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

Size: px
Start display at page:

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

Transcription

1 DEVELOPMENTAL DYNAMICS 236: , 2007 SPECIAL ISSUE REVIEWS A PEER REVIEWED FORUM Translational Embryology: Using Embryonic Principles to Generate Pancreatic Endocrine Cells From Embryonic Stem Cells Jason R. Spence and James M. Wells* Diseases that affect endodermally derived organs such as the lungs, liver, and pancreas include cystic fibrosis, chronic hepatitis, and diabetes, respectively. Despite the prevalence of these diseases, cures remain elusive. While several promising transplantation-based therapies exist for some diseases such as Type 1 diabetes, they are currently limited by the availability of donor-derived tissues. Embryonic stem cells are a promising and renewable source of tissue for transplantation; however, directing their differentiation into specific, adult cell lineages remains a significant challenge. In this review, we will focus on one endodermally derived organ, the pancreas, and discuss how studies of embryonic pancreas development have been used as the basis for the directed, step-wise differentiation of mouse and human embryonic stem cells into pancreatic endocrine cells that are capable of rescuing Type 1 diabetes in animal models. Developmental Dynamics 236: , Wiley-Liss, Inc. Key words: endoderm organ ogenesis; diabetes; pancreas; -cell; insulin Accepted 27 September 2007 INTRODUCTION The endoderm germ layer gives rise to the epithelial lining of the digestive and respiratory tracts, as well as organs such as the thyroid, lungs, liver, gall bladder, and pancreas. Diseases involving endodermally derived organs, particularly the lungs, liver, and pancreas include cystic fibrosis, chronic hepatitis, and diabetes, respectively, affect tens of millions of people in North America. An emerging approach to treating patients with a disease is to stimulate regeneration of damaged tissue in vivo or to generate replacement tissue ex vivo. This broad area of research is often referred to as regenerative medicine. Stem cells have effectively been used to treat diseases of mesodermally derived organs (the hematopoietic system), and ectodermally derived tissue (skin) has been grown and expanded in culture for the treatment of burn victims. Given these successes, it is likely that regenerative medicine will prove similarly effective to treat diseases of the endoderm. Many existing transplantationbased therapies are currently limited by the availability of donor-derived tissues. One key goal of regenerative medicine is to identify a renewable source of cells and tissues for replacement therapies. Embryonic stem cells (ESCs) represent a promising source of material for transplantation because of their unique properties. ESCs are primary cells that can be expanded indefinitely in culture and, thus, are a renewable source of tissue. In addition, ESCs are capable of becoming all cell types of the body and, thus, could be used to generate cells for replacement therapy of any diseased or damaged cell. One significant challenge in using ESCs therapeutically is to identify methods that promote their differentiation into specific and functional adult lineages. Over the past 2 decades, developmental biologists have made significant progress in identifying the molecular basis of pancreas development. Studies in model organisms, including Xenopus, zebrafish, chick, and mouse, have identified signaling pathways Division of Developmental Biology, Cincinnati Children s Hospital Research Foundation, Cincinnati Ohio Grant sponsor: Juvenile Diabetes Research Foundation; Grant sponsor: NIH; Grant number: GM072915; Grant number: T32 HD *Correspondence to: James M. Wells, Division of Developmental Biology, Cincinnati Children s Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH james.wells@cchmc.org DOI /dvdy Published online 31 October 2007 in Wiley InterScience ( Wiley-Liss, Inc.

2 TRANSLATIONAL EMBRYOLOGY 3219 Fig. 1. The lineage of the developing pancreas in vivo and in embryonic stem (ES) cultures. The left panels are mouse embryos at different stages of development (embryonic day 3.5 through 13.5 [E3.5 E13.5]), and the black bars on the right indicate the equivalent stages in human development. Embryos are oriented with anterior to the left and posterior to the right. The E13.5 stage shows the dissected stomach, pancreas, and duodenum with the stomach (s) and dorsal pancreas (dp) to the left and the ventral pancreas (vp) and duodenum (d) to the right. The LacZ staining in the E9.5 and 13.5 embryos shows expression of Pdx1 (Pdx1 LacZ/ animals were from Chris Wright at Vanderbilt University and were described in Offield et al., 1996). The lower left panel is a pancreatic islet showing glucagon-expressing -cells (green) and insulin-expressing -cells (red). The curved arrows highlight several signaling pathways involved pancreas development that have been used to direct ESCs into the pancreatic lineage. The middle panel indicates the lineage of the developing endocrine pancreas. The arrowheads in the cell lineage diagram indicate two separate roles of Nodal/Activin signaling: to initiate gastrulation and to promote endoderm vs. mesoderm fate in a dose-dependent manner. and downstream transcriptional networks that direct cell fate decisions throughout the formation of the pancreas. In this review, we will briefly highlight several key stages of pancreas development and several molecular pathways involved in these cell fate decisions (Fig. 1; for reviews that focus on endoderm and pancreas development, see Slack, 1995; Wells and Melton, 1999; Lewis and Tam, 2006; Murtaugh, 2007; Zorn and Wells, 2007). We will then discuss how these embryonic principles have been used as the basis for the directed, step-wise differentiation of mouse and human embryonic stem cells into specific cell types in culture, in particular, pancreatic endocrine cells (Fig. 1). There has been great progress made over the past few years in directing ESCs into this lineage, and we will discuss some hurdles that remain in the way of creating fully functional pancreatic -cells that could be used therapeutically. EMBRYONIC DEVELOPMENT OF THE PANCREAS Endoderm Formation During vertebrate development, the morphogenetic process of gastrulation results in a group of undifferentiated cells forming the three principal germ layers: ectoderm, mesoderm, and endoderm. There is increasing evidence from studies in fish, frogs, and mice, that the endoderm and mesoderm share a common progenitor referred to as mesendoderm. Cell lineage analyses of early to mid-stage mouse gastrula indicate that mesendoderm progenitor cells are concentrated at the anterior primitive streak near the node and that they adopt either a mesoderm or endoderm fate as they migrate through the primitive streak and incorporate into their respective germ layers (Lawson and Pedersen, 1987; Lawson et al., 1991). However, the exact nature of when and how endoderm cells acquire their identity during gastrulation is not well understood. The node is the source of Nodal, a member of the transforming growth factor-beta (TGF ) family of growth factors. The nodal-related growth factor signaling pathway is required for establishing the anterior posterior axis by restricting gastrulation to the posterior of the embryo (Conlon et al., 1994; Brennan et al., 2001; Perea-Gomez et al., 2002). Nodal subsequently plays a role in promoting mesoderm and endoderm formation (for a review, see Ang and Constam, 2004; Zorn and Wells, 2007). Studies in frogs, fish, and mice indicate that the dose of nodal signaling directs a mesendoderm cell into either the endoderm or mesoderm lineage. For example, the use of hypomorphic nodal alleles in mouse revealed that high levels of nodal signaling are required for an endoderm fate whereas lower levels promote a mesoderm fate (Tremblay et al., 2000; Lowe et al., 2001; Vincent et al., 2003). These data suggest that nodal dose is important in endoderm vs. mesoderm fate. Consistent with this finding, endoderm cells arise from the anterior primitive streak that is close to the node, which is the principal source of nodal (Lawson and Pedersen, 1987; Lawson et al., 1991). Presumptive endoderm cells may also be exposed to higher levels of nodal ligand as they traverse the primitive streak and come into closer proximity with the node during their anterior migration. Given that Nodal activity is additionally regulated through proteolytic processing, secreted antagonists, and by the presence or absence of receptor complexes (Bouwmeester et al., 1996; Ding et al., 1998; Roebroek et al., 1998; Constam and Robertson, 2000; Perea-Gomez et al., 2002), it is clear that establishing the correct dose of Nodal is a critical step in specifying endoderm. There is also evidence that the Wnt signaling pathway is involved in the mesoderm vs. endoderm cell fate choice. Embryos lacking -catenin, a key effector of the canonical Wnt pathway, have ectopic mesoderm cells forming in the endoderm germ layer (Lickert et al., 2002). The transcription factors that act downstream of

