Share this post on:

T endodermal cell derivatives from ESC can be generated through the in vitro recapitulation of major developmental signalling pathways occurring in vivo [1]. For instance, a conserved mechanism for mesoderm-endoderm lineage commitment involves Nodal, a TGFb family member, and can be mimicked in vitro by activin A, yielding a high percentage of endodermal-like cells [2,3,4]. From this cell CP21 chemical information population, different studies have used instructive signals playing a role in pancreatic organogenesis and b-cell differentiation to commit ESC to similar fates in vitro in order to 11967625 obtain a source of replaceable b-cells for diabeticpatients [5,6,7]. In addition to the endocrine compartment, the pancreas is composed by exocrine cells including ductal and acinar cells. Acinar cells are responsible for the synthesis of secretory digestive enzymes, and alterations in the acinar differentiation program have been linked to exocrine pancreatic diseases, such as chronic pancreatitis and adenocarcinoma [8]. Therefore, providing normal in vitro models of acinar differentiation from ESC could be helpful to understand better these processes as primary acinar cultures fail to retain a differentiated phenotype [9,10]. We previously demonstrated the generation of acinar cells from mESC on the basis of the genetic selection of elastase 1 (Ela1)-producing cells and the differentiation with conditioned medium from the culture of fetal pancreatic tissues [11]. As this medium contains signals that also promote the differentiation of other pancreatic cell lineages, the isolation of the acinar-like cells was required. In this sense, one important aspect missing in many pancreatic differentiation protocols is toPancreatic Acinar Differentiation of Mouse ESCassess the extent of selectivity in cell lineage induction. In this regard, other studies have reported the expression of acinar markers from ESC by manipulating several developmental pathways already established for endocrine differentiation or without examining their role on endocrine gene expression [12,13,14,15]. Therefore, progress in the knowledge of how acinar cells are formed during embryogenesis is 3397-23-7 essential for the improvement of strategies assessing ESC exocrine differentiation. Pancreatic organogenesis is a highly regulated process controlled by the gut microenvironment that orchestrates the expression of key transcription factors that, in turn, specify the different pancreatic cell types [16]. Both endocrine and exocrine cells derive from a common pool of progenitors present in the foregut endoderm. The cross-talk between several pathways including the inhibition of Shh and RA signalling activation specifies the pancreatic domain at early stages and regulates the emergence of Pdx1-expressing progenitors that can be expanded by FGF10 [17,18,19,20]. In addition, Ptf1a is a bHLH protein essential for pancreatic formation and in its absence pancreatic progenitors assume an intestinal fate [21,22]. Gradual reduction of Ptf1a dosage in mice leads to pancreatic hypoplasia and delayed exocrine cytodifferentiation [23]. In the adult, Ptf1a is only expressed in acinar cells as a component of PTF1, a heterotrimeric transcriptional complex including a ubiquitous E-protein and Recombination signal-binding protein J ike (Rbpjl) [24,25,26]. During early pancreatic development, Ptf1a requires the interaction with the Rbpj isoform for pancreatic growth and morphogenesis. Then at the onset of acinar cell development, Rbpj is r.T endodermal cell derivatives from ESC can be generated through the in vitro recapitulation of major developmental signalling pathways occurring in vivo [1]. For instance, a conserved mechanism for mesoderm-endoderm lineage commitment involves Nodal, a TGFb family member, and can be mimicked in vitro by activin A, yielding a high percentage of endodermal-like cells [2,3,4]. From this cell population, different studies have used instructive signals playing a role in pancreatic organogenesis and b-cell differentiation to commit ESC to similar fates in vitro in order to 11967625 obtain a source of replaceable b-cells for diabeticpatients [5,6,7]. In addition to the endocrine compartment, the pancreas is composed by exocrine cells including ductal and acinar cells. Acinar cells are responsible for the synthesis of secretory digestive enzymes, and alterations in the acinar differentiation program have been linked to exocrine pancreatic diseases, such as chronic pancreatitis and adenocarcinoma [8]. Therefore, providing normal in vitro models of acinar differentiation from ESC could be helpful to understand better these processes as primary acinar cultures fail to retain a differentiated phenotype [9,10]. We previously demonstrated the generation of acinar cells from mESC on the basis of the genetic selection of elastase 1 (Ela1)-producing cells and the differentiation with conditioned medium from the culture of fetal pancreatic tissues [11]. As this medium contains signals that also promote the differentiation of other pancreatic cell lineages, the isolation of the acinar-like cells was required. In this sense, one important aspect missing in many pancreatic differentiation protocols is toPancreatic Acinar Differentiation of Mouse ESCassess the extent of selectivity in cell lineage induction. In this regard, other studies have reported the expression of acinar markers from ESC by manipulating several developmental pathways already established for endocrine differentiation or without examining their role on endocrine gene expression [12,13,14,15]. Therefore, progress in the knowledge of how acinar cells are formed during embryogenesis is essential for the improvement of strategies assessing ESC exocrine differentiation. Pancreatic organogenesis is a highly regulated process controlled by the gut microenvironment that orchestrates the expression of key transcription factors that, in turn, specify the different pancreatic cell types [16]. Both endocrine and exocrine cells derive from a common pool of progenitors present in the foregut endoderm. The cross-talk between several pathways including the inhibition of Shh and RA signalling activation specifies the pancreatic domain at early stages and regulates the emergence of Pdx1-expressing progenitors that can be expanded by FGF10 [17,18,19,20]. In addition, Ptf1a is a bHLH protein essential for pancreatic formation and in its absence pancreatic progenitors assume an intestinal fate [21,22]. Gradual reduction of Ptf1a dosage in mice leads to pancreatic hypoplasia and delayed exocrine cytodifferentiation [23]. In the adult, Ptf1a is only expressed in acinar cells as a component of PTF1, a heterotrimeric transcriptional complex including a ubiquitous E-protein and Recombination signal-binding protein J ike (Rbpjl) [24,25,26]. During early pancreatic development, Ptf1a requires the interaction with the Rbpj isoform for pancreatic growth and morphogenesis. Then at the onset of acinar cell development, Rbpj is r.

Share this post on:

Author: Proteasome inhibitor