Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
Nat Commun ; 13(1): 4540, 2022 08 04.
Article En | MEDLINE | ID: mdl-35927244

During pancreas development endocrine cells leave the ductal epithelium to form the islets of Langerhans, but the morphogenetic mechanisms are incompletely understood. Here, we identify the Ca2+-independent atypical Synaptotagmin-13 (Syt13) as a key regulator of endocrine cell egression and islet formation. We detect specific upregulation of the Syt13 gene and encoded protein in endocrine precursors and the respective lineage during islet formation. The Syt13 protein is localized to the apical membrane of endocrine precursors and to the front domain of egressing endocrine cells, marking a previously unidentified apical-basal to front-rear repolarization during endocrine precursor cell egression. Knockout of Syt13 impairs endocrine cell egression and skews the α-to-ß-cell ratio. Mechanistically, Syt13 is a vesicle trafficking protein, transported via the microtubule cytoskeleton, and interacts with phosphatidylinositol phospholipids for polarized localization. By internalizing a subset of plasma membrane proteins at the front domain, including α6ß4 integrins, Syt13 modulates cell-matrix adhesion and allows efficient endocrine cell egression. Altogether, these findings uncover an unexpected role for Syt13 as a morphogenetic driver of endocrinogenesis and islet formation.


Endocrine Cells , Islets of Langerhans , Integrins , Morphogenesis , Pancreas , Synaptotagmins/genetics
3.
Int J Mol Sci ; 22(22)2021 Nov 20.
Article En | MEDLINE | ID: mdl-34830411

Synaptotagmin-13 (Syt13) is an atypical member of the vesicle trafficking synaptotagmin protein family. The expression pattern and the biological function of this Ca2+-independent protein are not well resolved. Here, we have generated a novel Syt13-Venus fusion (Syt13-VF) fluorescence reporter allele to track and isolate tissues and cells expressing Syt13 protein. The reporter allele is regulated by endogenous cis-regulatory elements of Syt13 and the fusion protein follows an identical expression pattern of the endogenous Syt13 protein. The homozygous reporter mice are viable and fertile. We identify the expression of the Syt13-VF reporter in different regions of the brain with high expression in tyrosine hydroxylase (TH)-expressing and oxytocin-producing neuroendocrine cells. Moreover, Syt13-VF is highly restricted to all enteroendocrine cells in the adult intestine that can be traced in live imaging. Finally, Syt13-VF protein is expressed in the pancreatic endocrine lineage, allowing their specific isolation by flow sorting. These findings demonstrate high expression levels of Syt13 in the endocrine lineages in three major organs harboring these secretory cells. Collectively, the Syt13-VF reporter mouse line provides a unique and reliable tool to dissect the spatio-temporal expression pattern of Syt13 and enables isolation of Syt13-expressing cells that will aid in deciphering the molecular functions of this protein in the neuroendocrine system.


Brain/metabolism , Intestines/metabolism , Pancreas/metabolism , Synaptotagmins/genetics , Animals , Brain/pathology , Cell Line, Tumor , Cell Lineage/genetics , Cell Movement/genetics , Gene Expression Regulation/genetics , Humans , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Mice , Neurosecretory Systems/metabolism , Neurosecretory Systems/pathology , Oxytocin/genetics , Synaptotagmins/metabolism , Tyrosine 3-Monooxygenase/genetics
5.
Nat Cell Biol ; 23(7): 692-703, 2021 07.
Article En | MEDLINE | ID: mdl-34168324

It is generally accepted that epiblast cells ingress into the primitive streak by epithelial-to-mesenchymal transition (EMT) to give rise to the mesoderm; however, it is less clear how the endoderm acquires an epithelial fate. Here, we used embryonic stem cell and mouse embryo knock-in reporter systems to combine time-resolved lineage labelling with high-resolution single-cell transcriptomics. This allowed us to resolve the morphogenetic programs that segregate the mesoderm from the endoderm germ layer. Strikingly, while the mesoderm is formed by classical EMT, the endoderm is formed independent of the key EMT transcription factor Snail1 by mechanisms of epithelial cell plasticity. Importantly, forkhead box transcription factor A2 (Foxa2) acts as an epithelial gatekeeper and EMT suppressor to shield the endoderm from undergoing a mesenchymal transition. Altogether, these results not only establish the morphogenetic details of germ layer formation, but also have broader implications for stem cell differentiation and cancer metastasis.