3 3220 SPENCE AND WELLS the nodal and Wnt signals to direct endoderm formation are also remarkably conserved across vertebrate species and include Mix-like homeodomain proteins, Gata zinc finger factors, Sox HMG (high mobility group) domain factors, and Fox forkhead domain factors. In particular, Sox17 paralogs are required for normal endoderm development in all vertebrates analyzed (for review, see Zorn and Wells, 2007). Foregut Development and Pancreas Specification Once formed, the endoderm germ layer in mouse is a simple squamous epithelium (embryonic day [e] 7.5). Within 24 hr, morphogenetic movements in the anterior region of the endoderm result in the formation of the foregut and at this stage the endoderm cells are a cuboidal epithelium (e8.5 in mouse). The foregut will give rise to the thyroid, lungs, liver, stomach, and pancreas. The dynamic nature of foregut morphogenesis brings the endoderm into proximity with several mesodermal tissues that provide patterning signals to establish the presumptive organ domains within the foregut. Signaling pathways that have been implicated in foregut formation and patterning include the fibroblast growth factor (FGF), Wnt, retinoic acid (RA), and hedgehog pathways. These pathways are known to regulate the expression of key transcription factors, including Fox/HNF, ParaHox, and Hox factors, which are important mediators of cell fate (Grapin-Botton, 2005). Formation of the foregut depends on proper anterior posterior (A-P) patterning of the endoderm (Wells and Melton, 1999). Experiments in mouse and frog have demonstrated that gastrula stage endoderm cells are unspecified, but will adopt an A-P fate in response to signals from mesoderm (Wells and Melton, 2000; Horb and Slack, 2001). Subsequent studies identified that FGF and Wnt signaling pathways, which are restricted to the posterior at this stage of development, are key modulators of A-P patterning (Dessimoz et al., 2006; McLin et al., 2007). These pathways act to posteriorize endoderm at the gastrula stage and excluding FGF and Wnt signaling from the anterior is necessary for proper development of the foregut. As morphogenesis of the foregut commences, it has been shown that additional signaling pathways are involved in establishing the pancreatic domain within the developing gut tube. These pathways include the FGF, RA, and hedgehog (HH) signaling pathways. It is suggested that RA signaling is important for the global patterning of the foregut endoderm, because alterations in RA signaling cause defects in numerous foregut derivatives, including the lung, the stomach, and the pancreas. In particular, development of posterior/dorsal foregut derivatives like the stomach and dorsal pancreas are severely perturbed in embryos lacking active RA signaling due to absence of the enzyme retinaldehyde dehydrogenase 2 (Raldh2 knockout; Martin et al., 2005; Molotkov et al., 2005; Wang et al., 2006b). RA signaling in the foregut was shown to act upstream of key transcription factors, including the Pancreatic duodenal homeobox factor 1 (Pdx1) and several Hox genes, again suggesting that RA-mediated signaling functions to pattern the foregut before specification of the pancreatic domain (Kumar et al., 2003). In addition to Pdx1, which is necessary for proper pancreas development (Jonsson et al., 1994; Offield et al., 1996), there are several other transcription factors involved in foregut patterning and early pancreas development, including HNF6 (onecut), HNF1, Hlxb9, and Ptf1a/p48 (for review, see Jensen, 2004; Murtaugh, 2007). Hedgehog signaling is involved in global patterning of the gut tube and in early pancreatic development (Hebrok et al., 1998, 2000; Kim and Melton, 1998; Ramalho-Santos et al., 2000). During the early stages of gut tube development (e8.5 to e9 in the mouse), Sonic hedgehog (Shh) is broadly expressed by the endoderm of the developing gut tube along the A-P axis. Coincident with the start of pancreas development, Shh expression is repressed in the dorsal and ventral pancreatic buds. The repression of Shh in the dorsal bud depends on signals from the notochord that include FGF2 and activin (Hebrok et al., 1998). Deletion of the notochord results in ectopic expression of Shh in the dorsal pancreatic bud and loss of pancreatic gene expression (Kim et al., 1997). Inhibition of hedgehog signaling with the alkaloid Cyclopamine causes ectopic pancreatic buds to form in cultured gut tube explants (Kim and Melton, 1998). Pancreatic, Endocrine, and Exocrine Progenitor Cells After pancreatic specification and the onset of bud formation, there are continued signaling processes that regulate the proliferation and differentiation of progenitor cells for the endocrine and exocrine lineages. FGF10-mediated signaling is important for expansion of a pool of Pdx1 pancreatic progenitor cells (Bhushan et al., 2001). Notch- Delta signaling results in the expression of the NGN3 transcription factor in a subset of pancreatic cells, and cell lineage experiments have demonstrated that these Ngn3-expressing cells are a multipotent progenitor that gives rise to all cells of the endocrine lineage (Apelqvist et al., 1999; Gu et al., 2002). Loss- and gain-of-function experiments confirm that Ngn3 is key for endocrine cell development (Gradwohl et al., 2000; Schwitzgebel et al., 2000; Grapin-Botton et al., 2001). Of interest, a null mutation in human Ngn3 does not result in overt glucose intolerance, suggesting the existence of an Ngn3-independent pathway to endocrine cell development not revealed by studies in mice (Wang et al., 2006a). Additional transcription factors, specifically Ptf1a/p48, have been shown to be required for development of multiple pancreatic lineages (Krapp et al., 1998) and recent studies indicate the -catenin/wnt signaling pathway is required for maintenance of Ptf1a/p48 exocrine progenitor cells (Murtaugh et al., 2005; Wells et al., 2007). Microarray data indicate that there is a reduction in Ptf1a/p48 levels that coincide with a premature burst of exocrine gene expression in -catenin null pancreata at e14.5. One possibility is that there is premature differentiation of exocrine progenitor cells, which could explain why exocrine pancreas development appears to arrest these animals between e14.5 and e16.5 (Wells et al., 2007).

4 TRANSLATIONAL EMBRYOLOGY 3221 Specifying the Different Endocrine Lineages There are five endocrine cell types in the mature islet: glucagon-expressing -cells, insulin-expressing -cells, somatostatin-expressing -cells, ghrelin-expressing -cells, and pancreatic polypeptide-expressing cells (PP; Fig. 1). In addition to Ngn3, which is required for specification of all endocrine lineages in mice, loss-of-function experiments have identified several other transcription factors that play a role in specifying multiple endocrine lineages in the developing mouse pancreas (for review, see Jensen, 2004). The basic helix loop helix transcription factor NeuroD, the zinc-finger protein Insm1 and the LIM homeodomain protein Isl1 are all required for the development of multiple endocrine cell lineages and are thought to act downstream of Ngn3 (Ahlgren et al., 1997; Naya et al., 1997; Gierl et al., 2006). Expression of additional factors in a combinatorial manner further restricts cells into distinct endocrine lineages, including the /glucagon cells (Pax6, Nkx2.2, Foxa2, Arx), /somatostatin cells (Pax4, Pax6), PP cells (Nkx2.2), and /insulin cells (Pax4, Pax6, Nkx2.2, Nkx6.1, Hlxb9, Pdx1) (Sosa-Pineda et al., 1997; St-Onge et al., 1997; Sussel et al., 1998; Li et al., 1999; Lee et al., 2002; Collombat et al., 2003, 2007; Prado et al., 2004; Heller et al., 2005). Given the recent discovery of /ghrelin cells, it is not known what transcription factors are necessary for specification of this lineage in the pancreas, however, like the other endocrine lineages, these cells derive from a Ngn3 progenitor cell (Heller et al., 2005). At the phenotypic level, some of these transcription factors appear to act synergistically to promote one lineage, while at the same time act to antagonize the activity of other factors. For example, -cells seem to be replaced by -cells in Nkx2.2 null animals, suggesting that Nkx2.2 positively promote -cell fate, but also might be required to repress an -cell fate (Prado et al., 2004). At the biochemical level, there are relatively few studies that have shown how these factors synergistically regulate the transcription of cell-specific target genes during endocrine cell development (Edlund, 1998; Jensen, 2004). Also, surprisingly little is know about the signaling pathways that promote these specific endocrine lineages during embryonic development in vivo. As mentioned above, cell lineage studies have been important in identifying the developmental lineage of pancreatic endocrine cells. For example, it is thought that all pancreatic cell types transition through a Pdx1 and a Ptf1a/p48 stage during development (Gu et al., 2002; Kawaguchi et al., 2002) and that all endocrine cells are derived from a Ngn3 progenitor cell (Gu et al., 2002). Equally important have been cell lineage analyses that show how the different hormone lineages are not derived. For example, cells that express both insulin and glucagon have been described in the developing pancreas, and it has long been thought that these cells represent a common progenitor for - and -cells. However, lineage-tracing experiments using the insulin and glucagon promoters to label developing and -cells cells have demonstrated that mature -cells have never expressed glucagon and mature -cells have never expressed insulin (Herrera, 2000). Therefore, embryonic cells that express both insulin and glucagon are not a bipotential progenitor but are perhaps a developmental dead end. Similarly, it had been reported that multipotent progenitor cells in the pancreas may express Nestin (Zulewski et al., 2001), but expression and cell lineage experiments have clearly shown that this is not the case (Selander and Edlund, 2002; Treutelaar et al., 2003; Delacour et al., 2004). Below, we will discuss the importance of understanding the correct embryonic origin of the endocrine pancreas in attempts to generate endocrine cells from embryonic stem cells. TRANSLATING EMBRYOLOGY INTO DIRECTED DIFFERENTIATION OF EMBRYONIC STEM CELLS Differentiation of ESCs into tissues for replacement therapy is a challenging task. Several different protocols have been described for differentiation of mouse and human ESCs (mescs or hescs) into pancreatic cells. Successful attempts in efficiently differentiating ESCs into endoderm and pancreas have largely been due to the lessons learned from basic developmental biology research. We will focus on some recent breakthroughs, where ESCs have been efficiently differentiated into definitive endoderm and endocrine pancreas using the molecular pathways described above. Efficient Differentiation of Definitive Endoderm From ESCs Directing ESCs into the endoderm lineage is a prerequisite for generating therapeutic endoderm derivatives. As discussed above, nodal signaling is required for induction of mesoderm and endoderm in vertebrates. However, until recently, a commercial source of biologically active nodal protein was unavailable (Tada et al., 2005). Therefore, initial efforts to derive endoderm from both mouse and hescs used activin, another member of the TGF- superfamily (Kubo et al., 2004; D Amour et al., 2005). Activin has been used to mimic nodal activity in studies of mesoderm and endoderm formation during amphibian development (Smith et al., 1990; Thomsen et al., 1990; Gamer and Wright, 1995; Henry et al., 1996). Activin binds and activates the same receptors as nodal. However, activin does not require the coreceptor cripto for receptor binding, whereas nodal does (Gray et al., 2003). The use of activin to differentiate ESCs into endoderm was first reported with mouse ESCs using embryoid body formation (aggregated ESCs grown in suspension) and then using hescs differentiated as a monolayer (Kubo et al., 2004; D Amour et al., 2005). In both of these studies, the effects of activin on ESC differentiation were nearly identical to its effect on naive ectoderm cells (animal caps) in frog; high levels of activin induced the formation of endoderm cells (Fig. 2B). As with endoderm formation in vivo, definitive endoderm (DE) cells in culture express a unique combination of transcription factors, including Sox17 and Foxa2. One difference between mescs and hescs is in the efficiency of differentiation using different methods. Mouse embryoid bodies consisted of 50% endoderm, as measured by expression of Foxa2, where as hescs differentiated as

