CB29CH19-Sander

ARI

24 July 2013

V I E W

A

Review in Advance first posted online on July 31, 2013. (Changes may still occur before final publication online and in print.)

N

I N

C E

S

R

E

12:44

D V A

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

Pancreas Organogenesis: From Lineage Determination to Morphogenesis Hung Ping Shih,∗ Allen Wang,∗ and Maike Sander Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, California 92093-0695; email: [email protected]

Annu. Rev. Cell Dev. Biol. 2013. 29:19.1–19.25

Keywords

The Annual Review of Cell and Developmental Biology is online at http://cellbio.annualreviews.org

pancreas, development, progenitor, exocrine, endocrine, pluripotent stem cell

This article’s doi: 10.1146/annurev-cellbio-101512-122405 c 2013 by Annual Reviews. Copyright  All rights reserved ∗

Equal contributions.

Abstract The pancreas is an essential organ for proper nutrient metabolism and has both endocrine and exocrine function. In the past two decades, knowledge of how the pancreas develops during embryogenesis has significantly increased, largely from developmental studies in model organisms. Specifically, the molecular basis of pancreatic lineage decisions and cell differentiation is well studied. Still not well understood are the mechanisms governing threedimensional morphogenesis of the organ. Strategies to derive transplantable β-cells in vitro for diabetes treatment have benefited from the accumulated knowledge of pancreas development. In this review, we provide an overview of the current understanding of pancreatic lineage determination and organogenesis, and we examine future implications of these findings for treatment of diabetes mellitus through cell replacement.

19.1

Changes may still occur before final publication online and in print

CB29CH19-Sander

ARI

24 July 2013

12:44

Contents

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

INTRODUCTION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19.2 OVERVIEW OF PANCREAS DEVELOPMENT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19.2 LINEAGE DECISIONS DURING PANCREAS DEVELOPMENT . . . . . . . . . . . . . . . 19.5 Pancreas Induction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19.5 Establishing and Maintaining Pancreatic Identity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19.7 Signaling During Early Pancreatic Growth . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19.8 Compartmentalizing the Organ: Patterning of Tips and Trunk . . . . . . . . . . . . . . . . . . . 19.9 Development of the Acinar Lineage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .19.10 The Endocrine Versus Ductal Cell-Fate Choice . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .19.11 Differentiation of the Islet Cell Types . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .19.12 PANCREAS MORPHOGENESIS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .19.13 Intracellular Signals Governing Pancreas Morphogenesis . . . . . . . . . . . . . . . . . . . . . . . . .19.14 Coordinating Morphogenesis and Cell Differentiation . . . . . . . . . . . . . . . . . . . . . . . . . . . .19.14 Morphogenetic Changes and Endocrine Cell Differentiation . . . . . . . . . . . . . . . . . . . . . .19.15 APPLICATION OF DEVELOPMENTAL PRINCIPLES TOWARD STEM CELL DIFFERENTIATION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .19.15 Generating Pancreatic Progenitors from Human Pluripotent Stem Cells . . . . . . . . . .19.16 Beyond Progenitors: How to Generate Functional Endocrine Cells In Vitro . . . . . . .19.16 Human Pluripotent Stem Cells as a Model for Understanding Pancreas Biology and Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .19.17 FUTURE PROSPECTS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .19.18

INTRODUCTION The pancreas is essential for nutrient metabolism and has both exocrine and endocrine functions. The exocrine pancreas consists of acinar and ductal cells, whose prime role is nutrient digestion. The acinar cells secrete pancreatic juice containing enzymes that catalyze the breakdown of proteins, carbohydrates, and lipids. Upon release from the acinar cells, the pancreatic juice is transported into the duodenum via the ductal system (Figure 1). Functioning largely independent from exocrine cells, the endocrine pancreas is responsible for regulating glucose homeostasis by releasing hormones into the blood stream. The endocrine pancreas comprises five endocrine cell types, which reside in small cell clusters called islets of Langerhans. The biology of the pancreas has been studied intensely, largely driven by the hope of finding better treatments for devastating pancreatic diseases, such as diabetes mellitus, pancreatitis, and pancreatic adenocarcinoma. In particular, advancements in stem cell technology have recently sparked optimism that diabetes could be cured by harnessing stem cells for therapeutic use. This has led to heightened interest in understanding embryonic development of the pancreas, specifically the events involved in cell fate decisions and endocrine cell differentiation. Recent successes in deriving pancreatic cells from human pluripotent stem cells (hPSCs) prove exemplary for the fruitful application of developmental principles to stem cell differentiation.

OVERVIEW OF PANCREAS DEVELOPMENT The invention of genetic tools, in particular transgenic mice, has been instrumental in defining the molecular events leading to pancreas organogenesis. Genetic lineage-tracing experiments revealed 19.2

Sander

· · Shih

Wang

Changes may still occur before final publication online and in print

CB29CH19-Sander

ARI

24 July 2013

12:44

Stomach Vein

Liver

Pancreatic ducts

Artery Endocrine cells

Gall bladder

Spleen

Islet of Lagerhans

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

Common bile duct

Pancreas

Duodenum Acinar cells

Figure 1 Overview of adult pancreas anatomy. The pancreas lies behind the stomach in the abdomen and attaches to the duodenum. For illustration purposes, the stomach is outlined with a dashed line and the duodenum is shown as a cut view. Exocrine acinar cells secrete pancreatic juice containing enzymes for the breakdown of proteins, carbohydrates, and lipids. The pancreatic juice is transported through a ductal network that releases the juice into the duodenum. The endocrine pancreas consists of small cell clusters, called islets of Langerhans, containing five endocrine cell types, namely glucagon-producing α-cells, insulin-producing β-cells, somatostatin-producing δ-cells, and pancreatic polypeptide-producing PP cells. Note, the figure shows the islet organization characteristic for mice with centrally located β-cells. Pancreatic islets are surrounded by a dense capillary network through which the hormones are released into the blood stream. See inset for an enlarged cutaway view of the pancreas.

a common origin of pancreatic exocrine and endocrine cells from a pool of progenitor cells in the gut endoderm (Gu et al. 2002). In mice, pancreas development first becomes morphologically evident around embryonic day (e) 8.75 with the emergence of epithelial buds on opposing sides of the foregut endoderm (Figure 2a). The two pancreatic buds subsequently elongate alongside the presumptive duodenum and stomach and, as a result of gut rotation, eventually fuse into a single organ by e12.5 (Figure 2b). Simultaneously, cell proliferation leads to a rapid size increase of the pancreatic buds, which also change their shape and begin to form branched tubular structures in a process known as branching morphogenesis. During this early phase of pancreatic development, which has been referred to as the primary transition (Pictet et al. 1972), the still-undifferentiated pancreatic epithelium is tightly surrounded by mesenchymal cells. During the secondary transition from e12.5 until birth, the pancreatic epithelium continues to expand and branch into a complex yet highly ordered tubular network (Figure 2c). These morphologic changes are paralleled by the differentiation of endocrine, acinar, and ductal cells. By the end of the secondary transition, the mouse pancreas has largely acquired the topological organization of the mature organ, with clusters of acinar cells coalesced around the ends of the ductal network and endocrine cell aggregates scattered throughout the organ (Figure 2c,d ). www.annualreviews.org • Pancreas Organogenesis

Changes may still occur before final publication online and in print

19.3

CB29CH19-Sander

a

ARI

24 July 2013

12:44

b

e9.5–e11.5

e12.5

Basement membrane

Dorsal pancreas Mesenchyme Microlumen Basement membrane Blood vessel

Tip cells

Central lumen

Trunk cells Rosette

Ventral pancreas

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

Blood vessel

c

Acinar cells

e15.5

Basement membrane

Blood vessels

Acinar cells

Progenitor cord Delaminated Ngn3+ cell

Endocrine cluster Endocrine cluster

Duct

d

Mature pancreas

Blood vessel

Secretory unit

Duct Islet Acinar cells

Artery

Islet of Langerhans Vein Duct

Endocrine cells

19.4

Sander

· · Shih

Wang

Changes may still occur before final publication online and in print

CB29CH19-Sander

ARI

24 July 2013

12:44

Over the past two decades, significant progress has been made in deciphering key molecular mechanisms that underlie the multiple lineage decisions leading to the formation of the pancreatic cell types. In comparison, far less is known is about the regulation of pancreas morphogenesis and whether a connection exists between morphogenetic events and cell differentiation. This review covers current knowledge of these subjects and discusses how these discoveries have begun to enable the development of cell-replacement strategies for diabetes.

LINEAGE DECISIONS DURING PANCREAS DEVELOPMENT

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

Pancreas Induction Prior to outgrowth of the pancreatic buds, a ventral and dorsal prepancreatic region is specified in the gut endoderm around e8.5 (Slack 1995). Although ventral and dorsal pancreatic buds arise independently in distinct locations, the molecular programs governing their specification are remarkably similar. In contrast to most epithelial cells of the early gut endoderm, the expression of sonic hedgehog (Shh) is selectively excluded from the presumptive pancreatic endoderm (Hebrok et al. 1998, Kim & Melton 1998). Transgenic misexpression experiments in mice revealed that the exclusion of Shh from the prepancreatic region is necessary for the induction of pancreatic markers (Apelqvist et al. 1997). Like Shh, retinoic acid (RA) also participates in patterning the gut endoderm. Absence of RA leads to anteriorization of the endoderm, resulting in complete pancreas agenesis in zebrafish and lack of the dorsal pancreas in Xenopus and mice (Chen et al. 2004, Mart´ın et al. 2005, Molotkov et al. 2005, Stafford & Prince 2002). RA seems to induce the dorsal pancreatic program by a Shh-independent mechanism, because loss of RA is not associated with ectopic Shh expression in the prepancreatic region in mice (Mart´ın et al. 2005). The ventral foregut region gives rise to both ventral pancreas and liver, which develop in close proximity to each other (for review, see Zorn & Wells 2009). Fibroblast growth factor (FGF) and bone morphogenetic protein (BMP) signaling are critical prohepatic cues, which are secreted by the cardiac mesoderm surrounding the prehepatic region (Deutsch et al. 2001, Rossi et al. 2001). The discovery of signals necessary for normal pancreas and liver induction has found practical applications in directed differentiation protocols of hPSCs, which use RA and inhibitors of Shh and BMP to induce pancreatic identity in vitro (D’Amour et al. 2006, Nostro et al. 2011). Dorsal bud emergence appears to also be regulated by extrinsic signals from endothelial cells. The dorsal and ventral pancreatic buds develop from regions of foregut endoderm that are in direct contact with the endothelium of the fusing dorsal aortae and vitelline veins, respectively ←−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−− Figure 2 Illustrated overview of pancreatic organogenesis. (a) The dorsal and ventral pancreatic epithelium evaginates into the surrounding mesenchyme between embryonic day (e) 9.5 and e11.5 in mice. At this stage, the pancreatic epithelium is comprised of a multilayered core of unpolarized cells engulfed by a basement membrane. Scattered microlumens (light yellow) arise between epithelial cells. Blood vessels surround but have not yet penetrated the epithelial buds. (b) At e12.5, the outer tip cell layer ( green) of the pancreatic epithelium forms recognizable branch protrusions. In the trunk portion of the pancreatic epithelium, microlumens fuse to form a primitive plexus, and groups of newly polarized cells ( purple) organize into rosettes around a lumen. At the same time, blood vessels begin to intercalate into the epithelium and contact trunk cells. (c) At e15.5, the luminal plexus progressively remodels into a single-layered epithelium consisting of highly branched primitive ducts (also known as progenitor cords) and newly differentiated acinar cells. Ngn3-expressing endocrine precursors (orange) delaminate and migrate away from the progenitor cords to form endocrine clusters. Blood vessels are intercalated between nascent branches of the pancreatic ductal tree. (d ) In the mature pancreas, acinar cells cap the endings of small terminal ducts and form functional exocrine secretory units. Endocrine cells are clustered in so-called islets of Langerhans, which are penetrated by a dense network of blood vessels. See insets for enlarged views of depicted areas. www.annualreviews.org • Pancreas Organogenesis