Blastocyst/physiology , Cell Plasticity , Endoderm/physiology , Epithelial Cells/physiology , Epithelial-Mesenchymal Transition , Gastrulation , Mouse Embryonic Stem Cells/physiology , Animals , Blastocyst/cytology , Blastocyst/metabolism , Cell Differentiation , Cell Line , Endoderm/cytology , Endoderm/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Gestational Age , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Mice , Mice, Transgenic , Mouse Embryonic Stem Cells/metabolism , Phenotype , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Time Factors
6.
Mol Metab ; 49: 101188, 2021 07.
Article En | MEDLINE | ID: mdl-33582383

OBJECTIVE: Islets of Langerhans contain heterogeneous populations of insulin-producing ß-cells. Surface markers and respective antibodies for isolation, tracking, and analysis are urgently needed to study ß-cell heterogeneity and explore the mechanisms to harness the regenerative potential of immature ß-cells. METHODS: We performed single-cell mRNA profiling of early postnatal mouse islets and re-analyzed several single-cell mRNA sequencing datasets from mouse and human pancreas and islets. We used mouse primary islets, iPSC-derived endocrine cells, Min6 insulinoma, and human EndoC-ßH1 ß-cell lines and performed FAC sorting, Western blotting, and imaging to support and complement the findings from the data analyses. RESULTS: We found that all endocrine cell types expressed the cluster of differentiation 81 (CD81) during pancreas development, but the expression levels of this protein were gradually reduced in ß-cells during postnatal maturation. Single-cell gene expression profiling and high-resolution imaging revealed an immature signature of ß-cells expressing high levels of CD81 (CD81high) compared to a more mature population expressing no or low levels of this protein (CD81low/-). Analysis of ß-cells from different diabetic mouse models and in vitro ß-cell stress assays indicated an upregulation of CD81 expression levels in stressed and dedifferentiated ß-cells. Similarly, CD81 was upregulated and marked stressed human ß-cells in vitro. CONCLUSIONS: We identified CD81 as a novel surface marker that labels immature, stressed, and dedifferentiated ß-cells in the adult mouse and human islets. This novel surface marker will allow us to better study ß-cell heterogeneity in healthy subjects and diabetes progression.


Cell Differentiation , Insulin-Secreting Cells/metabolism , Tetraspanin 28/genetics , Tetraspanin 28/metabolism , Animals , Cell Line , Diabetes Mellitus/metabolism , Female , Gene Expression , Gene Expression Profiling/methods , Genetic Heterogeneity , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Pancreas/metabolism , RNA, Messenger/metabolism , Up-Regulation
7.
Nat Biotechnol ; 38(9): 1061-1072, 2020 09.
Article En | MEDLINE | ID: mdl-32341565

Methods for differentiating human pluripotent stem cells to pancreatic and liver lineages in vitro have been limited by the inability to identify and isolate distinct endodermal subpopulations specific to these two organs. Here we report that pancreatic and hepatic progenitors can be isolated using the surface markers CD177/NB1 glycoprotein and inducible T-cell costimulatory ligand CD275/ICOSL, respectively, from seemingly homogeneous definitive endoderm derived from human pluripotent stem cells. Anterior definitive endoderm (ADE) subpopulations identified by CD177 and CD275 show inverse activation of canonical and noncanonical WNT signaling. CD177+ ADE expresses and synthesizes the secreted WNT, NODAL and BMP antagonist CERBERUS1 and is specified toward the pancreatic fate. CD275+ ADE receives canonical Wnt signaling and is specified toward the liver fate. Isolated CD177+ ADE differentiates more homogeneously into pancreatic progenitors and into more functionally mature and glucose-responsive ß-like cells in vitro compared with cells from unsorted differentiation cultures.