5 3222 SPENCE AND WELLS Fig. 2. Efficient differentiation of human embryonic stem cells (hescs) into definitive endoderm (DE). A: The ESCs are directed to become pancreatic endocrine cells over the course of 2 3 weeks. By adding various factors at different stages (see text and D Amour et al., 2006), hescs are first differentiated into a mesendodermal progenitor and then into DE. DE then transitions through stages reminiscent of primitive gut tube, foregut endoderm, pancreatic endoderm, and finally into the endocrine cell lineage. By analyzing different markers (shown under each cell stage), hesc differentiation is correlated with pancreas development in vivo. Figure modified from (D Amour et al., 2006). B: hesc differentiation into DE. The hesc cell line H9 was cultured using standard techniques. To induce differentiation, ESCs were cultured in RPMI Activin (100 ng/ml) and low serum (as described in D Amour et al., 2005) for 3 days. Before differentiation, hescs express Oct4 (image shows one ESC colony). After differentiation, 80% of cells are endoderm cells that coexpress Sox17 and FoxA2 (these cells are a monolayer). A small number of HESCs differentiate into mesoderm and express brachyury (TBra). monolayers consisted of 80% DE as measured by coexpression of Foxa2 and Sox17. In contrast, deriving definitive endoderm from hescs by means of embryoid body formation is much less efficient, with efficiencies ranging from 5 to 19% (Kim et al., 2007). Indeed, microarray analysis of hesc- DE derived by means of monolayer cultures indicates that numerous well-documented markers of DE are up-regulated after 3 days of culture in activin (Fig. 3). Whereas this protocol is highly efficient in producing DE ( 80%), it is important to point out that other cell lineages exist in these cultures at low levels including ectoderm, visceral endoderm, and mesoderm (Fig. 2B, TBra staining, and D Amour et al., 2005). This finding suggests that activin is not sufficient to fully repress these other cell types. More recently, efficient differentiation of mouse ESCs into mesoderm and endoderm was accomplished using the tetracycline transactivator system to drive expression of nodal (Takenaga et al., 2007). Nodal expression induced a mesendodermal progenitor population in 70% of cells when compared with treatment with exogenous activin, which induced a progenitor population 50%. More efficient differentiation of endoderm induced by nodal-expressing cells may occur for several reasons. It is known that commercially available nodal protein is much less active than Activin, and this finding is thought to be due, in part, to inefficient processing of nodal. However, in the above experiments, the ESCs produce their own nodal, and presumably are exposed to a constant, uniform level of biologi- Figure 3 Fig. 3. Microarray analysis of human embryonic stem cells (hescs) and human definitive endoderm (DE). A: RNA was isolated from undifferentiated hescs and from cells differentiated in Activin (100) for 3 days and subjected to Affymetrix microarray analysis. All experiments were performed in quadruplicate. Over 100 mr- NAs were either increased or decreased 3.5- fold during endoderm differentiation. B: The most highly up-regulated transcripts are welldocumented DE markers. References for endoderm gene expression are Gata3 (Debacker et al., 1999), Sox17 (Kanai-Azuma et al., 2002; Zorn and Wells, 2007), Isl1 (Lin et al., 2006), Foxa1 and Foxa2 (Friedman and Kaestner, 2006), and Cxcr4 (D Amour et al., 2005).

