kalbouneh Dr. Heba Kalbouneh Dr. Heba Kalbouneh Dr. Heba Kalbouneh Basement membrane: What is the basement membrane? - It is a layer of ECM separating the epithelial cells from the underlying connective tissue Basement

More information

In mammals, the pancreas regulates the response to. Control of Cell Identity in Pancreas Development and Regeneration

In mammals, the pancreas regulates the response to. Control of Cell Identity in Pancreas Development and Regeneration GASTROENTEROLOGY 2013;144:1170 1179 Control of Cell Identity in Pancreas Development and Regeneration Ben Z. Stanger 1 Matthias Hebrok 2 1 Division of Gastroenterology, Department of Medicine, Department

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

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

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

Fig. S1. RT-PCR analyses of the expression and distribution of Xdscr6 transcripts during early development.

Fig. S1. RT-PCR analyses of the expression and distribution of Xdscr6 transcripts during early development. Fig. S1. RT-PCR analyses of the expression and distribution of Xdscr6 transcripts during early development. (A) Temporal expression of Xdscr6 at various stages (numbers on the top) and its distribution

More information

Neural stem cells and the neurobiology of ageing. Chen Siyun 1, Dawe G.S. 2

Neural stem cells and the neurobiology of ageing. Chen Siyun 1, Dawe G.S. 2 ABSTRACT Neural stem cells and the neurobiology of ageing Chen Siyun 1, Dawe G.S. 2 Department of Physics, Faculty of Science, National University of Singapore 10 Kent Ridge Road, Singapore 117546 The

More information

In Vitro Differentiation and Expansion of Human Pluripotent Stem Cell-Derived Pancreatic Progenitors

In Vitro Differentiation and Expansion of Human Pluripotent Stem Cell-Derived Pancreatic Progenitors Reprint from The Review of DIABETIC STUDIES Vol 11 No 1 2014 Special Edition Stem Cells and Pancreas Regeneration The Review of DIABETIC STUDIES REVIEW In Vitro Differentiation and Expansion of Human Pluripotent

More information

TITLE: A PSCA Promoter Based Avian Retroviral Transgene Model of Normal and Malignant Prostate

TITLE: A PSCA Promoter Based Avian Retroviral Transgene Model of Normal and Malignant Prostate AD Award Number: DAMD17-03-1-0163 TITLE: A PSCA Promoter Based Avian Retroviral Transgene Model of Normal and Malignant Prostate PRINCIPAL INVESTIGATOR: Robert Reiter, M.D. CONTRACTING ORGANIZATION: The

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

Regulation of Neurod1 Contributes to the Lineage Potential of Neurogenin3+ Endocrine Precursor Cells in the Pancreas

Regulation of Neurod1 Contributes to the Lineage Potential of Neurogenin3+ Endocrine Precursor Cells in the Pancreas Regulation of Neurod1 Contributes to the Lineage Potential of Neurogenin3+ Endocrine Precursor Cells in the Pancreas Teresa L. Mastracci 1 a, Keith R. Anderson 2. b, James B. Papizan 1., Lori Sussel 1,2

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

The toll-like receptor 4 ligands Mrp8 and Mrp14 play a critical role in the development of autoreactive CD8 + T cells

The toll-like receptor 4 ligands Mrp8 and Mrp14 play a critical role in the development of autoreactive CD8 + T cells 1 SUPPLEMENTARY INFORMATION The toll-like receptor 4 ligands Mrp8 and Mrp14 play a critical role in the development of autoreactive CD8 + T cells Karin Loser 1,2,6, Thomas Vogl 2,3, Maik Voskort 1, Aloys

More information