Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 2483, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38509065

ABSTRACT

Missense variants are the most common type of coding genetic variants. Their functional assessment is fundamental for defining any implication in human diseases and may also uncover genes that are essential for human organ development. Here, we apply CRISPR-Cas9 gene editing on human iPSCs to study a heterozygous missense variant in GLI2 identified in two siblings with early-onset and insulin-dependent diabetes of unknown cause. GLI2 is a primary mediator of the Hedgehog pathway, which regulates pancreatic ß-cell development in mice. However, neither mutations in GLI2 nor Hedgehog dysregulation have been reported as cause or predisposition to diabetes. We establish and study a set of isogenic iPSC lines harbouring the missense variant for their ability to differentiate into pancreatic ß-like cells. Interestingly, iPSCs carrying the missense variant show altered GLI2 transcriptional activity and impaired differentiation of pancreatic progenitors into endocrine cells. RNASeq and network analyses unveil a crosstalk between Hedgehog and WNT pathways, with the dysregulation of non-canonical WNT signaling in pancreatic progenitors carrying the GLI2 missense variant. Collectively, our findings underscore an essential role for GLI2 in human endocrine development and identify a gene variant that may lead to diabetes.


Subject(s)
Diabetes Mellitus , Islets of Langerhans , Humans , Mice , Animals , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Zinc Finger Protein Gli2/genetics , Mutation, Missense/genetics , Islets of Langerhans/metabolism , Kruppel-Like Transcription Factors/metabolism , Nuclear Proteins/metabolism
2.
Dev Cell ; 59(3): 326-338.e5, 2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38237591

ABSTRACT

During organ formation, progenitor cells need to acquire different cell identities and organize themselves into distinct structural units. How these processes are coordinated and how tissue architecture(s) is preserved despite the dramatic cell rearrangements occurring in developing organs remain unclear. Here, we identified cellular rearrangements between acinar and ductal progenitors as a mechanism to drive branching morphogenesis in the pancreas while preserving the integrity of the acinar-ductal functional unit. Using ex vivo and in vivo mouse models, we found that pancreatic ductal cells form clefts by protruding and pulling on the acinar basement membrane, which leads to acini splitting. Newly formed acini remain connected to the bifurcated branches generated by ductal cell rearrangement. Insulin growth factor (IGF)/phosphatidylinositol 3-kinase (PI3K) pathway finely regulates this process by controlling pancreatic ductal tissue fluidity, with a simultaneous impact on branching and cell fate acquisition. Together, our results explain how acinar structure multiplication and branch bifurcation are synchronized during pancreas organogenesis.


Subject(s)
Phosphatidylinositol 3-Kinase , Phosphatidylinositol 3-Kinases , Mice , Animals , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Pancreas , Acinar Cells/metabolism , Morphogenesis/physiology , Intercellular Signaling Peptides and Proteins/metabolism
3.
Int J Mol Sci ; 24(12)2023 Jun 17.
Article in English | MEDLINE | ID: mdl-37373416

ABSTRACT

The pancreas is a complex organ consisting of differentiated cells and extracellular matrix (ECM) organized adequately to enable its endocrine and exocrine functions. Although much is known about the intrinsic factors that control pancreas development, very few studies have focused on the microenvironment surrounding pancreatic cells. This environment is composed of various cells and ECM components, which play a critical role in maintaining tissue organization and homeostasis. In this study, we applied mass spectrometry to identify and quantify the ECM composition of the developing pancreas at the embryonic (E) day 14.5 and postnatal (P) day 1 stages. Our proteomic analysis identified 160 ECM proteins that displayed a dynamic expression profile with a shift in collagens and proteoglycans. Furthermore, we used atomic force microscopy to measure the biomechanical properties and found that the pancreatic ECM was soft (≤400 Pa) with no significant change during pancreas maturation. Lastly, we optimized a decellularization protocol for P1 pancreatic tissues, incorporating a preliminary crosslinking step, which effectively preserved the 3D organization of the ECM. The resulting ECM scaffold proved suitable for recellularization studies. Our findings provide insights into the composition and biomechanics of the pancreatic embryonic and perinatal ECM, offering a foundation for future studies investigating the dynamic interactions between the ECM and pancreatic cells.