6 TRANSLATIONAL EMBRYOLOGY 3223 cally active nodal protein. It will be interesting to see if Nodal-conditioned medium will similarly promote efficient differentiation of hescs into definitive endoderm. That neither nodal nor activin are fully capable of transforming all of the ESCs into endoderm cells suggests that other factors play a role in endoderm formation. Nodal is thought to initiate gastrulation in vivo, which results in expression of several Wnt and FGF ligands in the primitive streak. These ligands are known to regulate gastrulation movements and cell fate cell fate decisions. In ESC cultures, it is likely that activin/nodal is important for both initiating a gastrulation-like process and for directing mesendoderm progenitors into the endoderm lineage. In support of this, activin treatment of ESCs induces the expression of gastrulation factors including Wnt and FGF ligands in a manner that is temporally similar to gastrulation in vivo (D Amour et al., 2005). Although in vivo it is not well understood how these factors act in combination to promote endoderm formation, ESC-derived endoderm cultures are a powerful tool to perform cellular, molecular, and biochemical analyses of this process. It is well documented in studies of Xenopus and zebrafish embryos that a common mesendoderm progenitor cell exists that gives rise to both the mesoderm and the endoderm (for review, see Zorn and Wells, 2007). It has not been conclusively demonstrated that mammals have a mesendoderm progenitor; however, one study in mouse suggests that such a cell exists in vivo (Lawson et al., 1991). In this cell lineage study, a population of cells in the anterior primitive streak was capable of giving rise to descendants that populate the mesoderm and endoderm germ layers, but not ectoderm. Recent studies have used mouse and hesc cultures as a basic research tool, and the data now support the idea that a common mesendoderm progenitor exists in mammals (Kubo et al., 2004; D Amour et al., 2005; Takenaga et al., 2007). For example, Kubo et al. sorted activin-treated ESCs using the mesodermal marker Brachyury (Bra, also called T or TBra; Wilkinson et al., 1990) and found that high levels of activin induced this Brachyury population of cells to express the endoderm markers Foxa2, Sox17, Hhex, and Albumin, whereas low activin-induced mesoderm markers. This finding suggested that the Brachyury-sorted cells were a bipotential mesendoderm progenitor. Tada et al. used a similar cell-sorting approach, which supports the existence of a mesendoderm progenitor cell (Tada et al., 2005). Under differentiation conditions, a population of Goosecoid-positive (Gsc ), E-cadherin positive (ECD ), platelet-derived growth factor receptor-alpha positive (PDGFR- ) cells is produced that will then give rise to two different populations of cells. One population is Gsc, ECD, PDGFR-, which will preferentially give rise to definitive endoderm. The other population of cells is Gsc, ECD, PDGFR-, and will differentiate into mesoderm (Tada et al., 2005). The hescs were shown to go through a mesendoderm-like state early during the endoderm-differentiation process in which cells coexpress mesoderm and endoderm markers such as Brachyury and Sox17. In these cultures, Brachyury is rapidly down-regulated and markers of the DE are up-regulated, including FoxA2 and Sox17 (D Amour et al., 2005). Although these experiments support the idea that some portion of the endoderm comes from a mesendoderm progenitor, they do not rule out the possibility that some endoderm cells are derived by means of a separate pathway as in Xenopus. Differentiation of ESC- Derived Endoderm Into the Pancreatic Lineage The aforementioned studies provide an important proof of principle that molecules that direct embryonic development in vivo can be used to direct the differentiation of ESCs into specific cell lineages in culture. While this remarkable finding highlights the utility of ESC cultures to study developmental biology, the therapeutic goal is to generate cells for replacement therapy. Because the DE derived from ESCs is theoretically capable of becoming any endoderm derivative (liver hepatocytes, lung alveolar cells, pancreatic -cells, etc.), the challenge now is to determine protocols for the efficient generation of specific and functional cell types. We will focus on a recent study that has successfully used developmental paradigms to generate pancreatic endocrine cells from human ESCs. D Amour et al. developed a protocol to generate pancreatic cells with 20% efficiency and insulin-producing cells with as high as 12% efficiency (D Amour et al., 2006). This was accomplished by manipulating many of the signaling pathways discussed above to mimic the in vivo development of the endoderm, foregut, pancreatic progenitors, and endocrine progenitor cells (Fig. 2A). In this stepwise differentiation protocol, hescs are first differentiated into DE with activin, then cells were treated with a combination of RA, FGF10, and cyclopamine (a hedgehog inhibitor). RA and cyclopamine are known to promote a posterior foregut/pancreatic fate in vivo, and in these cultures induces the expression of the posterior foregut markers Hnf6, Hnf1, Hlxb9, and Pdx1. In vivo, FGF10 is known to promote the proliferation of Pdx1 pancreatic progenitor cells and, in this context, appears to have a similar activity. In the later stages of this differentiation protocol, cells were cultured in the presence of extendin 4 (Ext4), insulin-like growth factor (IGF1), and hepatocyte growth factor (HGF). Although the role of these factors in pancreas development is not well defined, these factors have been shown to promote endocrine cell differentiation in various contexts (Anastasi et al., 2005; Tei et al., 2005). Although these cells have high insulin content, contain secretory granules, and secrete insulin in response to several secretagogues, they do not respond to glucose, suggesting that they are not mature -cells. Differentiation of ESCs into insulin cells is more efficient using monolayer cultures rather than embryoid body cultures. This finding is likely due to the fact that cells are more evenly exposed to the soluble factors in the medium, where as the three-dimensional structure of embryoid bodies will favor more cell cell communication resulting in secondary induction of other cell types. It therefore seems that efficient differentiation protocols based on three-dimensional cultures will be challenging, as supported by mouse ES protocols that are based on embryoid body formation where only 1% of the total popula-

7 3224 SPENCE AND WELLS tion of cells are insulin-positive after differentiation (Ku et al., 2004). While addition of factors such as activin increased the number of insulin-producing cells to almost 3%, this is still an inefficient process. One explanation for why embryoid body formation is an inefficient method was suggested by Mfopou et al., who showed that secondary inductions resulted in the expression of sonic hedgehog, a known inhibitor of pancreas development (Mfopou et al., 2005). It is possible that a combination of two- and three-dimensional culturing approaches might work best to derive functional pancreatic -cells. One such method was used to generate insulinproducing cells from hescs (Jiang et al., 2007a), where hesc monolayers were exposed to low activin and sodium butyrate to generate Sox17/Foxa2/ Cxcr4-positive DE, which was then scraped off the plate and grown in suspension. Over several weeks, FGF2, noggin, and endodermal growth factor were added to the medium to promote the formation of Pdx1-positive cells. This method resulted in up to 25% the cells becoming Pdx1-positive and 8% expressing insulin C-peptide. The C-peptide positive clusters of cells also contained glucagon- and somatostatinexpressing cells, which is reminiscent of pancreatic islets. Additionally, these islet-like structures contained secretory granules when examined by electron microscopy and were able to secrete C- peptide in response to glucose stimulation (Jiang et al., 2007a). Taken together the above studies could suggest that monolayer cultures are best suited for efficient differentiation of hescs into pancreatic and endocrine progenitor cells, whereas three-dimensional cultures might be more effective for maturation of progenitors into functional -cells. In support of this statement, several reports have shown that grafting ESC-derived Pdx1 cells in vivo promotes their maturation into -cells, and this process will be discussed below. Transplantation of Endoderm Tissue Derived From ESCs Toward the goal of generating therapeutics, it is necessary that ESC-derived tissues are capable of ameliorating disease symptoms in animal models. There have now been several reports investigating the impact of ESC-derived insulin cells in mouse models of type-1 diabetes. Several groups have reported the derivation of insulin-producing cells from mouse ESCs using a method based on selection of nestin progenitor cells (Lumelsky et al., 2001; Hori et al., 2002; Blyszczuk et al., 2003). One group transplanted these cells into streptozocin (STZ)-induced diabetic mice and observed the maintenance of insulin-expressing cells over different periods of time, without restoring euglycemia in STZ-treated animals (Lumelsky et al., 2001). Further characterization of the insulin-producing cells derived from this method showed that these cells were not pancreatic -cells but were neuronal. These insulin-producing cells rarely expressed C- peptide, were devoid of -cell secretory granules, and were unable to rescue STZ-induced hyperglycemia (Sipione et al., 2004). In some cases, insulin-expressing cells derived using the nestin selection method were able to rescue STZ-induced diabetes, however, this rescue ultimately failed because the transplanted cells formed teratomas (Fujikawa et al., 2005). These findings support the idea that, to generate functional -cells (and other tissues), protocols that recapitulate development are essential. While the nestin selection protocol is not useful for generating -cells, it has played an important role in our understanding of how (not) to experimentally approach ESC differentiation. Recent studies support the conclusion that human ESCs differentiated into endoderm and pancreatic lineages may have therapeutic potential. D Amour et al. have demonstrated that grafting human ESC-DE under the kidney capsule of mice promotes their further differentiation into more mature endodermal derivatives as measured by expression of markers for intestinal cells and liver hepatocytes (D Amour et al., 2005). Human embryoid bodies that were differentiated using activin and RA to enrich the FoxA2/Sox17/Pdx1 positive populations of cells were transplanted into STZ-treated mice (Shim et al., 2007). While insulin production was minimal in these cells before transplantation, after transplantation, the Pdx1-positive cells were more differentiated and expressed insulin (C-peptide and proinsulin) and glucagon. In some cases, insulin and glucagon were expressed in the same cell, reminiscent of the insulin/glucagon double-positive cells observed in the developing pancreas, which are not progenitors of -cells (Herrera, 2000). While these grafts rescued the hyperglycemia induced by STZ injections, it is not clear if any of the grafted cells were bona fide -cell progenitors capable of giving rise to mature, self-renewing -cells. A study by Jiang et al., resulted in a population of cells expressing C-peptide, insulin, glucagon, and glut2 before transplantation under the kidney capsule (Jiang et al., 2007b). After transplantation, the grafts contained multiple pancreatic cell types, including endocrine, ductal, and exocrine cells. Moreover, the endocrine population of cells appeared more mature, having secretory vesicles and being glucose responsive. These cells also expressed additional -cell markers Pdx1, Nkx6.1, and Pax6, among others and the grafted cells were able to rescue the STZ-induced hyperglycemia. Importantly, mice were followed for 3 months after transplantation and no teratoma formation was observed (Jiang et al., 2007b). Again, these studies highlight the importance of using methods of differentiation that will guide cells down a developmental path to becoming fully committed, mature cells that are not capable of uncontrolled growth. While the Jiang et al. study is promising, more thorough transplantation studies are needed to determine the risks (teratoma formation) of ESC-DE transplantations. PERSPECTIVES There has been great progress made over the past few years in directing ESCs into the pancreatic endocrine lineage. The successful approaches in this field have all been based on an embryonic blueprint where ESCs are differentiated in a step-wise manner that mimics pancreas development in vivo. Experimental approaches that deviate from embryonic principles have proven less effective at generating pancreatic cell types. For example, attempts to shortcut development by