Changes may still occur before final publication online and in print

19.5

CB29CH19-Sander

ARI

24 July 2013

12:44

(Lammert et al. 2001). Tissue recombination experiments and studies of endothelium-deficient Flk1−/− mice have shown that the absence of endothelial cells prevents induction of early pancreatic genes (Lammert et al. 2001, Yoshitomi & Zaret 2004). However, contrary to these reports in mice, studies in zebrafish revealed pancreas specification in the absence of vascular endothelium (Field et al. 2003), a discrepancy that may reflect species differences.

a

Pdx1 Prox1

Primary transition

Oc1

Ptf1a Foxa1/2 Tcf2 Sox9 Gata4/6 Hes1

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

Pancreatic progenitors Secondary transition

b

Notch

Tip

Ptf1a c-Myc

Trunk

Ptf1a

Nkx6.1 Oc1 Nkx6.2 Tcf2

Nkx6.1

Cpa

Sox9

Hes1

Notch

c

Hes1

Acinus

Endocrine Ngn3 precursors

Sox9

Duct

Ngn3 PTF1-L

Oc1 Tcf2

Nr5a2 α

β

Glucagon

Insulin

Hes1 Sox9

Glis3 Prox1

d

Arx

19.6

Sander

· · Shih

Pax4

Nkx6.1

Pdx1

Wang

Changes may still occur before final publication online and in print

CB29CH19-Sander

ARI

24 July 2013

12:44

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

Establishing and Maintaining Pancreatic Identity Once the ventral and dorsal prepancreatic domains are specified, several transcription factors become specifically expressed in the prospective pancreatic regions. Among the earliest transcription factors that mark this region are Pdx1, Ptf1a, and Sox9 (Ahlgren et al. 1996, Guz et al. 1995, Kawaguchi et al. 2002, Krapp et al. 1998, Seymour et al. 2007). Gata4/6, Foxa1/2, Tcf2, Onecut1/2, Hes1, Prox1, and Mnx1 are also expressed in the prepancreatic domain, but their expression expands more broadly throughout the foregut endoderm (Figure 3a) (comprehensively reviewed in Gittes 2009, Pan & Wright 2011, Seymour & Sander 2011). Mice lacking any one of these factors display varying degrees of pancreas hypoplasia or agenesis; however, they still show pancreatic budding, which indicates that absence of one single factor does not entirely prevent initiation of the pancreatic program (reviewed in Gittes 2009, Mastracci & Sussel 2012, Pan & Wright 2011, Seymour & Sander 2011). Although budding of the pancreas is still observed, evidence strongly indicates that these transcription factors are important for determining pancreatic fate. For example, Ptf1a deletion leads to a reallocation of subsets of pancreas-fated progenitors to other endodermal lineages. This was shown by in vivo lineagetracing experiments, which revealed a contribution of Ptf1a-deficient cells to adjacent duodenal and common bile duct endoderm (Burlison et al. 2008, Kawaguchi et al. 2002). Further evidence that these early pancreatic transcription factors possess pancreatic fate-determining activity comes from gain-of-function experiments in Xenopus, which have shown that constitutively active forms of either Pdx1 or Ptf1a can allocate liver progenitors to the pancreatic lineage (Horb et al. 2003, Jarikji et al. 2007). Also, when ectopically expressed, Pdx1 and Ptf1a in combination are sufficient to convert duodenal precursors to the pancreatic fate (Afelik et al. 2006). Thus, in a developmental context, Pdx1 and Ptf1a can confer pancreatic identity to nonpancreatic cells. The extent to which Pdx1 and Ptf1a are also capable of activating pancreatic genes in nonpancreatic adult cells remains to be investigated. A better understanding of context-dependent effects of these early pancreatic transcription factors on gene expression could lead to novel strategies for reprogramming fibroblasts directly into pancreatic cells, as shown recently for neurons and cardiomyocytes (Qian et al. 2012, Thier et al. 2012). In addition to their role in pancreas specification, all early pancreatic transcription factors are necessary for further development of pancreatic buds (for reviews, see Gittes 2009, Pan & Wright 2011, and Seymour & Sander 2011). Mechanistically, the profound effect on the development of the organ caused by loss of only one single transcription factor may result from these early factors functioning in a transcriptional network with extensive cross-regulation between individual factors. For example, the expression of both Tcf2 and Ptf1a depends on Onecut-1 (Haumaitre et al. 2005, Maestro et al. 2003); simultaneously, Onecut-1 expression depends on Tcf2 and Ptf1a ←−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−− Figure 3 Lineage decisions during pancreas development. (a) During the primary transition, the transcription factors Pdx1, Prox1, Onecut-1 (Oc1), Ptf1a, Foxa1/2, Tcf2, Sox9, Gata4/6, and Hes1 mediate expansion of pancreatic progenitor cells and maintain pancreatic identity. (b) At the onset of the secondary transition, pancreatic progenitors adopt either tip or trunk identity. Notch activity promotes the trunk while repressing tip identity. Cross-repression between Ptf1a and Nkx6.1 mediates tip/trunk separation. (c) Tip cells adopt an acinar phenotype, which is maintained by a positive-regulatory loop between PTF1-L and Nr5a2. Trunk cells are bipotential for the ductal and endocrine cell fate. The ductal versus endocrine fate decision is controlled by graded Notch activity. High Notch promotes the ductal fate by activating both Hes1, a repressor of Ngn3, and Sox9, an Ngn3 activator. At low Notch activity, only Sox9 is activated, leading to endocrine differentiation. (d ) Endocrine precursors further differentiate into five different cell types, of which only α- and β-cells are depicted. Cross-repression between the α-cell determinant Arx and β-cell determinants Pax4, Nkx6.1, and Pdx1 separates the two lineages. www.annualreviews.org • Pancreas Organogenesis

Changes may still occur before final publication online and in print

19.7

CB29CH19-Sander

ARI

24 July 2013

12:44

(Poll et al. 2006, Thompson et al. 2012). Similar cross-regulation has been demonstrated between Sox9, Pdx1, Hes1, and Ptf1a (Ahnfelt-Rønne et al. 2012; Hale et al. 2005; Seymour et al. 2007, 2012; Thompson et al. 2012). Thus, elimination of a single factor has repercussions that ultimately affect multiple components of the pancreatic program. The advent of genome-scale tools and the availability of stem cell–based in vitro models of pancreas development (D’Amour et al. 2006, Kroon et al. 2008) now enable experiments to globally define the relevant transcriptional networks. Such knowledge should help inform reprogramming strategies for deriving pancreatic cells from other somatic cells.

Signaling During Early Pancreatic Growth

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

Whereas we have a relatively detailed understanding of the transcription factors regulating early pancreatic development, less is known about the signals controlling their expression. Both Hes1 and Sox9 are regulated by Notch signaling (Kageyama et al. 2007, Shih et al. 2012), which suggests that Notch signaling is important for early pancreas growth. As evidence to support this, embryos deficient for the Notch effector RBP-jκ have been shown to exhibit early arrest in pancreatic growth, similar in degree to that seen after Hes1 or Sox9 deletion (Apelqvist et al. 1999, Fujikura et al. 2006, Jensen et al. 2000, Seymour et al. 2007). Hes1 deletion is also associated with upregulation of the cell cycle inhibitor p57 in pancreatic progenitors (Georgia et al. 2006). Thus, there appears to be a direct link between Notch activity, transcription factor expression, and cell cycle regulation in the early pancreas. Known and putative roles for Notch in the developing pancreas have been expertly reviewed (Afelik & Jensen 2012). In addition to Notch signaling within the epithelium, pancreatic growth is also controlled by extrinsic signals from the surrounding mesenchyme. Seminal studies in the 1960s demonstrated the importance of the mesenchyme for early pancreatic growth (Golosow & Grobstein 1962, Rutter et al. 1964, Wessells & Cohen 1966). However, we have begun only recently to understand how mesenchymal signals control growth of the early pancreas. The best-understood pathway that relays pro-proliferative signals from the mesenchyme to the epithelium is the FGF signaling pathway. During the primary transition, FGF10 is highly expressed in pancreatic mesenchyme, whereas its receptor FGFR2 is expressed throughout the epithelium (Bhushan et al. 2001, Dichmann et al. 2003, Seymour et al. 2012). Gain- and loss-of-function experiments in mice further demonstrated that FGF10 stimulates progenitor cell proliferation and is required for early growth of the pancreatic buds (Bhushan et al. 2001). FGF10 has been shown to regulate Ptf1a and Sox9 expression in the epithelium ( Jacquemin et al. 2006, Seymour et al. 2012), which explains why FGF10 deletion results in a phenotype similar to that observed in either Ptf1a- or Sox9-deficient pancreas (Bhushan et al. 2001, Kawaguchi et al. 2002, Seymour et al. 2007). During the early growth phase of the pancreatic buds, Sox9, FGFR2, and FGF10 form a feed-forward loop, with mesenchymal FGF10 maintaining epithelial Sox9 expression and Sox9 upholding receptivity to FGF10 by cell-autonomously controlling FGFR2 expression (Seymour et al. 2012). If this loop is disrupted at any point, pancreatic epithelial cells can no longer maintain their identity, and liver genes are activated instead (Seymour et al. 2012). This demonstrates genetic coupling of early pancreatic growth and maintenance of organ identity. Additional signaling pathways critical to epithelial growth include the EGF, Wnt, and BMP pathways (Ahnfelt-Rønne et al. 2010, Miettinen et al. 2000, Murtaugh et al. 2005). Wnt and BMP signals appear not to be relayed to the epithelium; instead, they are transduced by mesenchymal cells (Ahnfelt-Rønne et al. 2010, Jonckheere et al. 2008, Landsman et al. 2011). It is unknown whether the ligand is secreted by the epithelium or the mesenchyme itself. Thus, signaling between the mesenchyme and epithelium likely involves multiple signaling loops. Recent in vitro studies 19.8

Sander

· · Shih

Wang

Changes may still occur before final publication online and in print

CB29CH19-Sander

ARI

24 July 2013

12:44

by Sneddon et al. (2012) on pancreatic lineage intermediates derived from hPSCs supported this contention, demonstrating that coculture with mesenchyme effectively expands progenitors and that this effect is specific both to the mesenchymal cell line used and to the progenitor being amplified. They further showed that no single growth factor or combination of factors is sufficient to reproduce the magnitude of effect of coculture with mesenchyme, reinforcing the notion that multiple factors cooperate to stimulate epithelial expansion. The identification of factors orchestrating pancreatic growth should be a high priority to aid the development of scalable protocols for the in vitro production of pancreatic cells.