Endoderm/cytology , Endoderm/metabolism , Insulin-Secreting Cells/cytology , Isoantigens/metabolism , Receptors, Cell Surface/metabolism , Adolescent , Adult , Biomarkers/metabolism , Cell Differentiation , Cell Line , Cell Lineage , Cytokines/metabolism , Female , GPI-Linked Proteins/metabolism , Humans , Inducible T-Cell Co-Stimulator Ligand/metabolism , Insulin-Secreting Cells/metabolism , Liver/cytology , Liver/metabolism , Male , Middle Aged , Pancreas/cytology , Pancreas/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Receptors, CXCR4/metabolism , Wnt Signaling Pathway/physiology , Young Adult
8.
Mol Metab ; 30: 16-29, 2019 12.
Article En | MEDLINE | ID: mdl-31767167

OBJECTIVE: Translation of basic research from bench-to-bedside relies on a better understanding of similarities and differences between mouse and human cell biology, tissue formation, and organogenesis. Thus, establishing ex vivo modeling systems of mouse and human pancreas development will help not only to understand evolutionary conserved mechanisms of differentiation and morphogenesis but also to understand pathomechanisms of disease and design strategies for tissue engineering. METHODS: Here, we established a simple and reproducible Matrigel-based three-dimensional (3D) cyst culture model system of mouse and human pancreatic progenitors (PPs) to study pancreatic epithelialization and endocrinogenesis ex vivo. In addition, we reanalyzed previously reported single-cell RNA sequencing (scRNA-seq) of mouse and human pancreatic lineages to obtain a comprehensive picture of differential expression of key transcription factors (TFs), cell-cell adhesion molecules and cell polarity components in PPs during endocrinogenesis. RESULTS: We generated mouse and human polarized pancreatic epithelial cysts derived from PPs. This system allowed to monitor establishment of pancreatic epithelial polarity and lumen formation in cellular and sub-cellular resolution in a dynamic time-resolved fashion. Furthermore, both mouse and human pancreatic cysts were able to differentiate towards the endocrine fate. This differentiation system together with scRNA-seq analysis revealed how apical-basal polarity and tight and adherens junctions change during endocrine differentiation. CONCLUSIONS: We have established a simple 3D pancreatic cyst culture system that allows to tempo-spatial resolve cellular and subcellular processes on the mechanistical level, which is otherwise not possible in vivo.


Cell Culture Techniques/methods , Organoids/metabolism , Pancreas/metabolism , Animals , Cell Differentiation , Cell Line , Epithelial Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Organogenesis/physiology , Organoids/physiology , Stem Cells/metabolism , Transcription Factors/metabolism
9.
Curr Opin Cell Biol ; 61: 48-55, 2019 12.
Article En | MEDLINE | ID: mdl-31377680

The pancreas is derived from the foregut endoderm during embryonic development. After gastrulation and endoderm germ layer formation complex morphogenetic events coupled with cell differentiation programs pattern the gut tube and induce pancreas organogenesis. This results in formation of exocrine, ductal and hormone-producing endocrine cells. Among these, endocrine cells are responsible for blood glucose homeostasis and their malfunction leads to diabetes mellitus, which cannot be stopped or reversed by the current standard treatments. Thus, intense efforts to regenerate or replace the lost or dysfunctional insulin-producing ß-cells are on the way. This depends on identifying the factors that coordinate pancreas organogenesis. Here, we highlight the contribution of canonical and non-canonical Wnt signaling branches in orchestrating endoderm formation, pancreatic morphogenesis as well as endocrine cell formation and function.