8 TRANSLATIONAL EMBRYOLOGY 3225 expressing pancreatic transcription factors including Pdx1, Ngn3, and Pax4 in ESCs are not an efficient method to generate pancreatic cells (Blyszczuk et al., 2003; Vincent et al., 2006) for the likely reason that the ESCs are not yet competent to respond to these factors. Now that efficient protocols have been established for generating foregut endoderm cells, it would be interesting to re-evaluate the ability of these pancreatic transcription factors to efficiently direct differentiation. Differentiation protocols that were not based on pancreas development, such as the Nestin-selection approach discussed above that generated insulin-expressing neurons, have frustrated subsequent efforts at generating bona fide -cells. Studies of pancreas development continue to help researchers design better protocols and overcome obstacles toward generating mature -cells and eliminating residual hescs that are capable of forming teratomas. For example, most published ESC studies describe a population of insulin-expressing cells that arises early in these cultures that often times coexpresses other hormones such as glucagon. Lineage tracing studies have shown that these cells are probably not progenitors of the -cell lineage but are possibly the transient population of hormone-positive cells that arise early during pancreas development, which do not give rise to the endocrine cells found in adult animals (Herrera, 2000, 2002). Researchers are focusing on populations of cells that coexpress -cell progenitor markers (Nkx2.2, Nkx6.1, Pax4, Pax6) but are insulin. These may be progenitor cells that can become bona fide -cells that are capable of self-renewal. Whether or not early hormone-expressing cells in ESC cultures have an in vivo equivalent, or are aborted attempts at endocrine differentiation, it is clear that we still lack information on the full complement of factors that are required for generating functional endocrine cells. There is still a considerable amount of information in the current literature that could be used to improve existing ESC differentiation protocols. For example, little attention has been placed on developing conditions that promote endocrine vs. exocrine fate by controlling the levels of notch, TGF, and Wnt signaling. There is every reason to believe that further studies of normal pancreatic development and -cell maturation will continue to increase both the efficiency and functionality of HESC-derived insulin-expressing cells. ACKNOWLEDGMENTS J.M.W. and J.R.S. are supported by a Career Development Award from the Juvenile Diabetes Research Foundation and the NIH. J.R.S. is supported by an NIH training grant in Developmental and Perinatal Endocrinology. REFERENCES Ahlgren U, Pfaff SL, Jessell TM, Edlund T, Edlund H Independent requirement for ISL1 in formation of pancreatic mesenchyme and islet cells. Nature 385: Anastasi E, Santangelo C, Bulotta A, Dotta F, Argenti B, Mincione C, Gulino A, Maroder M, Perfetti R, Di Mario U The acquisition of an insulin-secreting phenotype by HGF-treated rat pancreatic ductal cells (ARIP) is associated with the development of susceptibility to cytokine-induced apoptosis. J Mol Endocrinol 34: Ang SL, Constam DB A gene network establishing polarity in the early mouse embryo. Semin Cell Dev Biol 15: 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: Bhushan A, Itoh N, Kato S, Thiery JP, Czernichow P, Bellusci S, Scharfmann R Fgf10 is essential for maintaining the proliferative capacity of epithelial progenitor cells during early pancreatic organogenesis. Development 128: Blyszczuk P, Czyz J, Kania G, Wagner M, Roll U, St-Onge L, Wobus AM Expression of Pax4 in embryonic stem cells promotes differentiation of nestin-positive progenitor and insulin-producing cells. Proc Natl Acad Sci U S A 100: Bouwmeester T, Kim S, Sasai Y, Lu B, De Robertis EM Cerberus is a headinducing secreted factor expressed in the anterior endoderm of Spemann s organizer. Nature 382: Brennan J, Lu CC, Norris DP, Rodriguez TA, Beddington RS, Robertson EJ Nodal signalling in the epiblast patterns the early mouse embryo. Nature 411: Collombat P, Mansouri A, Hecksher-Sorensen J, Serup P, Krull J, Gradwohl G, Gruss P Opposing actions of Arx and Pax4 in endocrine pancreas development. Genes Dev 17: Collombat P, Hecksher-Sorensen J, Krull J, Berger J, Riedel D, Herrera PL, Serup P, Mansouri A Embryonic endocrine pancreas and mature beta cells acquire alpha and PP cell phenotypes upon Arxmisexpression. JClinInvest117: Conlon FL, Lyons KM, Takaesu N, Barth KS, Kispert A, Herrmann B, Robertson EJ A primary requirement for nodal in the formation and maintenance of the primitive streak in the mouse. Development 120: Constam DB, Robertson EJ SPC4/ PACE4 regulates a TGFbeta signaling network during axis formation. Genes Dev 14: D Amour KA, Agulnick AD, Eliazer S, Kelly OG, Kroon E, Baetge EE Efficient differentiation of human embryonic stem cells to definitive endoderm. Nat Biotechnol 23: 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: Debacker C, Catala M, Labastie MC Embryonic expression of the human GATA-3 gene. Mech Dev 85: Delacour A, Nepote V, Trumpp A, Herrera PL Nestin expression in pancreatic exocrine cell lineages. Mech Dev 121: Dessimoz J, Opoka R, Kordich JJ, Grapin- Botton A, Wells JM FGF signaling is necessary for establishing gut tube domains along the anterior-posterior axis in vivo. Mech Dev 123: Ding J, Yang L, Yan YT, Chen A, Desai N, Wynshaw-Boris A, Shen MM Cripto is required for correct orientation of the anterior-posterior axis in the mouseembryo[seecomment].nature395: Edlund H Transcribing pancreas. Diabetes 47: Friedman JR, Kaestner KH The Foxa family of transcription factors in development and metabolism. Cell Mol Life Sci 63: Fujikawa T, Oh SH, Pi L, Hatch HM, Shupe T, Petersen BE Teratoma formation leads to failure of treatment for type I diabetes using embryonic stem cell-derived insulin-producing cells. Am J Pathol 166: Gamer LW, Wright CV Autonomous endodermal determination in Xenopus: regulation of expression of the pancreatic gene XlHbox 8. Dev Biol 171: Gierl MS, Karoulias N, Wende H, Strehle M, Birchmeier C The zinc-finger factor Insm1 (IA-1) is essential for the development of pancreatic beta cells and intestinal endocrine cells. Genes Dev 20: Gradwohl G, Dierich A, LeMeur M, Guillemot F neurogenin3 is required for the development of the four endocrine