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

Compartmentalizing the Organ: Patterning of Tips and Trunk Shortly after the primary transition, the nascent pancreatic buds have not yet acquired the macroscopic structure of the mature organ, nor have they formed the differentiated cell types necessary for pancreatic function. Instead, they are almost entirely composed of multipotent progenitor cells (MPCs). The transition of noncommitted MPCs into an organ with different cell compartments is a multistep process, of which the first step is the segregation of a tip and trunk domain at the onset of the secondary transition (Figure 3b) (Zhou et al. 2007). During this time, the developing organ continues to grow rapidly. Concomitantly, the pancreatic epithelium undergoes dynamic structural changes, resulting in multiple protrusions that bud from the edges. These structures, marked by expression of Ptf1a, c-Myc, and Cpa, represent the tip domain; conversely, the inner cells representing the trunk domain are identified by Nkx6.1/6.2, Sox9, Tcf2, Onecut-1, Prox1, and Hes1 (Figure 3b) ( Jacquemin et al. 2003, Klinck et al. 2011, Kopinke et al. 2011, Kopp et al. 2011, Schaffer et al. 2010, Solar et al. 2009, Vanhorenbeeck et al. 2007, Wang et al. 2005, Zhou et al. 2007). These morphological changes mark the beginning of a complex sequence of cell rearrangements (Villasenor et al. 2010), resulting in the mature organ structure (discussed in detail in the section on Pancreas Morphogenesis). In addition to the changes in organ shape, the formerly multipotent MPCs have taken the first step toward lineage commitment. Lineage-tracing experiments revealed that the trunk predominantly gives rise to the endocrine and ductal cell lineages, whereas the tips are quickly restricted to an acinar fate (Figure 3b) (Kopinke et al. 2011, Kopp et al. 2011, Pan et al. 2013, Solar et al. 2009, Zhou et al. 2007). The transcription factors Nkx6.1/6.2 and Ptf1a act as master regulators during this process. These factors are initially coexpressed in MPCs, but their expression domains entirely segregate during tip and trunk compartmentalization: Nkx6.1/6.2 promote trunk identity and segregate to trunk cells, and Ptf1a has an equivalent role in the tip compartment (Schaffer et al. 2010). Mechanistically, this process is initiated by transcriptional cross-repression between Nkx6.1/6.2 and Ptf1a (Schaffer et al. 2010). The example of Ptf1a, which first specifies pancreatic fate and later governs acinar cell development (Kawaguchi et al. 2002, Krapp et al. 1998, Masui et al. 2010), illustrates that developmental transcription factors can regulate different processes depending on cellular context. Genetic studies have provided insight into the signaling pathways regulating tip/trunk segregation, revealing that Notch signaling plays an essential role in promoting trunk identity. Repression of Notch signaling causes excessive tip formation at the expense of the trunk population (Afelik et al. 2012, Horn et al. 2012, Magenheim et al. 2011b). Conversely, constitutively active Notch prevents tip formation and promotes expansion of the trunk domain partly by activating the trunk marker Nkx6.1 (Afelik et al. 2012, Esni et al. 2004, Murtaugh et al. 2003, Schaffer et al. 2010). Although Notch signaling clearly plays a pivotal role in coordinating tip and trunk segregation, it remains unknown how Notch is regulated during this process. www.annualreviews.org • Pancreas Organogenesis

Changes may still occur before final publication online and in print

19.9

CB29CH19-Sander

ARI

24 July 2013

12:44

Development of the Acinar Lineage

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

In addition to their role in digestion, pancreatic acinar cells are an attractive target for β-cellreplacement therapy, because they are plentiful and retain the cellular plasticity to undergo reprogramming into β-cells (Zhou et al. 2008). Moreover, because they are the initiating cell type for precursor lesions of pancreatic cancer (Kopp et al. 2012), acinar cells are also a study focus for oncologists. As discussed, acinar cells predominantly arise from precursor cells in the tip domain (Pan et al. 2013, Zhou et al. 2007). By e15.5, most tip cells have undergone acinar cell differentiation, and subsequent expansion of the acinar domain is largely driven by cell proliferation (Pan et al. 2013, Zhou et al. 2007). Acinar cell differentiation is synergistically orchestrated by the transcription factors Ptf1a, Rbp-jl, and Nr5a2/LRH-1, which exhibit first tip cell–specific and later acinar cell–specific expression (Holmstrom et al. 2011; Krapp et al. 1996, 1998; Masui et al. 2007, 2010; Rose et al. 2001; Zhou et al. 2007). Prior to acinar cell differentiation, Ptf1a interacts with the b-HLH transcription factor Rbp-jκ (Beres et al. 2006, Masui et al. 2007, Miyatsuka et al. 2007). Together, these factors are responsible for the activation of Rbp-jl, a constitutively active, acinar-restricted paralog of Rbp-jκ (Masui et al. 2007). During the secondary transition, Rbp-jl then replaces Rbpjκ in the complex with Ptf1a. This complex, termed PTF1-L, establishes the acinar phenotype by direct activation of essential acinar genes (i.e., genes encoding digestive enzymes and proteins for exocytosis of zymogen granules), as well as Ptf1a and Rbp-jl themselves (Masui et al. 2007, 2008, 2010; Rose et al. 2001). Thus, PTF1-L induces and maintains the acinar phenotype through an autoactivation loop that sustains high expression of acinar genes (Figure 3c). Similarly, Nr5a2, a direct target gene of Ptf1a, is necessary for acinar cell differentiation (Holmstrom et al. 2011, Thompson et al. 2012). Nr5a2 binds to and regulates a set of genes similar to those regulated by the PTF1-L complex, and Nr5a2 and PTF1-L cooperate in the regulation of acinar cell–specific genes (Holmstrom et al. 2011). Together, these studies demonstrate that Ptf1a promotes acinar cell differentiation by initiating and stabilizing a gene-regulatory network that drives acinar gene expression. Although Ptf1a is an indispensable coordinator of acinar cell development, forced expression of Ptf1a alone is not sufficient to effectively induce acinar cell differentiation (Schaffer et al. 2010). Initiation of acinar differentiation may require other transcription factors functioning in parallel with Ptf1a. One candidate is Mist1, which is strongly expressed in acinar cells but does not depend on the PTF1-L complex for its expression (Masui et al. 2010, Yoshida et al. 2001). Mist1-deficient mice exhibit loss of differentiated characteristics of acinar cells, including loss of cellular polarity and defective exocytosis (Pin et al. 2001). Thus, the PTF1-L complex, Nr5a2, and Mist1 are all required to maintain the acinar phenotype. Genome-wide analysis of Mist target genes may reveal whether Mist1 occupies genomic regions similar to those of PTF1-L and Nr5a2 and could therefore regulate acinar development jointly with these factors. Less understood than acinar cell specification and differentiation are the mechanisms underlying the rapid proliferative expansion of acinar cells until birth. Unlike Mist1 and Ptf1a, which negatively regulate acinar cell expansion ( Jia et al. 2008, Rodolosse et al. 2004), Nr5a2 promotes proliferation of exocrine cell lines (Benod et al. 2011, Botrugno et al. 2004), revealing Nr5a2 as a potential candidate for driving acinar cell expansion. Additional candidates include c-Myc and β-catenin, whose inactivation in early pancreatic progenitors causes acinar cell hypoplasia (Bonal et al. 2009, Murtaugh et al. 2005, Nakhai et al. 2008, Wells et al. 2007). Temporally and spatially controlled inactivation of other acinar factors in mice may yield additional insight into molecular control of acinar cell differentiation, expansion, and maintenance.

19.10

Sander

· · Shih

Wang

Changes may still occur before final publication online and in print

CB29CH19-Sander

ARI

24 July 2013

12:44

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

The Endocrine Versus Ductal Cell-Fate Choice Whereas tip cells are fated to become acinar cells, the trunk domain is bipotential and produces ductal and endocrine cells (Figure 3c) (Kopinke et al. 2011, Kopp et al. 2011, Solar et al. 2009). After the tip and trunk domains have separated at around e12.5, cells in the trunk undergo extensive morphogenetic changes to form a 3-D network of tubules lined by a single layer of polarized epithelial cells (Hick et al. 2009, Kesavan et al. 2009, Villasenor et al. 2010). This network of tubules is referred to as the primitive ducts or progenitor cords and is the epithelium that gives rise to all endocrine cells (Hick et al. 2009, Kopp et al. 2011). During the secondary transition, a subset of cells in the progenitor cords initiate expression of the transcription factor Neurogenin3 (Ngn3), which marks the onset of endocrine cell differentiation (Gradwohl et al. 2000, Gu et al. 2002, Johansson et al. 2007, Schwitzgebel et al. 2000). Trunk epithelial cells that do not activate Ngn3 eventually contribute to the ductal tree (Beucher et al. 2012, Magenheim et al. 2011b, Wang et al. 2010). Thus, precise control of Ngn3 induction is the key factor balancing the endocrine versus ductal cell-fate decision. Many of the same transcription factors (e.g., Sox9, Tcf2, Onecut-1, Hes1, Prox1, and Glis3) that segregate to the trunk domain during tip/trunk separation later become restricted to the ducts and excluded from the endocrine compartment (Figure 3c) (Delous et al. 2012, Kang et al. 2009, Maestro et al. 2003, Pierreux et al. 2006, Shih et al. 2012, Wang et al. 2005, Westmoreland et al. 2012, Zhang et al. 2009). Consistent with their duct-specific expression, their inactivation in mice causes defects in ductal cell morphology and abnormal architecture of the ductal tree. For example, pancreata deficient for Sox9, Glis3, or Onecut-1 exhibit ductal cysts and lack ductal primary cilia, a functionally important organelle of ductal epithelial cells (Kang et al. 2009, Pierreux et al. 2006, Shih et al. 2012). Whether these ductal malformations reflect a functional requirement of these factors in the maintenance of ductal epithelial properties or a role in ductal cell differentiation is unknown. Conditional inactivation of the genes encoding ductal factors in developing and mature ducts should further define their role in ductal development and function. This knowledge could be relevant to understanding the pathogenesis of duct-related illnesses, including pancreatitis and pancreatic ductal adenoma carcinoma. Unlike ductal cell development, endocrine differentiation has been studied intensely, driven by hopes that such knowledge could help produce replacement β-cells for diabetes therapy. Because activation of Ngn3 expression is essential for initiating endocrine cell differentiation, there have been substantial efforts to understand Ngn3 regulation. Interestingly, many transcription factors controlling ductal development, such as Sox9, Tcf2, Glis3, and Onecut-1, are also necessary for Ngn3 induction ( Jacquemin et al. 2000, Kang et al. 2009, Kim et al. 2012, Lynn et al. 2007, Maestro et al. 2003, Seymour et al. 2008, Zhang et al. 2009), which indicates dual roles for these factors in the development of both lineages. However, Ngn3 is expressed in only a subset of cells within the primitive ducts, which raises the question of how its expression is repressed in the majority of embryonic ductal cells. Biochemical and genetic evidence suggests that the transcription factor Hes1 plays an important role in repressing Ngn3 transcription and preventing widespread Ngn3 activation (Ahnfelt-Rønne et al. 2012, Apelqvist et al. 1999, Jensen et al. 2000, Lee et al. 2001). Recently, Hes1 was found to accelerate Ngn3 protein degradation (Qu et al. 2013), which suggests that additional posttranscriptional mechanisms contribute to Ngn3 regulation. Supporting this idea, Villasenor et al. (2008) reported that Ngn3 mRNA is detected in a much broader domain than is the protein. Generally, the detailed mechanisms controlling Ngn3 expression are still poorly understood. Progress has been slow owing to the lack of a cell system in which Ngn3 regulation can be studied ex vivo. Because Ngn3 activation follows a pattern during pancreatic differentiation of hPSCs similar to that seen in vivo

www.annualreviews.org • Pancreas Organogenesis

Changes may still occur before final publication online and in print

19.11

ARI

24 July 2013

12:44

(D’Amour et al. 2006, Xie et al. 2013), hPSCs could prove an informative model for further studies. Because ducts simultaneously express positive and negative regulators of Ngn3, their relative levels will determine whether or not a progenitor activates Ngn3 and adopts endocrine identity. Evidence suggests that the Notch pathway plays an important role in this decision. Constitutive activation of Notch signaling prevents Ngn3 activation and promotes ductal cell differentiation (Greenwood et al. 2007, Murtaugh et al. 2003). This suggests that bipotential ductal/endocrine progenitors must escape Notch signaling to initiate endocrine differentiation. Surprisingly, Notch has been found to induce both negative and positive regulators of Ngn3. High Notch signaling promotes the expression of both Hes1 and Sox9, resulting in Ngn3 repression; conversely, lower Notch activity promotes the expression of Sox9 alone, allowing for Ngn3 activation (Figure 3c) (Shih et al. 2012). Thus, the select activation of Ngn3 in small subsets of progenitors appears to be controlled by graded Notch activity in the progenitor epithelium. How individual cells within the progenitor cords acquire different levels of Notch activity remains unknown. After the secondary transition, endocrine precursors gradually stop arising from the ductal epithelium, which suggests either that postnatal ducts lose competency to form endocrine precursors or that proendocrine signals are missing (Kopinke et al. 2011, Kopp et al. 2011, Solar et al. 2009). Studies of pancreatic injury models and clonal analysis of ductal cells in vitro indicate that adult ducts retain the capacity to activate Ngn3 and thus are competent to initiate, at least to some extent, endocrine programs ( Jin et al. 2013, Kopp et al. 2011). Adult ductal cell plasticity is a field of intense study that has been reviewed comprehensively (Desgraz et al. 2011, Reichert & Rustgi 2011).