Embryonic Development/physiology , Endoderm/embryology , Pancreas/embryology , Wnt Signaling Pathway/physiology , Animals , Cell Differentiation/physiology , Diabetes Mellitus/physiopathology , Exocrine Glands/embryology , Humans , Insulin , Organogenesis
10.
Nucleic Acids Res ; 47(17): 9069-9086, 2019 09 26.
Article En | MEDLINE | ID: mdl-31350899

Pioneer transcription factors (PTF) can recognize their binding sites on nucleosomal DNA and trigger chromatin opening for recruitment of other non-pioneer transcription factors. However, critical properties of PTFs are still poorly understood, such as how these transcription factors selectively recognize cell type-specific binding sites and under which conditions they can initiate chromatin remodelling. Here we show that early endoderm binding sites of the paradigm PTF Foxa2 are epigenetically primed by low levels of active chromatin modifications in embryonic stem cells (ESC). Priming of these binding sites is supported by preferential recruitment of Foxa2 to endoderm binding sites compared to lineage-inappropriate binding sites, when ectopically expressed in ESCs. We further show that binding of Foxa2 is required for chromatin opening during endoderm differentiation. However, increased chromatin accessibility was only detected on binding sites which are synergistically bound with other endoderm transcription factors. Thus, our data suggest that binding site selection of PTFs is directed by the chromatin environment and that chromatin opening requires collaboration of PTFs with additional transcription factors.


Chromatin/metabolism , Hepatocyte Nuclear Factor 3-beta/metabolism , Mouse Embryonic Stem Cells/metabolism , Animals , Binding Sites/genetics , Cell Differentiation/genetics , Chromatin Assembly and Disassembly/genetics , Endoderm/cytology , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Gene Expression Regulation, Developmental/genetics , Hepatocyte Nuclear Factor 3-beta/genetics , Histone Code , Histones/metabolism , Mice , Mice, Knockout , Models, Genetic , Mouse Embryonic Stem Cells/cytology , Signal Transduction
11.
Development ; 146(12)2019 06 17.
Article En | MEDLINE | ID: mdl-31160421

Deciphering mechanisms of endocrine cell induction, specification and lineage allocation in vivo will provide valuable insights into how the islets of Langerhans are generated. Currently, it is ill defined how endocrine progenitors segregate into different endocrine subtypes during development. Here, we generated a novel neurogenin 3 (Ngn3)-Venus fusion (NVF) reporter mouse line, that closely mirrors the transient endogenous Ngn3 protein expression. To define an in vivo roadmap of endocrinogenesis, we performed single cell RNA sequencing of 36,351 pancreatic epithelial and NVF+ cells during secondary transition. This allowed Ngn3low endocrine progenitors, Ngn3high endocrine precursors, Fev+ endocrine lineage and hormone+ endocrine subtypes to be distinguished and time-resolved, and molecular programs during the step-wise lineage restriction steps to be delineated. Strikingly, we identified 58 novel signature genes that show the same transient expression dynamics as Ngn3 in the 7260 profiled Ngn3-expressing cells. The differential expression of these genes in endocrine precursors associated with their cell-fate allocation towards distinct endocrine cell types. Thus, the generation of an accurately regulated NVF reporter allowed us to temporally resolve endocrine lineage development to provide a fine-grained single cell molecular profile of endocrinogenesis in vivo.


Basic Helix-Loop-Helix Transcription Factors/genetics , Nerve Tissue Proteins/genetics , Pancreas/embryology , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Animals , Cell Differentiation/genetics , Cell Lineage , Endocrine Cells/cytology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genes, Reporter , Insulin-Secreting Cells/cytology , Mice , Regeneration , Signal Transduction , Stem Cells/cytology , Wnt Proteins/metabolism
12.
Development ; 144(16): 2873-2888, 2017 08 15.
Article En | MEDLINE | ID: mdl-28811309

The pancreas is an endoderm-derived glandular organ that participates in the regulation of systemic glucose metabolism and food digestion through the function of its endocrine and exocrine compartments, respectively. While intensive research has explored the signaling pathways and transcriptional programs that govern pancreas development, much remains to be discovered regarding the cellular processes that orchestrate pancreas morphogenesis. Here, we discuss the developmental mechanisms and principles that are known to underlie pancreas development, from induction and lineage formation to morphogenesis and organogenesis. Elucidating such principles will help to identify novel candidate disease genes and unravel the pathogenesis of pancreas-related diseases, such as diabetes, pancreatitis and cancer.


Pancreas/embryology , Animals , Endoderm/embryology , Endoderm/metabolism , Gene Expression Regulation, Developmental , Humans , Pancreas/metabolism , Signal Transduction
...