9 3226 SPENCE AND WELLS cell lineages of the pancreas. Proc Natl Acad Sci U S A 97: Grapin-Botton A Antero-posterior patterning of the vertebrate digestive tract: 40 years after Nicole Le Douarin s PhD thesis. Int J Dev Biol 49: Grapin-Botton A, Majithia AR, Melton DA Key events of pancreas formation are triggered in gut endoderm by ectopic expression of pancreatic regulatory genes. Genes Dev 15: Gray PC, Harrison CA, Vale W Cripto forms a complex with activin and type II activin receptors and can block activin signaling. Proc Natl Acad Sci U S A 100: 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: Hebrok M, Kim SK, Melton DA Notochord repression of endodermal Sonic hedgehog permits pancreas development. Genes Dev 12: Hebrok M, Kim SK, St Jacques B, McMahon AP, Melton DA Regulation of pancreas development by hedgehog signaling. Development 127: Heller RS, Jenny M, Collombat P, Mansouri A, Tomasetto C, Madsen OD, Mellitzer G, Gradwohl G, Serup P Genetic determinants of pancreatic epsiloncell development. Dev Biol 286: Henry GL, Brivanlou IH, Kessler DS, Hemmati-Brivanlou A, Melton DA TGF-beta signals and a pattern in Xenopus laevis endodermal development. Development 122: Herrera PL Adult insulin- and glucagon-producing cells differentiate from two independent cell lineages. Development 127: Herrera PL Defining the cell lineages of the islets of Langerhans using transgenic mice. Int J Dev Biol 46: Horb ME, Slack JM Endoderm specification and differentiation in Xenopus embryos. Dev Biol 236: Hori Y, Rulifson IC, Tsai BC, Heit JJ, Cahoy JD, Kim SK Growth inhibitors promote differentiation of insulinproducing tissue from embryonic stem cells. Proc Natl Acad Sci U S A 19:19. Jensen J Gene regulatory factors in pancreatic development. Dev Dyn 229: Jiang J, Au M, Lu K, Eshpeter A, Korbutt G, Fisk G, Majumdar AS. 2007a. Generation of insulin-producing islet-like clusters from human embryonic stem cells. Stem Cells 25: Jiang W, Shi Y, Zhao D, Chen S, Yong J, Zhang J, Qing T, Sun X, Zhang P, Ding M, Deng H. 2007b. In vitro derivation of functional insulin-producing cells from human embryonic stem cells. Cell Res 17: Jonsson J, Carlsson L, Edlund T, Edlund H Insulin-promoter-factor 1 is required for pancreas development in mice. Nature 371: Kanai-Azuma M, Kanai Y, Gad JM, Tajima Y, Taya C, Kurohmaru M, Sanai Y, Yonekawa H, Yazaki K, Tam PP, Hayashi Y Depletion of definitive gut endoderm in Sox17-null mutant mice. Development 129: 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: Kim SK, Melton DA Pancreas development is promoted by cyclopamine, a hedgehog signaling inhibitor. Proc Natl Acad Sci U S A 95: Kim SK, Hebrok M, Melton DA Notochord to endoderm signaling is required for pancreas development. Development 124: Kim SE, Lee WJ, Choi KY The PI3 kinase-akt pathway mediates Wnt3a-induced proliferation. Cell Signal 19: Krapp A, Knofler M, Ledermann B, Burki K, Berney C, Zoerkler N, Hagenb chle O, Wellauer PK The bhlh protein PTF1 p48 is essential for the formation of the exocrine and the correct spatial organization of the endocrine pancreas. Genes Dev 12: Ku HT, Zhang N, Kubo A, O Connor R, Mao M, Keller G, Bromberg JS Committing embryonic stem cells to early endocrine pancreas in vitro. Stem Cells 22: Kubo A, Shinozaki K, Shannon J, Kouskoff V, Kennedy M, Woo S, Fehling H, Keller G Development of definitive endoderm from embryonic stem cells in culture. Development 131: Kumar M, Jordan N, Melton D, Grapin- Botton A Signals from lateral plate mesoderm instruct endoderm toward a pancreatic fate. Dev Biol 259: Lawson KA, Pedersen RA Cell fate, morphogenetic movement and population kinetics of embryonic endoderm at the time of germ layer formation in the mouse. Development 101: Lawson KA, Meneses JJ, Pedersen RA Clonal analysis of epiblast fate during germ layer formation in the mouse embryo. Development 113: Lee CS, Sund NJ, Vatamaniuk MZ, Matschinsky FM, Stoffers DA, Kaestner KH Foxa2 controls Pdx1 gene expression in pancreatic beta-cells in vivo. Diabetes 51: Lewis SL, Tam PP Definitive endoderm of the mouse embryo: formation, cell fates, and morphogenetic function. Dev Dyn 235: Li H, Arber S, Jessell TM, Edlund H Selective agenesis of the dorsal pancreas in mice lacking homeobox gene Hlxb9. Nat Genet 23: Lickert H, Kutsch S, Kanzler B, Tamai Y, Taketo MM, Kemler R Formation of multiple hearts in mice following deletion of beta-catenin in the embryonic endoderm. Dev Cell 3: Lin L, Bu L, Cai CL, Zhang X, Evans S Isl1 is upstream of sonic hedgehog in a pathway required for cardiac morphogenesis. Dev Biol 295: Lowe LA, Yamada S, Kuehn MR Genetic dissection of nodal function in patterning the mouse embryo. Development 128: Lumelsky N, Blondel O, Laeng P, Velasco I, Ravin R, McKay R Differentiation of embryonic stem cells to insulinsecreting structures similar to pancreatic islets. Science 292: Martin M, Gallego-Llamas J, Ribes V, Kedinger M, Niederreither K, Chambon P, Dolle P, Gradwohl G Dorsal pancreas agenesis in retinoic acid-deficient Raldh2 mutant mice. Dev Biol 284: McLin VA, Rankin SA, Zorn AM Repression of Wnt/beta-catenin signaling in the anterior endoderm is essential for liver and pancreas development. Development 134: Mfopou JK, Willems E, Leyns L, Bouwens L Expression of regulatory genes for pancreas development during murine embryonic stem cell differentiation. Int J Dev Biol 49: Molotkov A, Molotkova N, Duester G Retinoic acid generated by Raldh2 in mesoderm is required for mouse dorsal endodermal pancreas development. Dev Dyn 232: Murtaugh LC Pancreas and betacell development: from the actual to the possible. Development 134: Murtaugh LC, Law AC, Dor Y, Melton DA Beta-catenin is essential for pancreatic acinar but not islet development. Development 132: Naya FJ, Huang HP, Qiu Y, Mutoh H, De- Mayo FJ, Leiter AB, Tsai MJ Diabetes, defective pancreatic morphogenesis, and abnormal enteroendocrine differentiation in BETA2/neuroD-deficient mice. Genes Dev 11: Offield MF, Jetton TL, Labosky PA, Ray M, Stein RW, Magnuson MA, Hogan BL, Wright CV PDX-1 is required for pancreatic outgrowth and differentiation of the rostral duodenum. Development 122: Perea-Gomez A, Vella FD, Shawlot W, Oulad-Abdelghani M, Chazaud C, Meno C, Pfister V, Chen L, Robertson E, Hamada H, Behringer RR, Ang SL Nodal antagonists in the anterior visceral endoderm prevent the formation of multiple primitive streaks. Dev Cell 3: Prado CL, Pugh-Bernard AE, Elghazi L, Sosa-Pineda B, Sussel L Ghrelin cells replace insulin-producing beta cells in two mouse models of pancreas development. Proc Natl Acad Sci U S A 101: Ramalho-Santos M, Melton DA, McMahon AP Hedgehog signals regulate multiple aspects of gastrointestinal development. Development 127: Roebroek AJ, Umans L, Pauli IG, Robertson EJ, van Leuven F, Van de Ven WJ,

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

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

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

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

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

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

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

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

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

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

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

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

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

Axis Formation and Mesoderm Induction

Axis Formation and Mesoderm Induction Developmental Biology Biology 4361 Axis Formation and Mesoderm Induction October 27, 2005 Amphibian anteroposterior specification polarized eggs animal/vegetal pigment yolk v. clear cytoplasm mitochondrial

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

Generation of Insulin-Producing Islet-Like Clusters from Human Embryonic Stem Cells

Generation of Insulin-Producing Islet-Like Clusters from Human Embryonic Stem Cells EMBRYONIC STEM CELLS Generation of Insulin-Producing Islet-Like Clusters from Human Embryonic Stem Cells JIANJIE JIANG, a MELINDA AU, a KUANGHUI LU, a ALANA ESHPETER, b GREGORY KORBUTT, b GREG FISK, a

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

The Evolution and Development of the Gut. Dr Mike Wride School of Natural Sciences Zoology Department

The Evolution and Development of the Gut. Dr Mike Wride School of Natural Sciences Zoology Department The Evolution and Development of the Gut Dr Mike Wride School of Natural Sciences Zoology Department email: wridem@tcd.ie The gut? Gut Function and Regulation (Dr. Alan Tuffery) Absorption of nutrients

More information

Vertebrate Limb Patterning

Vertebrate Limb Patterning Vertebrate Limb Patterning What makes limb patterning an interesting/useful developmental system How limbs develop Key events in limb development positioning and specification initiation of outgrowth establishment

More information

Cell Type Nervous System I. Developmental Readout. Foundations. Stem cells. Organ formation. Human issues.