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

CB29CH19-Sander

Differentiation of the Islet Cell Types Once Ngn3 is expressed, progenitors exit the cell cycle and delaminate from the progenitor cords into the surrounding stromal tissue (Gouzi et al. 2011, Miyatsuka et al. 2011). Although individual endocrine precursors are unipotent (Desgraz & Herrera 2009), Ngn3+ cells as a whole generate five different endocrine cell types: glucagon-producing α-cells, insulin-producing βcells, somatostatin-producing δ-cells, pancreatic polypeptide–producing PP-cells, and ghrelinproducing ε-cells. Johansson et al. (2007) have shown that the competence of endocrine precursors to produce different endocrine cell types is temporally controlled. When Ngn3 expression was induced at different developmental time points from a conditional Ngn3 transgene, progenitors first differentiated into α-cells, then β- and δ-cells, and finally PP-cells. The mechanisms controlling this change in developmental potential are unknown. Studies in the developing neurons, muscle, and pituitary suggest that Notch signaling could be involved (Mizutani & Saito 2005, Mourikis et al. 2012, Zhu et al. 2006). Here, the duration of exposure to Notch is critical for determining progenitor cell competence. If the pancreas uses a similar mechanism, optimizing Notch exposure time could be key to maximizing β-cell yield in directed differentiation protocols for hPSCs. When differentiating into hormone-producing cells, Ngn3+ endocrine precursors undergo dynamic changes in gene expression, resulting in activation of both Ngn3-dependent transcription factors (Pax4, Arx, Rfx6, NeuroD1, Pax6, Isl1, and IA2) and hormones. Analysis of mouse mutants revealed that these factors control many aspects of endocrine development, including cell differentiation, cell maintenance, and islet cell–type identity (comprehensively reviewed in Mastracci & Sussel 2012, Pan & Wright 2011). Here we focus on the mechanisms governing islet cell–type identity, specifically on the cell fate choice between the β- and α-cell lineage. As with the mechanisms governing tip/trunk segregation, fate distinction between β- and αcell precursors is thought to rely on mutual repression between opposing lineage determinants. 19.12

Sander

· · Shih

Wang

Changes may still occur before final publication online and in print

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

CB29CH19-Sander

ARI

24 July 2013

12:44

The transcription factors Pax4, Pdx1, and Nkx6.1 act as critical β-cell determinants (Collombat et al. 2003, Gannon et al. 2008, Henseleit et al. 2005, Holland et al. 2002, Sosa-Pineda et al. 1997), whereas Arx determines α-cell identity (Collombat et al. 2003, 2005). Forced expression of Pax4, Pdx1, or Nkx6.1 in endocrine precursors favors the β-cell fate at the expense of α-cells (Collombat et al. 2009, Schaffer et al. 2013, Yang et al. 2011). Conversely, Arx gain of function results in excess α-cells and reduced β-cells (Collombat et al. 2007). Loss-of-function experiments revealed reciprocal phenotypes, although the consequences of Pdx1 deletion in endocrine precursors are yet to be explored. Both Pax4 and Nkx6.1 are direct transcriptional repressors of Arx; conversely, Arx represses Pax4, Nkx6.1, and Pdx1 by direct or indirect mechanisms (Figure 3d ) (Collombat et al. 2005, 2007; Schaffer et al. 2013; Yang et al. 2011). One interesting but mechanistically poorly understood observation is that some transcription factors, which can change the lineage identity of endocrine precursors, lose this ability when misexpressed in differentiated endocrine cells. For example, Pdx1 or Nkx6.1 is insufficient to convert differentiated α-cells into β-cells (Schaffer et al. 2013, Yang et al. 2011). This suggests that endocrine differentiation restrains developmental plasticity. The molecular mechanisms restricting cell plasticity during cell differentiation are poorly understood. Recent studies indicate a role for DNA methylation (Dhawan et al. 2011, Papizan et al. 2011), but additional studies are needed to understand how such epigenetic mechanisms limit cell plasticity. A better understanding of these mechanisms could enable strategies for reversing epigenetic constraints in efforts to reprogram non-β endocrine cells into β-cells.

PANCREAS MORPHOGENESIS Coincident with the differentiation of endocrine and exocrine cells, the pancreas develops into a highly branched organ, wherein groups of acinar cells coalesce around a small ductal lumen to form functional exocrine secretory units, and ductal cells organize into the tubular network of the pancreatic ductal system (Pictet et al. 1972). This process, called branching morphogenesis (Figure 2), is driven by dynamic cell rearrangements and spatiotemporally controlled cell proliferation. Although pancreatic cell differentiation and its molecular control have been studied since the 1990s, knowledge of how the pancreas acquires its final shape and becomes a functional organ is still rudimentary. Two landmark studies describing the cellular changes associated with pancreatic tubulogenesis (Kesavan et al. 2009, Villasenor et al. 2010) now provide the framework for exploration of the mechanisms governing pancreatic branching morphogenesis. Prior to the initiation of epithelial branching at around e12, the pancreatic buds form a multilayered core of unpolarized cells engulfed by a single basement membrane abutting the mesenchyme (Figure 2a) (Hick et al. 2009, Kesavan et al. 2009, Villasenor et al. 2010). Pancreatic tubulogenesis begins with individual cells acquiring apicobasal polarity: Cells develop a defined apical membrane that faces a central lumen and a basal surface attached to a layer of extracellular matrix (ECM) (Kesavan et al. 2009, Villasenor et al. 2010). Groups of newly polarized cells then form rosettes, wherein adjacent cells are linked by intercellular junctional complexes around a nascent central lumen (Figure 2b, inset) (Villasenor et al. 2010). Newly formed microlumens are initially unconnected but eventually fuse into a luminal plexus (Figure 2c). Remodeling of the luminal plexus produces a tubular network lined by a polarized, monolayered epithelium attached to a basement membrane (Figure 2c, inset) (Kesavan et al. 2009, Villasenor et al. 2010). At birth, the tubes have organized into a highly branched ductal tree, connecting exocrine secretory units to the ductal system (Figure 2d, inset). Concomitant to epithelial branching, blood vessels and nerves also begin to penetrate the epithelium, intercalating between nascent branches of the pancreatic ductal tree (Figure 2b–d ) (Lindsay et al. 2006, www.annualreviews.org • Pancreas Organogenesis

Changes may still occur before final publication online and in print

19.13

CB29CH19-Sander

ARI

24 July 2013

12:44

Magenheim et al. 2011a, Pierreux et al. 2010). The stereotypical organization of cells in the mature pancreas illustrates that proper pancreas morphogenesis requires highly coordinated cell rearrangements involving acinar, ductal, endocrine, endothelial, neuronal, and mesenchymal cells. Currently, little is known about the molecular cues governing these cell rearrangements; about how pancreas morphogenesis is coordinated with cell differentiation; or about how differentiation of pancreatic endocrine and exocrine cells influences surrounding nonepithelial cells, and vice versa.

Intracellular Signals Governing Pancreas Morphogenesis

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

In many organs, including pancreas, epithelial morphogenesis is controlled by Rho family GTPases, which regulate cell polarity, adhesion, and migration by organizing the actin cytoskeleton (Heasman & Ridley 2008). Kesavan et al. (2009) demonstrated that deletion of the Rho GTPase Cdc42 interrupts cell polarization during early morphogenesis, disrupting epithelial morphology and aborting tubulogenesis at microlumen formation. Premature arrest of tubulogenesis was also observed in mice deficient in RhoA GTPase inhibitor Strad13 (Petzold et al. 2013), whereas pancreas-specific deletion of Rho GTPase Rac1 had no obvious effect on pancreas morphogenesis (Heid et al. 2011). This implies that different Rho GTPases have distinct roles in shaping the organ. Studies in other organs show that Rho GTPases are regulated by Ephrin, EGF, and ECMintegrin signaling (Heasman & Ridley 2008, Huveneers & Danen 2009, Poliakov et al. 2004). The hypobranched pancreas seen in Ephrin and EGF receptor–mutant mice suggests that these signaling pathways could also influence Rho GTPase activity in the pancreas (Miettinen et al. 2000, Villasenor et al. 2010). How extrinsic cues coordinate epithelial morphogenesis and differentiation in a spatiotemporally controlled manner is an active area of research.

Coordinating Morphogenesis and Cell Differentiation Evidence is emerging that mesenchymal cells, blood vessels, and ECM provide locally restricted signals that influence morphogenesis and cell fate decisions in the developing pancreas. Tip/trunk segregation not only marks the first cell fate choice of pancreatic progenitors but also coincides with the formation of branches in the tip region (Figure 2b, inset) (Zhou et al. 2007). The tissue remodeling associated with branch formation results in the repositioning of mesenchymal and epithelial cells and basement membrane components (Kesavan et al. 2009, Landsman et al. 2011). Real-time imaging and static analysis of pancreas morphogenesis suggest that new branches form by tip-splitting (i.e., individual tips repeatedly split into new tips) (Puri & Hebrok 2007, Villasenor et al. 2010). Whether this is driven directly by differential proliferation of tip cells or indirectly by cell remodeling in the trunk is unknown. However, tip cells have been observed to proliferate more quickly than trunk cells (Petzold et al. 2013, Zhou et al. 2007), which implies that tip proliferation could drive branch formation. One candidate cue for regulating tip-cell proliferation is FGF, which is secreted by mesenchymal cells that surround the tips when branching is initiated (Bhushan et al. 2001). Removal of mesenchyme as an FGF source halts morphogenesis of pancreatic explants, whereas adding FGF to cultures restores branching and cell proliferation (Miralles et al. 1999). Similar to their association with mesenchymal cells, emerging tip cells are also directly apposed to the basement membrane at the beginning of epithelial branching (Kesavan et al. 2009, Villasenor et al. 2010). Explant culture studies have shown that knockdown or blocking of antibodies against the ECM component laminin-1 reduces expression of early acinar cell markers and impairs branching (Kesavan et al. 2009, Li et al. 2004). Because the branching process leads to increased cellular exposure to ECM, the process of morphogenesis as such could cause cells to adopt acinar identity. 19.14

Sander

· · Shih

Wang

Changes may still occur before final publication online and in print

CB29CH19-Sander

ARI

24 July 2013

12:44

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

Whereas mesenchymal cells and ECM components favor acinar identity and promote branching, blood vessels have the opposite effect. Forced VEGFA expression in pancreatic progenitors caused increased expression of the trunk marker Hes1 and concomitant loss of the tip/acinar markers Cpa and amylase, whereas VEGF ablation favored acinar formation and caused hyperbranching (Magenheim et al. 2011a, Pierreux et al. 2010). Consistent with their trunk-promoting effect, blood vessels remain physically associated with the trunk and become separated from the tips (Magenheim et al. 2011a, Pierreux et al. 2010) (Figure 2b–c, inset). These examples illustrate that organ morphogenesis and cell differentiation are intricately linked. Further studies should focus on identifying the extrinsic and intrinsic cues mediating cross talk between endothelial, mesenchymal, and epithelial cells and on deciphering how these cues govern morphogenesis and cell differentiation. To analyze spatiotemporally dynamic intercellular interactions more precisely, it would be helpful to develop a system for studying pancreas organogenesis in vitro under defined conditions. A stem cell–based 3-D differentiation system could enable significant progress.

Morphogenetic Changes and Endocrine Cell Differentiation Morphogenetic changes are also associated with the initiation of endocrine differentiation in the progenitor cords. As cells initiate Ngn3 expression, they acquire a droplet shape and delaminate from the progenitor epithelium (Figure 2c, inset) (Gouzi et al. 2011, Rukstalis & Habener 2007). Gouzi et al. (2011) have shown that expression of Snail2, a protein that induces cell motility by triggering epithelial-to-mesenchymal transition (EMT) (Barrallo-Gimeno & Nieto 2005), is Ngn3 dependent. Ectopic Ngn3 expression also triggers EMT, which suggests a dual role for Ngn3 in inducing both cell differentiation and delamination (Gouzi et al. 2011). The extent to which these processes are interdependent is unknown. Conditional inactivation of Snail2, or of other effectors of EMT, may explain the connection between endocrine cell differentiation and delamination. Another mechanism that could enable cell delamination is asymmetric cell division, in which cells divide so that only one daughter cell inherits the apical membrane and remains in the progenitor epithelium, while the daughter cell inheriting the basal membrane escapes from the epithelial sheath and differentiates. This mechanism is associated with the asymmetric distribution of cell fate determinants to the daughter cells and is relevant for the differentiation of neurons and epidermal cells (reviewed in Knoblich 2008). It is unknown whether endocrine precursors are created by asymmetric cell divisions in progenitor cords. The knowledge may be important for devising appropriate in vitro culture conditions for production of functional endocrine cells from hPSCs. Rapidly improving real-time imaging technology, which now allows for the visualization of fluorescently labeled cells in whole organ explants at single-cell resolution, will likely prove useful for defining the cellular events associated with the emergence of endocrine precursors from the progenitor cords.

APPLICATION OF DEVELOPMENTAL PRINCIPLES TOWARD STEM CELL DIFFERENTIATION The ultimate goal of understanding pancreas development is to gain insight into pancreasassociated diseases and to develop novel therapies to treat such diseases. With the knowledge amassed by studying pancreas development, investigators are exploring novel therapeutic avenues for treating diabetes by replacing or regenerating β-cell mass. Such innovative strategies for diabetes treatment include increasing β-cell numbers by boosting their intrinsic ability to replicate; fate-converting other pancreatic cells into β-cells (reviewed in Desgraz et al. 2011); www.annualreviews.org • Pancreas Organogenesis

Changes may still occur before final publication online and in print

19.15

CB29CH19-Sander

ARI

24 July 2013

12:44

and most fascinatingly, both as a therapy and as a tool for study, generating β-cells in vitro by differentiating hPSCs.

Generating Pancreatic Progenitors from Human Pluripotent Stem Cells

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

The enthusiasm for deriving transplantable β-cells from hPSCs is the direct result of landmark achievements in cell transplantation, in particular the successful allogenic transplantation of islets, which has allowed diabetic patients to forego insulin injections for years (Shapiro et al. 2006). Unfortunately, the amount of donor islets required for widespread treatment significantly exceeds availability, and even when donor islets are available, recipients must undergo lifelong immunosuppression to prevent rejection. Although these limitations have prevented its widespread use in diabetic patients, the insulin independence achieved with islet transplantation provides proof of the principle that cell transplantation can be a viable therapy for diabetes. Unlike allografts, which are susceptible to donor shortages, hPSCs could yield an unlimited source of transplantable β-cells. Additionally, patient-derived induced pluripotent stem cells would obviate immunosuppression. For these reasons, there is currently a major push to generate pancreatic β-cells in vitro. Protocols have been established for deriving pancreatic precursors from hPSCs. hPSCs can be differentiated with high efficiency to definitive endoderm by adding high levels of Activin A, mimicking the proendodermal role of Nodal during in vivo gastrulation (reviewed by Zorn & Wells 2009). A landmark study by D’Amour et al. (2006) established a protocol to guide hPSC-derived endoderm toward the pancreatic lineage. Similar protocols have since been published (Kroon et al. 2008, Mfopou et al. 2010, Nostro et al. 2011, Rezania et al. 2012, Xu et al. 2011). The majority of pancreatic differentiation protocols induce pancreatic fate by simultaneously treating cells with RA and inhibitors of Shh and BMP, recapitulating the signaling environment that specifies the pancreas in vivo. Such treatment generates the in vitro equivalent of early multipotent pancreatic progenitors, expressing PDX1, SOX9, PTF1a, and NKX6.1. These in vitro–generated progenitors also pass the functional test of having the potential to form mature endocrine, ductal, and acinar cells when transplanted into mice (Kelly et al. 2011, Kroon et al. 2008, Rezania et al. 2012, Schulz et al. 2012, Xie et al. 2013). The resulting endocrine cells are functional, responding appropriately to glucose and properly regulating glucose homeostasis in diabetic animal models (Kroon et al. 2008, Rezania et al. 2012, Xie et al. 2013). Further evidence for their islet-like phenotype is that the transcriptional signature and cellular composition of hPSC-derived endocrine clusters are strikingly similar to those of human islets (Xie et al. 2013). Having accomplished the in vitro differentiation of functional pancreatic progenitors, the field has attempted to optimize differentiation efficiencies across different cell lines. In addition, to allow for large-scale cell production, strategies have been developed to expand developmental lineage intermediates in vitro (Cheng et al. 2012, Schulz et al. 2012, Sneddon et al. 2012). However, because it is unknown whether transplantation of pancreatic progenitors can cure diabetes in humans, the ultimate goal is to generate functional β-cells in vitro.

Beyond Progenitors: How to Generate Functional Endocrine Cells In Vitro Currently, endocrine cells that arise in vitro are non–glucose responsive and often coexpress multiple hormones. For pancreatic differentiation of hPSCs to fulfill its full therapeutic and scientific promise, methods to generate functional pancreatic cells in vitro must be developed. As discussed in the sections on Lineage Decisions During Pancreas Development and Pancreas Morphogenesis, above, the developing pancreas receives extrinsic signals from the mesenchyme, ECM, and endothelium. However, these components are not supplied in the majority of hPSC 19.16

Sander

· · Shih

Wang

Changes may still occur before final publication online and in print

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

CB29CH19-Sander

ARI

24 July 2013

12:44

differentiation protocols. Coculture with mesenchymal cell lines has been explored as a strategy to expand intermediate precursor populations, consistent with the role of mesenchyme in early pancreatic progenitor expansion (Sneddon et al. 2012). Possible instructive signals from the mesenchyme have been added to current pancreatic differentiation protocols and have resulted in increased efficiency of PDX1 induction (Guo et al. 2013). The role of mesenchyme and ECM at later steps of pancreatic development in vivo, especially during endocrine cell differentiation, is still poorly understood. Both should be considered as a possible source for signals when developing strategies to generate functional β-cells in vitro. Blood vessels may also be involved in islet development, because both native islets and in vivo– matured, transplanted hPSC-derived endocrine cells are highly vascularized with endocrine cells in direct contact with blood vessels. Although endothelial cells are clearly important for proper sensing of blood glucose levels and release of hormones, their role in pancreatic lineage determination is only beginning to be unveiled. Because blood vessels promote trunk cell formation at the expense of tip cell formation (see section on Coordinating Morphogenesis and Cell Differentiation) (Magenheim et al. 2011a, Pierreux et al. 2010), endothelial cells could provide critical missing cues for favoring the endocrine cell fate during in vitro differentiation. As discussed in the section on Pancreas Morphogenesis, the embryonic pancreas undergoes extensive morphogenetic changes in a 3-D epithelium, which may be coupled with cell-lineage decisions. Unlike in vivo development, in vitro pancreatic differentiation of hPSCs is primarily performed in 2-D culture systems. This difference between in vitro and in vivo differentiation conditions could, at least in part, explain our inability to generate functional β-cells in vitro. Protocols to generate functional cells of other endodermal lineages (e.g., the intestine) have employed 3-D culture systems. Such 3-D systems support proper morphogenesis and have been shown to produce mini organs with remarkable similarity to their in vivo counterparts (Sato et al. 2009, Spence et al. 2011). Organotypic culture systems may also facilitate the production of functional β-cells from hPSCs. An important question is whether the generation of islets or of only β-cells should be attempted. Engrafted hPSC-derived progenitors differentiate into functional islet-like clusters that contain all endocrine cell types, in a ratio similar to that in human islets (Xie et al. 2013). Although it is currently unknown whether non-β islet cells are required for β-cell function, evidence suggests that paracrine signaling between endocrine cell types may be important in humans (reviewed in Caicedo 2012). Therefore, future studies should consider the production of entire islets and not just of β-cells.

Human Pluripotent Stem Cells as a Model for Understanding Pancreas Biology and Disease Much information about pancreas development and organogenesis has been gleaned from animal models; there are, however, some limitations to these models. At a technical level, transient developmental cell populations are too sparse in embryos to isolate sufficient cells for large-scale genomic or biochemical analyses. The ability to differentiate such lineage intermediates from hPSC in virtually unlimited quantities has enabled such studies and recently has led to the identification of core transcriptional and epigenetic mechanisms of pancreatic lineage determination (Xie et al. 2013). Moreover, species-specific differences in gene regulation limit the use of animal models for studying human disease. One example is maturity onset diabetes of the young (MODY), which in humans results from single-gene mutations that are not always phenocopied in mice (Haumaitre et al. 2006, Mayer et al. 2008). It is hoped that protocols for the in vitro production of functional β-cells will facilitate studies aimed at understanding disease mechanisms of diabetes. Advances www.annualreviews.org • Pancreas Organogenesis

Changes may still occur before final publication online and in print

19.17

CB29CH19-Sander

ARI

24 July 2013

12:44

in genome editing now allow us to model effects of gene mutations or single-nucleotide polymorphisms in hPSCs. Thus, the production of β-cells in vitro not only will be a significant step toward regenerative medicine but also will help advance our understanding of the pathogenesis of diabetes.

FUTURE PROSPECTS

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

This review has covered the tremendous progress we have made in understanding pancreatic organogenesis and has briefly discussed the progress made in recapitulating this process in vitro using hPSCs. Reflecting on this body of work, a few conclusions become strikingly clear. First, much remains to be understood about the complex process of pancreas organogenesis, specifically, how morphogenesis is coupled with cell fate specification. Although we have made significant progress in understanding many of the transcription factors that are required at various stages of pancreas development, the signals that promote and regulate each developmental decision are still poorly understood. Clearly, the current success of in vitro pancreatic differentiation of hPSCs has relied heavily on studies employing animal models. Therefore, we must continue to apply the lessons learned in vivo to achieve the critical goal of generating functional β-cells in vitro.

DISCLOSURE STATEMENT The authors are not aware of any affiliations, memberships, funding, or financial holdings that might be perceived as affecting the objectivity of this review.