Cell Type Nervous System I. Developmental Readout. Foundations. Stem cells. Organ formation. Human issues. 7.72 10.11.06 Cell Type Nervous System I Human issues Organ formation Stem cells Developmental Readout Axes Cell type Axon guidance 3D structure Analysis Model + + organisms Foundations Principles 1 What

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

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

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

Tetrapod Limb Development

Tetrapod Limb Development IBS 8102 Cell, Molecular and Developmental Biology Tetrapod Limb Development February 11, 2008 Tetrapod Limbs Merlin D. Tuttle Vicki Lockard and Paul Barry Father Alejandro Sanchez Anne Fischer Limb Patterning

More information

Regionalization of the nervous system. Paul Garrity 7.68J/9.013J February 25, 2004

Regionalization of the nervous system. Paul Garrity 7.68J/9.013J February 25, 2004 Regionalization of the nervous system Paul Garrity 7.68J/9.013J February 25, 2004 Patterning along: Rostral/Caudal (AP) axis Dorsal/Ventral (DV) axis Start with DV axial patterning in Spinal Cord Dorsal/Ventral

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

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

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

Embryonic stem cells to beta-cells by understanding pancreas development

Embryonic stem cells to beta-cells by understanding pancreas development Embryonic stem cells to beta-cells by understanding pancreas development Marie Best, Michael Carroll, Neil A. Hanley, Karen Piper Hanley To cite this version: Marie Best, Michael Carroll, Neil A. Hanley,

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

Polarity and Segmentation. Chapter Two

Polarity and Segmentation. Chapter Two Polarity and Segmentation Chapter Two Polarization Entire body plan is polarized One end is different than the other Head vs. Tail Anterior vs. Posterior Front vs. Back Ventral vs. Dorsal Majority of neural

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

Combined ectopic expression of Pdx1 and Ptf1a/p48 results in the stable conversion of posterior endoderm into endocrine and exocrine pancreatic tissue

Combined ectopic expression of Pdx1 and Ptf1a/p48 results in the stable conversion of posterior endoderm into endocrine and exocrine pancreatic tissue RESEARCH COMMUNICATION Combined ectopic expression of Pdx1 and Ptf1a/p48 results in the stable conversion of posterior endoderm into endocrine and exocrine pancreatic tissue Solomon Afelik, Yonglong Chen,

More information

EMBRYONIC STEM CELLS/INDUCED PLURIPOTENT STEM CELLS

EMBRYONIC STEM CELLS/INDUCED PLURIPOTENT STEM CELLS EMBRYONIC STEM CELLS/INDUCED PLURIPOTENT STEM CELLS Biphasic Induction of Pdx1 in Mouse and Human Embryonic Stem Cells Can Mimic Development of Pancreatic -Cells ANDREIA S. BERNARDO, a CANDY H.-H. CHO,

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

Supplementary Figure 1. A microarray screen of organizers compared to non-organizer tissue reveals a putative organizer gene set.

Supplementary Figure 1. A microarray screen of organizers compared to non-organizer tissue reveals a putative organizer gene set. Supplementary Figure 1. A microarray screen of organizers compared to non-organizer tissue reveals a putative organizer gene set. (a, b) Venn diagrams of 31 enriched (a) and 17 depleted (b) genes significantly

More information

Derivation of insulin-producing cells from human embryonic stem cells

Derivation of insulin-producing cells from human embryonic stem cells Stem Cell Research (2009) 3, 73 87 available at www.sciencedirect.com www.elsevier.com/locate/scr REVIEW Derivation of insulin-producing cells from human embryonic stem cells Dennis Van Hoof a, Kevin A.

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

3D differentiation enhances the efficiency of differentiation of human induced pluripotent stem cells to insulin producing cells

3D differentiation enhances the efficiency of differentiation of human induced pluripotent stem cells to insulin producing cells University of Iowa Iowa Research Online Theses and Dissertations Fall 2014 3D differentiation enhances the efficiency of differentiation of human induced pluripotent stem cells to insulin producing cells

More information

Citation for published version: Myatt, E-J 2013, 'Differentiation of Pancreatic and Hepatic Cell Types', Ph.D., University of Bath.

Citation for published version: Myatt, E-J 2013, 'Differentiation of Pancreatic and Hepatic Cell Types', Ph.D., University of Bath. Citation for published version: Myatt, E-J 2013, 'Differentiation of Pancreatic and Hepatic Cell Types', Ph.D., University of Bath. Publication date: 2013 Document Version Publisher's PDF, also known as

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

Tetrapod Limb Development

Tetrapod Limb Development Biology 4361 Developmental Biology Tetrapod Limb Development July 29, 2009 Tetrapod Limbs Merlin D. Tuttle Vicki Lockard and Paul Barry Father Alejandro Sanchez Anne Fischer Limb Development - Overview

More information

Sonic Hedgehog and Other Soluble Factors from Differentiating Embryoid Bodies Inhibit Pancreas Development

Sonic Hedgehog and Other Soluble Factors from Differentiating Embryoid Bodies Inhibit Pancreas Development EMBRYONIC STEM CELLS Sonic Hedgehog and Other Soluble Factors from Differentiating Embryoid Bodies Inhibit Pancreas Development JOSUÉ K. MFOPOU, a VÉRONIQUE DE GROOTE, a XIABO XU, b HARRY HEIMBERG, b LUC

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

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

Urogenital Development

Urogenital Development 2-5-03 Urogenital Development Greg Dressler Assoc. Professor Dept. of Pathology x46490 Dressler@umich.edu The Origin of the Kidney In the vertebrate embryo, the first stage of kidney development occurs

More information

Differentiation of Mouse Embryonic Stem Cells into Endoderm without Embryoid Body Formation

Differentiation of Mouse Embryonic Stem Cells into Endoderm without Embryoid Body Formation Differentiation of Mouse Embryonic Stem Cells into Endoderm without Embryoid Body Formation Peter T. W. Kim 1, Brad G. Hoffman 1, Annette Plesner 2, Cheryl D. Helgason 1, C. Bruce Verchere 2, Stephen W.

More information

Characterization of ALDH Positive Cells in the Human Fetal Pancreas

Characterization of ALDH Positive Cells in the Human Fetal Pancreas Western University Scholarship@Western Electronic Thesis and Dissertation Repository December 2014 Characterization of ALDH Positive Cells in the Human Fetal Pancreas Amanda Oakie The University of Western

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

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

NIH Public Access Author Manuscript Expert Opin Biol Ther. Author manuscript; available in PMC 2012 January 1.

NIH Public Access Author Manuscript Expert Opin Biol Ther. Author manuscript; available in PMC 2012 January 1. NIH Public Access Author Manuscript Published in final edited form as: Expert Opin Biol Ther. 2011 January ; 11(1): 41 53. doi:10.1517/14712598.2011.540235. Stem cell-based strategies for the treatment

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

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

Tetrapod Limb Development

Tetrapod Limb Development Biology 4361 Developmental Biology Tetrapod Limb Development July 29, 2009 Tetrapod Limbs Merlin D. Tuttle Vicki Lockard and Paul Barry Father Alejandro Sanchez Anne Fischer Limb Development - Overview

More information

Heart Development. Origins of congenital heart defects Properties of cardiac progenitor cells. Robert G. Kelly

Heart Development. Origins of congenital heart defects Properties of cardiac progenitor cells. Robert G. Kelly ESC CBCS Summer School on Cardiovascular Sciences Heart Development 19th June 2013 Origins of congenital heart defects Properties of cardiac progenitor cells Robert G. Kelly Animal models of heart development

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

Prolonged FGF signaling is necessary for lung and liver induction in Xenopus

Prolonged FGF signaling is necessary for lung and liver induction in Xenopus Shifley et al. BMC Developmental Biology 2012, 12:27 RESEARCH ARTICLE Open Access Prolonged FGF signaling is necessary for lung and liver induction in Xenopus Emily T Shifley 1, Alan P Kenny 2, Scott A

More information

EMBRYONIC STEM CELLS/INDUCED PLURIPOTENT STEM CELLS

EMBRYONIC STEM CELLS/INDUCED PLURIPOTENT STEM CELLS EMBRYONIC STEM CELLS/INDUCED PLURIPOTENT STEM CELLS FGF2 Specifies hesc-derived Definitive Endoderm into Foregut/Midgut Cell Lineages in a Concentration-Dependent Manner JACQUELINE AMERI, ANDERS STÅHLBERG,

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

Lecture IV. Mechanisms of Neural. Neural Development

Lecture IV. Mechanisms of Neural. Neural Development Lecture IV. Mechanisms of Neural Bio 3411 Monday 1 Readings NEUROSCIENCE: 5 th ed, pp 477-506 (sorta) 4 th ed, pp 545-575 (sorta) References : Fainsod, A., Steinbeisser, H., & De Robertis, E. M. (1994).