ACKNOWLEDGMENTS We thank members of the Sander laboratory for constructive comments on the manuscript. We apologize to our colleagues whose references were omitted owing to space constraints. Work in the Sander laboratory is supported by grants from the National Institutes of Health (NIH), the Juvenile Diabetes Research Foundation, and the California Institute for Regenerative Medicine. H.P.S. and A.W. were supported by postdoctoral fellowships from the Juvenile Diabetes Research Foundation. A.W. was additionally supported by the NIH training program in diabetes research T32 DK7494-27. LITERATURE CITED Afelik S, Chen Y, Pieler T. 2006. Combined ectopic expression of Pdx1 and Ptf1a/p48 results in the stable conversion of posterior endoderm into endocrine and exocrine pancreatic tissue. Genes Dev. 20:1441–46 Afelik S, Jensen J. 2012. Notch signaling in the pancreas: patterning and cell fate specification. WIREs Dev. Biol. 2:531–44 Afelik S, Qu X, Hasrouni E, Bukys MA, Deering T, et al. 2012. Notch-mediated patterning and cell fate allocation of pancreatic progenitor cells. Development 139:1744–53 Ahlgren U, Jonsson J, Edlund H. 1996. The morphogenesis of the pancreatic mesenchyme is uncoupled from that of the pancreatic epithelium in IPF1/PDX1-deficient mice. Development 122:1409–16 Ahnfelt-Rønne J, Jørgensen MC, Klinck R, Jensen JN, Fuchtbauer E-M, et al. 2012. Ptf1a-mediated control ¨ of Dll1 reveals an alternative to the lateral inhibition mechanism. Development 139:33–45 Ahnfelt-Rønne J, Ravassard P, Pardanaud-Glavieux C, Scharfmann R, Serup P. 2010. Mesenchymal bone morphogenetic protein signaling is required for normal pancreas development. Diabetes 59:1948–56 Apelqvist A, Ahlgren U, Edlund H. 1997. Sonic hedgehog directs specialised mesoderm differentiation in the intestine and pancreas. Curr. Biol. 7:801–4 19.18

Sander

· · Shih

Wang

Changes may still occur before final publication online and in print

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

CB29CH19-Sander

ARI

24 July 2013

12:44

Apelqvist A, Li H, Sommer L, Beatus P, Anderson DJ, et al. 1999. Notch signalling controls pancreatic cell differentiation. Nature 400:877–81 Barrallo-Gimeno A, Nieto MA. 2005. The Snail genes as inducers of cell movement and survival: implications in development and cancer. Development 132:3151–61 Benod C, Vinogradova MV, Jouravel N, Kim GE, Fletterick RJ, Sablin EP. 2011. Nuclear receptor liver receptor homologue 1 (LRH-1) regulates pancreatic cancer cell growth and proliferation. Proc. Natl. Acad. Sci. USA 108:16927–31 Beres TM, Masui T, Swift GH, Shi L, Henke RM, MacDonald RJ. 2006. PTF1 is an organ-specific and Notch-independent basic helix-loop-helix complex containing the mammalian Suppressor of Hairless (RBP-J) or its paralogue, RBP-L. Mol. Cell. Biol. 26:117–30 Beucher A, Mart´ın M, Spenle C, Poulet M, Collin C, Gradwohl G. 2012. Competence of failed endocrine progenitors to give rise to acinar but not ductal cells is restricted to early pancreas development. Dev. Biol. 361:277–85 Bhushan A, Itoh N, Kato S, Thiery JP, Czernichow P, et al. 2001. Fgf10 is essential for maintaining the proliferative capacity of epithelial progenitor cells during early pancreatic organogenesis. Development 128:5109–17 Bonal C, Thorel F, Ait-Lounis A, Reith W, Trumpp A, Herrera PL. 2009. Pancreatic inactivation of cMyc decreases acinar mass and transdifferentiates acinar cells into adipocytes in mice. Gastroenterology 136:309–19.e9 Botrugno OA, Fayard E, Annicotte J-S, Haby C, Brennan T, et al. 2004. Synergy between LRH-1 and β-catenin induces G1 cyclin-mediated cell proliferation. Mol. Cell 15:499–509 Burlison JS, Long Q, Fujitani Y, Wright CV, Magnuson MA. 2008. Pdx-1 and Ptf1a concurrently determine fate specification of pancreatic multipotent progenitor cells. Dev. Biol. 316:74–86 Caicedo A. 2012. Paracrine and autocrine interactions in the human islet: more than meets the eye. Semin. Cell Dev. Biol. 24:11–21 Chen Y, Pan FC, Brandes N, Afelik S, Solter M, Pieler T. 2004. Retinoic acid signaling is essential for pancreas ¨ development and promotes endocrine at the expense of exocrine cell differentiation in Xenopus. Dev. Biol. 271:144–60 Cheng X, Ying L, Lu L, Galvao AM, Mills JA, et al. 2012. Self-renewing endodermal progenitor lines generated from human pluripotent stem cells. Cell Stem Cell 10:371–84 Collombat P, Hecksher-Sørensen J, Broccoli V, Krull J, Ponte I, et al. 2005. The simultaneous loss of Arx and Pax4 genes promotes a somatostatin-producing cell fate specification at the expense of the α- and β-cell lineages in the mouse endocrine pancreas. Development 132:2969–80 Collombat P, Hecksher-Sørensen J, Krull J, Berger J, Riedel D, et al. 2007. Embryonic endocrine pancreas and mature β cells acquire α and PP cell phenotypes upon Arx misexpression. J. Clin. Investig. 117:961–70 Collombat P, Mansouri A, Hecksher-Sørensen J, Serup P, Krull J, et al. 2003. Opposing actions of Arx and Pax4 in endocrine pancreas development. Genes Dev. 17:2591–603 Collombat P, Xu X, Ravassard P, Sosa-Pineda B, Dussaud S, et al. 2009. The ectopic expression of Pax4 in the mouse pancreas converts progenitor cells into α and subsequently β cells. Cell 138:449–62 D’Amour KA, Bang AG, Eliazer S, Kelly OG, Agulnick AD, et al. 2006. Production of pancreatic hormoneexpressing endocrine cells from human embryonic stem cells. Nat. Biotechnol. 24:1392–401 Delous M, Yin C, Shin D, Ninov N, Debrito Carten J, et al. 2012. sox9b is a key regulator of pancreaticobiliary ductal system development. PLoS Genet. 8:e1002754 Desgraz R, Bonal C, Herrera PL. 2011. β-Cell regeneration: the pancreatic intrinsic faculty. Trends Endocrinol. Metab. 22:34–43 Desgraz R, Herrera PL. 2009. Pancreatic neurogenin 3-expressing cells are unipotent islet precursors. Development 136:3567–74 Deutsch G, Jung J, Zheng M, Lora J, Zaret KS. 2001. A bipotential precursor population for pancreas and liver within the embryonic endoderm. Development 128:871–81 Dhawan S, Georgia S, Tschen SI, Fan G, Bhushan A. 2011. Pancreatic β cell identity is maintained by DNA methylation-mediated repression of Arx. Dev. Cell 20:419–29 Dichmann DS, Miller CP, Jensen J, Heller RS, Serup P. 2003. Expression and misexpression of members of the FGF and TGFβ families of growth factors in the developing mouse pancreas. Dev. Dyn. 226:663–74 www.annualreviews.org • Pancreas Organogenesis

Changes may still occur before final publication online and in print

19.19

ARI

24 July 2013

12:44

Esni F, Ghosh B, Biankin AV, Lin JW, Albert MA, et al. 2004. Notch inhibits Ptf1 function and acinar cell differentiation in developing mouse and zebrafish pancreas. Development 131:4213–24 Field HA, Dong PD, Beis D, Stainier DY. 2003. Formation of the digestive system in zebrafish. II. Pancreas morphogenesis. Dev. Biol. 261:197–208 Fujikura J, Hosoda K, Iwakura H, Tomita T, Noguchi M, et al. 2006. Notch/Rbp-j signaling prevents premature endocrine and ductal cell differentiation in the pancreas. Cell Metab. 3:59–65 Gannon M, Ables ET, Crawford L, Lowe D, Offield MF, et al. 2008. pdx-1 function is specifically required in embryonic β cells to generate appropriate numbers of endocrine cell types and maintain glucose homeostasis. Dev. Biol. 314:406–17 Georgia S, Soliz R, Li M, Zhang P, Bhushan A. 2006. p57 and Hes1 coordinate cell cycle exit with self-renewal of pancreatic progenitors. Dev. Biol. 298:22–31 Gittes GK. 2009. Developmental biology of the pancreas: a comprehensive review. Dev. Biol. 326:4–35 Golosow N, Grobstein C. 1962. Epitheliomesenchymal interaction in pancreatic morphogenesis. Dev. Biol. 4:242–55 Gouzi M, Kim YH, Katsumoto K, Johansson K, Grapin-Botton A. 2011. Neurogenin3 initiates stepwise delamination of differentiating endocrine cells during pancreas development. Dev. Dyn. 240:589–604 Gradwohl G, Dierich A, LeMeur M, Guillemot F. 2000. neurogenin3 is required for the development of the four endocrine cell lineages of the pancreas. Proc. Natl. Acad. Sci. USA 97:1607–11 Greenwood AL, Li S, Jones K, Melton DA. 2007. Notch signaling reveals developmental plasticity of Pax4+ pancreatic endocrine progenitors and shunts them to a duct fate. Mech. Dev. 124:97–107 Gu G, Dubauskaite J, Melton DA. 2002. Direct evidence for the pancreatic lineage: NGN3+ cells are islet progenitors and are distinct from duct progenitors. Development 129:2447–57 Guo T, Landsman L, Li N, Hebrok M. 2013. Factors expressed by murine embryonic pancreatic mesenchyme enhance generation of insulin-producing cells from hESCs. Diabetes 5:1581–92 Guz Y, Montminy MR, Stein R, Leonard J, Gamer LW, et al. 1995. Expression of murine STF-1, a putative insulin gene transcription factor, in β cells of pancreas, duodenal epithelium and pancreatic exocrine and endocrine progenitors during ontogeny. Development 121:11–18 Hale MA, Kagami H, Shi L, Holland AM, Elsasser HP, et al. 2005. The homeodomain protein PDX1 is required at mid-pancreatic development for the formation of the exocrine pancreas. Dev. Biol. 286:225– 37 Haumaitre C, Barbacci E, Jenny M, Ott MO, Gradwohl G, Cereghini S. 2005. Lack of TCF2/vHNF1 in mice leads to pancreas agenesis. Proc. Natl. Acad. Sci. USA 102:1490–95 Haumaitre C, Fabre M, Cormier S, Baumann C, Delezoide A-L, Cereghini S. 2006. Severe pancreas hypoplasia and multicystic renal dysplasia in two human fetuses carrying novel HNF1β/MODY5 mutations. Hum. Mol. Genet. 15:2363–75 Heasman SJ, Ridley AJ. 2008. Mammalian Rho GTPases: new insights into their functions from in vivo studies. Nat. Rev. Mol. Cell Biol. 9:690–701 Hebrok M, Kim SK, Melton DA. 1998. Notochord repression of endodermal Sonic hedgehog permits pancreas development. Genes Dev. 12:1705–13 Heid I, Lubeseder-Martellato C, Sipos B, Mazur PK, Lesina M, et al. 2011. Early requirement of Rac1 in a mouse model of pancreatic cancer. Gastroenterology 141:719–30.e7 Henseleit KD, Nelson SB, Kuhlbrodt K, Hennings JC, Ericson J, Sander M. 2005. NKX6 transcription factor activity is required for α- and β-cell development in the pancreas. Development 132:3139–49 Hick A-C, van Eyll JM, Cordi S, Forez C, Passante L, et al. 2009. Mechanism of primitive duct formation in the pancreas and submandibular glands: a role for SDF-1. BMC Dev. Biol. 9:66 Holland AM, Hale MA, Kagami H, Hammer RE, MacDonald RJ. 2002. Experimental control of pancreatic development and maintenance. Proc. Natl. Acad. Sci. USA 99:12236–41 Holmstrom SR, Deering T, Swift GH, Poelwijk FJ, Mangelsdorf DJ, et al. 2011. LRH-1 and PTF1-L coregulate an exocrine pancreas-specific transcriptional network for digestive function. Genes Dev. 25:1674–79 Horb ME, Shen CN, Tosh D, Slack JM. 2003. Experimental conversion of liver to pancreas. Curr. Biol. 13:105–15 Horn S, Kobberup S, Jørgensen MC, Kalisz M, Klein T, et al. 2012. Mind bomb 1 is required for pancreatic β-cell formation. Proc. Natl. Acad. Sci. USA 109:7356–61