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

Modeling Developmental Hematopoiesis Using Pluripotent Stem Cells

Modeling Developmental Hematopoiesis Using Pluripotent Stem Cells Modeling Developmental Hematopoiesis Using Pluripotent Stem Cells Christopher Sturgeon February 14, 2017 Pluripotent Stem Cells self-renewal hpsc Mesoderm blood cardiovascular muscle Endoderm lung liver

More information

Patterning the Embryo

Patterning the Embryo Patterning the Embryo Anteroposterior axis Regional Identity in the Vertebrate Neural Tube Fig. 2.2 1 Brain and Segmental Ganglia in Drosophila Fig. 2.1 Genes that create positional and segment identity

More information

Heart Development. Robert G. Kelly Developmental Biology Institute of Marseilles - Luminy

Heart Development. Robert G. Kelly Developmental Biology Institute of Marseilles - Luminy ESC CBCS Summer School on Cardiovascular Sciences 15th June 2011 Heart Development Robert G. Kelly Developmental Biology Institute of Marseilles - Luminy Animal models of heart development Tinman/Nkx2.5

More information

CNS Developmental. Anke van Eekelen, PhD. Telethon Institute for Child Health Research

CNS Developmental. Anke van Eekelen, PhD. Telethon Institute for Child Health Research CNS Developmental Anke van Eekelen, PhD Telethon Institute for Child Health Research (Some slides are modified versions of Prof. Alan Harvey s Neuroscience lecture at ANHB and Dr. Joanne Britto s Dev Neuroscience

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

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

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

How to make a functional β-cell

How to make a functional β-cell 2472 Development 40, 2472-2483 (203) doi:0.242/dev.09387 203. Published by The Company of Biologists Ltd How to make a functional β-cell Felicia W. Pagliuca,2 and Douglas A. Melton,2, * Summary Insulin-secreting

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

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

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

RECAPITULATING PANCREATIC DEVELOPMENT IN INDUCING IN-VITRO HUMAN EMBRYONIC STEM CELL DIFFERENTIATION. Maria Jaramillo

RECAPITULATING PANCREATIC DEVELOPMENT IN INDUCING IN-VITRO HUMAN EMBRYONIC STEM CELL DIFFERENTIATION. Maria Jaramillo RECAPITULATING PANCREATIC DEVELOPMENT IN INDUCING IN-VITRO HUMAN EMBRYONIC STEM CELL DIFFERENTIATION TITLE by Maria Jaramillo B.S Biomedical Engineering, Florida International University, 2006 Submitted

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

a) They are the most common cause of pediatric kidney failure. b) They are always symptomatic. c) They can be asymmetric.

a) They are the most common cause of pediatric kidney failure. b) They are always symptomatic. c) They can be asymmetric. Practice questions: 1. The paraxial mesoderm gives rise to somites. The structure of the somite a) is a loose mesenchymal sheet that will migrate toward the notochord. b) is an epithelial rosette with

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 Figure S1. Monolayer differentiation of mouse ESCs into telencephalic neural precursors. (a) Schematic representation of the protocols

Supplementary Figure S1. Monolayer differentiation of mouse ESCs into telencephalic neural precursors. (a) Schematic representation of the protocols Supplementary Figure S1. Monolayer differentiation of mouse ESCs into telencephalic neural precursors. (a) Schematic representation of the protocols used to differentiate mouse ESCs. (b) Representative

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

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

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

WNT2 and WNT7B Cooperative Signaling in Lung Development

WNT2 and WNT7B Cooperative Signaling in Lung Development University of Pennsylvania ScholarlyCommons Publicly Accessible Penn Dissertations 1-1-2012 WNT2 and WNT7B Cooperative Signaling in Lung Development Mayumi Miller University of Pennsylvania, mayumimiller@gmail.com

More information

Spatiotemporally regulated expression of transcription

Spatiotemporally regulated expression of transcription ORIGINAL ARTICLE Reduction of Ptf1a Gene Dosage Causes Pancreatic Hypoplasia and Diabetes in Mice Akihisa Fukuda, 1,2,3 Yoshiya Kawaguchi, 1 Kenichiro Furuyama, 1 Sota Kodama, 1 Masashi Horiguchi, 1 Takeshi

More information

From crypt stem cell to colorectal cancer

From crypt stem cell to colorectal cancer 19 3 2007 6 Chinese Bulletin of Life Sciences Vol. 19, No. 3 Jun., 2007 1004-0374(2007)03-0321-05 ( 510405) Wnt Notch BMP R735.35; R730.21 A From crypt stem cell to colorectal cancer WEN Bin*, CHEN Weiwen

More information

Transcription factor expression in the developing human fetal endocrine pancreas

Transcription factor expression in the developing human fetal endocrine pancreas Diabetologia (8) 5:9 8 DOI.7/s5-8--z ARTICLE Transcription factor expression in the developing human fetal endocrine pancreas B. M. Lyttle & J. Li & M. Krishnamurthy & F. Fellows & M. B. Wheeler & C. G.

More information

FGF4 and retinoic acid direct differentiation of hescs into PDX1-expressing foregut endoderm in a time- and concentration-dependent manner.

FGF4 and retinoic acid direct differentiation of hescs into PDX1-expressing foregut endoderm in a time- and concentration-dependent manner. FGF4 and retinoic acid direct differentiation of hescs into PDX1-expressing foregut endoderm in a time- and concentration-dependent manner. Johannesson, Martina; Ståhlberg, Anders; Ameri, Jacqueline; Wolfhagen

More information

Cell therapeutics for the Insulin-Dependent Diabetes Mellitus

Cell therapeutics for the Insulin-Dependent Diabetes Mellitus Cell therapeutics for the Insulin-Dependent Diabetes Mellitus Haekwon Kim Dept. of Biotechnology Seoul Women s University Introduction Type I diabetes is caused by the autoimmune destruction of pancreatic

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

University of Groningen. Embryogenesis and neogenesis of the endocrine pancreas Gangaram-Panday, Shanti Tireshma

University of Groningen. Embryogenesis and neogenesis of the endocrine pancreas Gangaram-Panday, Shanti Tireshma University of Groningen Embryogenesis and neogenesis of the endocrine pancreas Gangaram-Panday, Shanti Tireshma IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you

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

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

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

Biology Developmental Biology Spring Quarter Midterm 1 Version A

Biology Developmental Biology Spring Quarter Midterm 1 Version A Biology 411 - Developmental Biology Spring Quarter 2013 Midterm 1 Version A 75 Total Points Open Book Choose 15 out the 20 questions to answer (5 pts each). Only the first 15 questions that are answered

More information

Lung Physiology. Jamie Havrilak, PhD Postdoctoral Research Associate Layden Lab October 26th, 2018

Lung Physiology. Jamie Havrilak, PhD Postdoctoral Research Associate Layden Lab October 26th, 2018 Lung Physiology Jamie Havrilak, PhD Postdoctoral Research Associate Layden Lab October 26th, 2018 Nkx2.1- Lung epithelium Endomucin- Vasculature Alveoli/Capillaries: Site of Gas Exchange in the Lung

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

Making Mature Human Islet Cells from Stem Cells to Model Disease and Treat Diabetes

Making Mature Human Islet Cells from Stem Cells to Model Disease and Treat Diabetes University of British Columbia Departments of Surgery and Cellular & Physiological Sciences Making Mature Human Islet Cells from Stem Cells to Model Disease and Treat Diabetes 2016 International Conference

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

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland AD Award Number: W81XWH-12-1-0212 TITLE: Wnt/Beta-Catenin, Foxa2, and CXCR4 Axis Controls Prostate Cancer Progression PRINCIPAL INVESTIGATOR: Xiuping Yu CONTRACTING ORGANIZATION: Vanderbilt University

More information

Supplementary Figure 1.

Supplementary Figure 1. Supplementary Figure 1. Increased expression of cell cycle pathway genes in insulin + Glut2 low cells of STZ-induced diabetic islets. A) random blood glucose measuers of STZ and vehicle treated MIP-GFP

More information

Convergence of cmyc and β-catenin on Tcf7l1 enables endoderm specification

Convergence of cmyc and β-catenin on Tcf7l1 enables endoderm specification Manuscript EMBO-2015-92116 Convergence of cmyc and β-catenin on Tcf7l1 enables endoderm specification Gillian Morrison, Roberta Scognamiglio, Andreas Trumpp, and Austin Smith Corresponding author: Gillian

More information

Formation of Urine: Formation of Urine

Formation of Urine: Formation of Urine The Urinary outflow tract: monitors and regulates extra-cellular fluids excretes harmful substances in urine, including nitrogenous wastes (urea) returns useful substances to bloodstream maintain balance

More information