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

CB29CH19-Sander

19.20

Sander

· · Shih

Wang

Changes may still occur before final publication online and in print

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

CB29CH19-Sander

ARI

24 July 2013

12:44

Huveneers S, Danen EHJ. 2009. Adhesion signaling—crosstalk between integrins, Src and Rho. J. Cell Sci. 122:1059–69 Jacquemin P, Durviaux SM, Jensen J, Godfraind C, Gradwohl G, et al. 2000. Transcription factor hepatocyte nuclear factor 6 regulates pancreatic endocrine cell differentiation and controls expression of the proendocrine gene ngn3. Mol. Cell. Biol. 20:4445–54 Jacquemin P, Lemaigre FP, Rousseau GG. 2003. The Onecut transcription factor HNF-6 (OC-1) is required for timely specification of the pancreas and acts upstream of Pdx-1 in the specification cascade. Dev. Biol. 258:105–16 Jacquemin P, Yoshitomi H, Kashima Y, Rousseau GG, Lemaigre FP, Zaret KS. 2006. An endothelialmesenchymal relay pathway regulates early phases of pancreas development. Dev. Biol. 290:189–99 Jarikji ZH, Vanamala S, Beck CW, Wright CV, Leach SD, Horb ME. 2007. Differential ability of Ptf1a and Ptf1a-VP16 to convert stomach, duodenum and liver to pancreas. Dev. Biol. 304:786–99 Jensen J, Pedersen EE, Galante P, Hald J, Heller RS, et al. 2000. Control of endodermal endocrine development by Hes-1. Nat. Genet. 24:36–44 Jia D, Sun Y, Konieczny SF. 2008. Mist1 regulates pancreatic acinar cell proliferation through p21CIP1/WAF1 . Gastroenterology 135:1687–97 Jin L, Feng T, Shih HP, Zerda R, Luo A, et al. 2013. Colony-forming cells in the adult mouse pancreas are expandable in Matrigel and form endocrine/acinar colonies in laminin hydrogel. Proc. Natl. Acad. Sci. USA 110:3907–12 Johansson KA, Dursun U, Jordan N, Gu G, Beermann F, et al. 2007. Temporal control of neurogenin3 activity in pancreas progenitors reveals competence windows for the generation of different endocrine cell types. Dev. Cell 12:457–65 Jonckheere N, Mayes E, Shih H-P, Li B, Lioubinski O, et al. 2008. Analysis of mPygo2 mutant mice suggests a requirement for mesenchymal Wnt signaling in pancreatic growth and differentiation. Dev. Biol. 318:224– 35 Kageyama R, Ohtsuka T, Kobayashi T. 2007. The Hes gene family: repressors and oscillators that orchestrate embryogenesis. Development 134:1243–51 Kang HS, Kim Y-S, ZeRuth G, Beak JY, Gerrish K, et al. 2009. Transcription factor Glis3, a novel critical player in the regulation of pancreatic β-cell development and insulin gene expression. Mol. Cell. Biol. 29:6366–79 Kawaguchi Y, Cooper B, Gannon M, Ray M, MacDonald RJ, Wright CV. 2002. The role of the transcriptional regulator Ptf1a in converting intestinal to pancreatic progenitors. Nat. Genet. 32:128–34 Kelly OG, Chan MY, Martinson LA, Kadoya K, Ostertag TM, et al. 2011. Cell-surface markers for the isolation of pancreatic cell types derived from human embryonic stem cells. Nat. Biotechnol. 29:750–56 Kesavan G, Sand FW, Greiner TU, Johansson JK, Kobberup S, et al. 2009. Cdc42-mediated tubulogenesis controls cell specification. Cell 139:791–801 Kim SK, Melton DA. 1998. Pancreas development is promoted by cyclopamine, a hedgehog signaling inhibitor. Proc. Natl. Acad. Sci. USA 95:13036–41 Kim Y-S, Kang HS, Takeda Y, Hom L, Song H-Y, et al. 2012. Glis3 regulates neurogenin 3 expression in pancreatic β-cells and interacts with its activator, Hnf6. Mol. Cells 34:193–200 Klinck R, Fuchtbauer E-M, Ahnfelt-Rønne J, Serup P, Jensen JN, Jørgensen MC. 2011. A BAC transgenic ¨ Hes1-EGFP reporter reveals novel expression domains in mouse embryos. Gene Expr. Patterns 11:415–26 Knoblich JA. 2008. Mechanisms of asymmetric stem cell division. Cell 132:583–97 Kopinke D, Brailsford M, Shea JE, Leavitt R, Scaife CL, Murtaugh LC. 2011. Lineage tracing reveals the dynamic contribution of Hes1+ cells to the developing and adult pancreas. Development 138:431–41 Kopp JL, Dubois CL, Schaffer AE, Hao E, Shih HP, et al. 2011. Sox9+ ductal cells are multipotent progenitors throughout development but do not produce new endocrine cells in the normal or injured adult pancreas. Development 138:653–65 Kopp JL, von Figura G, Mayes E, Liu F-F, Dubois CL, et al. 2012. Identification of Sox9-dependent acinarto-ductal reprogramming as the principal mechanism for initiation of pancreatic ductal adenocarcinoma. Cancer Cell 22:737–50 www.annualreviews.org • Pancreas Organogenesis

Changes may still occur before final publication online and in print

19.21

ARI

24 July 2013

12:44

Krapp A, Knofler M, Frutiger S, Hughes GJ, Hagenbuchle O, Wellauer PK. 1996. The p48 DNA-binding ¨ ¨ subunit of transcription factor PTF1 is a new exocrine pancreas-specific basic helix-loop-helix protein. EMBO J. 15:4317–29 Krapp A, Knofler M, Ledermann B, Burki K, Berney C, et al. 1998. The bHLH protein PTF1-p48 is essential ¨ ¨ for the formation of the exocrine and the correct spatial organization of the endocrine pancreas. Genes Dev. 12:3752–63 Kroon E, Martinson LA, Kadoya K, Bang AG, Kelly OG, et al. 2008. Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat. Biotechnol. 26:443–52 Lammert E, Cleaver O, Melton D. 2001. Induction of pancreatic differentiation by signals from blood vessels. Science 294:564–67 Landsman L, Nijagal A, Whitchurch TJ, Vanderlaan RL, Zimmer WE, et al. 2011. Pancreatic mesenchyme regulates epithelial organogenesis throughout development. PLoS Biol. 9:e1001143 Lee JC, Smith SB, Watada H, Lin J, Scheel D, et al. 2001. Regulation of the pancreatic pro-endocrine gene Neurogenin3. Diabetes 50:928–36 Li Z, Manna P, Kobayashi H, Spilde T, Bhatia A, et al. 2004. Multifaceted pancreatic mesenchymal control of epithelial lineage selection. Dev. Biol. 269:252–63 Lindsay TH, Halvorson KG, Peters CM, Ghilardi JR, Kuskowski MA, et al. 2006. A quantitative analysis of the sensory and sympathetic innervation of the mouse pancreas. Neuroscience 137:1417–26 Lynn FC, Smith SB, Wilson ME, Yang KY, Nekrep N, German MS. 2007. Sox9 coordinates a transcriptional network in pancreatic progenitor cells. Proc. Natl. Acad. Sci. USA 104:10500–5 Maestro MA, Boj SF, Luco RF, Pierreux CE, Cabedo J, et al. 2003. Hnf6 and Tcf2 (MODY5) are linked in a gene network operating in a precursor cell domain of the embryonic pancreas. Hum. Mol. Genet. 12:3307–14 Magenheim J, Ilovich O, Lazarus A, Klochendler A, Ziv O, et al. 2011a. Blood vessels restrain pancreas branching, differentiation and growth. Development 138:4743–52 Magenheim J, Klein AM, Stanger BZ, Ashery-Padan R, Sosa-Pineda B, et al. 2011b. Ngn3+ endocrine progenitor cells control the fate and morphogenesis of pancreatic ductal epithelium. Dev. Biol. 359:26–36 Mart´ın M, Gallego-Llamas J, Ribes V, Kedinger M, Niederreither K, et al. 2005. Dorsal pancreas agenesis in retinoic acid-deficient Raldh2 mutant mice. Dev. Biol. 284:399–411 Mastracci TL, Sussel L. 2012. The endocrine pancreas: insights into development, differentiation and diabetes. WIREs Dev. Biol. 1:609–28 Masui T, Long Q, Beres TM, Magnuson MA, MacDonald RJ. 2007. Early pancreatic development requires the vertebrate Suppressor of Hairless (RBPJ) in the PTF1 bHLH complex. Genes Dev. 21:2629–43 Masui T, Swift GH, Deering T, Shen C, Coats WS, et al. 2010. Replacement of Rbpj with Rbpjl in the PTF1 complex controls the final maturation of pancreatic acinar cells. Gastroenterology 139:270–80 Masui T, Swift GH, Hale MA, Meredith DM, Johnson JE, MacDonald RJ. 2008. Transcriptional autoregulation controls pancreatic Ptf1a expression during development and adulthood. Mol. Cell. Biol. 28:5458–68 Mayer C, Bottcher Y, Kovacs P, Halbritter J, Stumvoll M. 2008. Phenotype of a patient with a de novo ¨ mutation in the hepatocyte nuclear factor 1β/maturity-onset diabetes of the young type 5 gene. Metab. Clin. Exp. 57:416–20 Mfopou JK, Chen B, Mateizel I, Sermon K, Bouwens L. 2010. Noggin, retinoids, and fibroblast growth factor regulate hepatic or pancreatic fate of human embryonic stem cells. Gastroenterology 138:2233–45.e14 Miettinen PJ, Huotari M-A, Koivisto T, Ustinov J, Palgi J, et al. 2000. Impaired migration and delayed differentiation of pancreatic islet cells in mice lacking EGF-receptors. Development 127:2617–27 Miralles F, Czernichow P, Ozaki K, Itoh N, Scharfmann R. 1999. Signaling through fibroblast growth factor receptor 2b plays a key role in the development of the exocrine pancreas. Proc. Natl. Acad. Sci. USA 96:6267–72 Miyatsuka T, Kosaka Y, Kim H, German MS. 2011. Neurogenin3 inhibits proliferation in endocrine progenitors by inducing Cdkn1a. Proc. Natl. Acad. Sci. USA 108:185–90 Miyatsuka T, Matsuoka TA, Shiraiwa T, Yamamoto T, Kojima I, Kaneto H. 2007. Ptf1a and RBP-J cooperate in activating Pdx1 gene expression through binding to Area III. Biochem. Biophys. Res. Commun. 362:905–9

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

CB29CH19-Sander

19.22

Sander

· · Shih

Wang

Changes may still occur before final publication online and in print

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

CB29CH19-Sander

ARI

24 July 2013

12:44

Mizutani K, Saito T. 2005. Progenitors resume generating neurons after temporary inhibition of neurogenesis by Notch activation in the mammalian cerebral cortex. Development 132:1295–304 Molotkov A, Molotkova N, Duester G. 2005. Retinoic acid generated by Raldh2 in mesoderm is required for mouse dorsal endodermal pancreas development. Dev. Dyn. 232:950–57 Mourikis P, Gopalakrishnan S, Sambasivan R, Tajbakhsh S. 2012. Cell-autonomous Notch activity maintains the temporal specification potential of skeletal muscle stem cells. Development 139:4536–48 Murtaugh LC, Law AC, Dor Y, Melton DA. 2005. β-Catenin is essential for pancreatic acinar but not islet development. Development 132:4663–74 Murtaugh LC, Stanger BZ, Kwan KM, Melton DA. 2003. Notch signaling controls multiple steps of pancreatic differentiation. Proc. Natl. Acad. Sci. USA 100:14920–25 Nakhai H, Siveke JT, Mendoza-Torres L, Schmid RM. 2008. Conditional inactivation of Myc impairs development of the exocrine pancreas. Development 135:3191–96 Nostro MC, Sarangi F, Ogawa S, Holtzinger A, Corneo B, et al. 2011. Stage-specific signaling through TGFβ family members and WNT regulates patterning and pancreatic specification of human pluripotent stem cells. Development 138:861–71 Pan FC, Bankaitis ED, Boyer D, Xu X, van de Casteele M, et al. 2013. Spatiotemporal patterns of multipotentiality in Ptf1a-expressing cells during pancreas organogenesis and injury-induced facultative restoration. Development 140:751–64 Pan FC, Wright C. 2011. Pancreas organogenesis: from bud to plexus to gland. Dev. Dyn. 240:530–65 Papizan JB, Singer RA, Tschen SI, Dhawan S, Friel JM, et al. 2011. Nkx2.2 repressor complex regulates islet β-cell specification and prevents β-to-α-cell reprogramming. Genes Dev. 25:2291–305 Petzold KM, Naumann H, Spagnoli FM. 2013. Rho signalling restriction by the RhoGAP Stard13 integrates growth and morphogenesis in the pancreas. Development 140:126–35 Pictet RL, Clark WR, Williams RH, Rutter WJ. 1972. An ultrastructural analysis of the developing embryonic pancreas. Dev. Biol. 29:436–67 Pierreux CE, Cordi S, Hick AC, Achouri Y, Ruiz de Almodovar C, et al. 2010. Epithelial: endothelial cross-talk regulates exocrine differentiation in developing pancreas. Dev. Biol. 347:216–27 Pierreux CE, Poll AV, Kemp CR, Clotman F, Maestro MA, et al. 2006. The transcription factor hepatocyte nuclear factor-6 controls the development of pancreatic ducts in the mouse. Gastroenterology 130:532–41 Pin CL, Rukstalis JM, Johnson C, Konieczny SF. 2001. The bHLH transcription factor Mist1 is required to maintain exocrine pancreas cell organization and acinar cell identity. J. Cell Biol. 155:519–30 Poliakov A, Cotrina M, Wilkinson DG. 2004. Diverse roles of eph receptors and ephrins in the regulation of cell migration and tissue assembly. Dev. Cell 7:465–80 Poll AV, Pierreux CE, Lokmane L, Haumaitre C, Achouri Y, et al. 2006. A vHNF1/TCF2-HNF6 cascade regulates the transcription factor network that controls generation of pancreatic precursor cells. Diabetes 55:61–69 Puri S, Hebrok M. 2007. Dynamics of embryonic pancreas development using real-time imaging. Dev. Biol. 306:82–93 Qian L, Huang Y, Spencer CI, Foley A, Vedantham V, et al. 2012. In vivo reprogramming of murine cardiac fibroblasts into induced cardiomyocytes. Nature 485:593–98 Qu X, Afelik S, Jensen JN, Bukys MA, Kobberup S, et al. 2013. Notch-mediated post-translational control of Ngn3 protein stability regulates pancreatic patterning and cell fate commitment. Dev. Biol. 376:1–12 Reichert M, Rustgi AK. 2011. Pancreatic ductal cells in development, regeneration, and neoplasia. J. Clin. Investig. 121:4572–78 Rezania A, Bruin JE, Riedel MJ, Mojibian M, Asadi A, et al. 2012. Maturation of human embryonic stem cell-derived pancreatic progenitors into functional islets capable of treating pre-existing diabetes in mice. Diabetes 61:2016–29 Rodolosse A, Chalaux E, Adell T, Hag`ege H, Skoudy A, Real FX. 2004. PTF1α/p48 transcription factor couples proliferation and differentiation in the exocrine pancreas [corrected]. Gastroenterology 27:937–49 Rose SD, Swift GH, Peyton MJ, Hammer RE, MacDonald RJ. 2001. The role of PTF1-P48 in pancreatic acinar gene expression. J. Biol. Chem. 276:44018–26 www.annualreviews.org • Pancreas Organogenesis

Changes may still occur before final publication online and in print

19.23

ARI

24 July 2013

12:44

Rossi JM, Dunn NR, Hogan BL, Zaret KS. 2001. Distinct mesodermal signals, including BMPs from the septum transversum mesenchyme, are required in combination for hepatogenesis from the endoderm. Genes Dev. 15:1998–2009 Rukstalis JM, Habener JF. 2007. Snail2, a mediator of epithelial-mesenchymal transitions, expressed in progenitor cells of the developing endocrine pancreas. Gene Expr. Patterns: GEP 7:471–79 Rutter WJ, Wessells NK, Grobstein C. 1964. Control of specific synthesis in the developing pancreas. Natl. Cancer Inst. Monogr. 13:51–65 Sato T, Vries RG, Snippert HJ, van de Wetering M, Barker N, et al. 2009. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 459:262–65 Schaffer AE, Freude KK, Nelson SB, Sander M. 2010. Nkx6 transcription factors and Ptf1a function as antagonistic lineage determinants in multipotent pancreatic progenitors. Dev. Cell 18:1022–29 Schaffer AE, Taylor BL, Benthuysen JR, Liu J, Thorel F, et al. 2013. Nkx6.1 controls a gene regulatory network required for establishing and maintaining pancreatic beta cell identity. PLoS Genet. 9:e1003274 Schulz TC, Young HY, Agulnick AD, Babin MJ, Baetge EE, et al. 2012. A scalable system for production of functional pancreatic progenitors from human embryonic stem cells. PLoS ONE 7:e37004 Schwitzgebel VM, Scheel DW, Conners JR, Kalamaras J, Lee JE, et al. 2000. Expression of neurogenin3 reveals an islet cell precursor population in the pancreas. Development 127:3533–42 Seymour PA, Freude KK, Dubois CL, Shih H-P, Patel NA, Sander M. 2008. A dosage-dependent requirement for Sox9 in pancreatic endocrine cell formation. Dev. Biol. 323:19–30 Seymour PA, Freude KK, Tran MN, Mayes EE, Jensen J, et al. 2007. SOX9 is required for maintenance of the pancreatic progenitor cell pool. Proc. Natl. Acad. Sci. USA 104:1865–70 Seymour PA, Sander M. 2011. Historical perspective: beginnings of the β-cell. Current perspectives in β-cell development. Diabetes 60:364–76 Seymour PA, Shih HP, Patel NA, Freude KK, Xie R, et al. 2012. A Sox9/Fgf feed-forward loop maintains pancreatic organ identity. Development 139:3363–72 Shapiro AM, Ricordi C, Hering BJ, Auchincloss H, Lindblad R, et al. 2006. International trial of the Edmonton protocol for islet transplantation. New Engl. J. Med. 355:1318–30 Shih HP, Kopp JL, Sandhu M, Dubois CL, Seymour PA, et al. 2012. A Notch-dependent molecular circuitry initiates pancreatic endocrine and ductal cell differentiation. Development 139:2488–99 Slack JM. 1995. Developmental biology of the pancreas. Development 121:1569–80 Sneddon JB, Borowiak M, Melton DA. 2012. Self-renewal of embryonic-stem-cell-derived progenitors by organ-matched mesenchyme. Nature 491:765–68 Solar M, Cardalda C, Houbracken I, Mart´ın M, Maestro MA, et al. 2009. Pancreatic exocrine duct cells give rise to insulin-producing β cells during embryogenesis but not after birth. Dev. Cell 17:849–60 Sosa-Pineda B, Chowdhury K, Torres M, Oliver G, Gruss P. 1997. The Pax4 gene is essential for differentiation of insulin-producing β cells in the mammalian pancreas. Nature 386:399–402 Spence JR, Mayhew CN, Rankin SA, Kuhar MF, Vallance JE, et al. 2011. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature 470:105–9 Stafford D, Prince VE. 2002. Retinoic acid signaling is required for a critical early step in zebrafish pancreatic development. Curr. Biol. 12:1215–20 Thier M, Worsd P, Lakes YB, Gorris R, Herms S, et al. 2012. Direct conversion of fibroblasts into stably ¨ orfer ¨ expandable neural stem cells. Cell Stem Cell 10:473–79 Thompson N, G´esina E, Scheinert P, Bucher P, Grapin-Botton A. 2012. RNA profiling and chromatin immunoprecipitation-sequencing reveal that PTF1a stabilizes pancreas progenitor identity via the control of MNX1/HLXB9 and a network of other transcription factors. Mol. Cell. Biol. 32:1189–99 Vanhorenbeeck V, Jenny M, Cornut JF, Gradwohl G, Lemaigre FP, et al. 2007. Role of the Onecut transcription factors in pancreas morphogenesis and in pancreatic and enteric endocrine differentiation. Dev. Biol. 305:685–94 Villasenor A, Chong DC, Cleaver O. 2008. Biphasic Ngn3 expression in the developing pancreas. Dev. Dyn. 237:3270–79 Villasenor A, Chong DC, Henkemeyer M, Cleaver O. 2010. Epithelial dynamics of pancreatic branching morphogenesis. Development 137:4295–305

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

CB29CH19-Sander

19.24

Sander

· · Shih

Wang

Changes may still occur before final publication online and in print

Annu. Rev. Cell Dev. Biol. 2013.29. Downloaded from www.annualreviews.org by Georgetown University on 08/06/13. For personal use only.

CB29CH19-Sander

ARI

24 July 2013

12:44

Wang J, Kilic G, Aydin M, Burke Z, Oliver G, Sosa-Pineda B. 2005. Prox1 activity controls pancreas morphogenesis and participates in the production of “secondary transition” pancreatic endocrine cells. Dev. Biol. 286:182–94 Wang S, Yan J, Anderson DA, Xu Y, Kanal MC, et al. 2010. Neurog3 gene dosage regulates allocation of endocrine and exocrine cell fates in the developing mouse pancreas. Dev. Biol. 339:26–37 Wells JM, Esni F, Boivin GP, Aronow BJ, Stuart W, et al. 2007. Wnt/β-catenin signaling is required for development of the exocrine pancreas. BMC Dev. Biol. 7:4 Wessells NK, Cohen JH. 1966. The influence of collagen and embryo extract on the development of pancreatic epithelium. Exp. Cell Res. 43:680–84 Westmoreland JJ, Kilic G, Sartain C, Sirma S, Blain J, et al. 2012. Pancreas-specific deletion of Prox1 affects development and disrupts homeostasis of the exocrine pancreas. Gastroenterology 142:999–1009.e6 Xie R, Everett LJ, Lim HW, Patel NA, Schug J, et al. 2013. Dynamic chromatin remodeling mediated by polycomb proteins orchestrates pancreatic differentiation of human embryonic stem cells. Cell Stem Cell 12:224–37 Xu X, Browning VL, Odorico JS. 2011. Activin, BMP and FGF pathways cooperate to promote endoderm and pancreatic lineage cell differentiation from human embryonic stem cells. Mech. Dev. 128:412–27 Yang Y-P, Thorel F, Boyer DF, Herrera PL, Wright CV. 2011. Context-specific α-to-β-cell reprogramming by forced Pdx1 expression. Genes Dev. 25:1680–85 Yoshida S, Ohbo K, Takakura A, Takebayashi H, Okada T, et al. 2001. Sgn1, a basic helix-loop-helix transcription factor delineates the salivary gland duct cell lineage in mice. Dev. Biol. 240:517–30 Yoshitomi H, Zaret KS. 2004. Endothelial cell interactions initiate dorsal pancreas development by selectively inducing the transcription factor Ptf1a. Development 131:807–17 Zhang H, Ables ET, Pope CF, Washington MK, Hipkens S, et al. 2009. Multiple, temporal-specific roles for HNF6 in pancreatic endocrine and ductal differentiation. Mech. Dev. 126:958–73 Zhou Q, Brown J, Kanarek A, Rajagopal J, Melton DA. 2008. In vivo reprogramming of adult pancreatic exocrine cells to β-cells. Nature 455:627–32 Zhou Q, Law AC, Rajagopal J, Anderson WJ, Gray PA, Melton DA. 2007. A multipotent progenitor domain guides pancreatic organogenesis. Dev. Cell 13:103–14 Zhu X, Zhang J, Tollkuhn J, Ohsawa R, Bresnick EH, et al. 2006. Sustained Notch signaling in progenitors is required for sequential emergence of distinct cell lineages during organogenesis. Genes Dev. 20:2739–53 Zorn AM, Wells JM. 2009. Vertebrate endoderm development and organ formation. Annu. Rev. Cell Dev. Biol. 25:221–51

www.annualreviews.org • Pancreas Organogenesis

Changes may still occur before final publication online and in print

19.25

Pancreas organogenesis: from lineage determination to morphogenesis.

The pancreas is an essential organ for proper nutrient metabolism and has both endocrine and exocrine function. In the past two decades, knowledge of ...
6MB Sizes 0 Downloads 0 Views