Subject(s)
Proteomics , Tissue Engineering , Tissue Engineering/methods , Proteomics/methods , Extracellular Matrix/metabolism , Pancreas/metabolism , Extracellular Matrix Proteins/metabolism , Pancreatic Hormones/metabolism , Tissue Scaffolds/chemistry
4.
Dev Cell ; 58(9): 727-743.e11, 2023 05 08.
Article in English | MEDLINE | ID: mdl-37040771

ABSTRACT

Pancreatic islet cells derived from human pluripotent stem cells hold great promise for modeling and treating diabetes. Differences between stem-cell-derived and primary islets remain, but molecular insights to inform improvements are limited. Here, we acquire single-cell transcriptomes and accessible chromatin profiles during in vitro islet differentiation and pancreas from childhood and adult donors for comparison. We delineate major cell types, define their regulomes, and describe spatiotemporal gene regulatory relationships between transcription factors. CDX2 emerged as a regulator of enterochromaffin-like cells, which we show resemble a transient, previously unrecognized, serotonin-producing pre-ß cell population in fetal pancreas, arguing against a proposed non-pancreatic origin. Furthermore, we observe insufficient activation of signal-dependent transcriptional programs during in vitro ß cell maturation and identify sex hormones as drivers of ß cell proliferation in childhood. Altogether, our analysis provides a comprehensive understanding of cell fate acquisition in stem-cell-derived islets and a framework for manipulating cell identities and maturity.


Subject(s)
Insulin-Secreting Cells , Islets of Langerhans , Pluripotent Stem Cells , Adult , Humans , Pancreas , Cell Differentiation/genetics
5.
Front Cell Dev Biol ; 11: 1083175, 2023.
Article in English | MEDLINE | ID: mdl-36819106

ABSTRACT

Cell-cell interactions underlay organ formation and function during homeostasis. Changes in communication between cells and their surrounding microenvironment are a feature of numerous human diseases, including metabolic disease and neurological disorders. In the past decade, cross-disciplinary research has been conducted to engineer novel synthetic multicellular organ systems in 3D, including organoids, assembloids, and organ-on-chip models. These model systems, composed of distinct cell types, satisfy the need for a better understanding of complex biological interactions and mechanisms underpinning diseases. In this review, we discuss the emerging field of building 3D multicellular systems and their application for modelling the cellular interactions at play in diseases. We report recent experimental and computational approaches for capturing cell-cell interactions as well as progress in bioengineering approaches for recapitulating these complexities ex vivo. Finally, we explore the value of developing such multicellular systems for modelling metabolic, intestinal, and neurological disorders as major examples of multisystemic diseases, we discuss the advantages and disadvantages of the different approaches and provide some recommendations for further advancing the field.

6.
Sci Rep ; 12(1): 12498, 2022 07 21.
Article in English | MEDLINE | ID: mdl-35864120

ABSTRACT

Development of the pancreas is driven by an intrinsic program coordinated with signals from other cell types in the epithelial environment. These intercellular communications have been so far challenging to study because of the low concentration, localized production and diversity of the signals released. Here, we combined scRNAseq data with a computational interactomic approach to identify signals involved in the reciprocal interactions between the various cell types of the developing pancreas. This in silico approach yielded 40,607 potential ligand-target interactions between the different main pancreatic cell types. Among this vast network of interactions, we focused on three ligands potentially involved in communications between epithelial and endothelial cells. BMP7 and WNT7B, expressed by pancreatic epithelial cells and predicted to target endothelial cells, and SEMA6D, involved in the reverse interaction. In situ hybridization confirmed the localized expression of Bmp7 in the pancreatic epithelial tip cells and of Wnt7b in the trunk cells. On the contrary, Sema6d was enriched in endothelial cells. Functional experiments on ex vivo cultured pancreatic explants indicated that tip cell-produced BMP7 limited development of endothelial cells. This work identified ligands with a restricted tissular and cellular distribution and highlighted the role of BMP7 in the intercellular communications contributing to vessel development and organization during pancreas organogenesis.


Subject(s)
Endothelial Cells , Organogenesis , Cell Differentiation/physiology , Endothelial Cells/metabolism , Ligands , Organogenesis/physiology , Pancreas/metabolism
7.
Stem Cell Reports ; 17(5): 1033-1047, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35487213

ABSTRACT

Induced pluripotent stem cells (iPSCs) are valuable in disease modeling because of their potential to expand and differentiate into virtually any cell type and recapitulate key aspects of human biology. Functional genomics are genome-wide studies that aim to discover genotype-phenotype relationships, thereby revealing the impact of human genetic diversity on normal and pathophysiology. In this review, we make the case that human iPSCs (hiPSCs) are a powerful tool for functional genomics, since they provide an in vitro platform for the study of population genetics. We describe cutting-edge tools and strategies now available to researchers, including multi-omics technologies, advances in hiPSC culture techniques, and innovations in drug development. Functional genomics approaches based on hiPSCs hold great promise for advancing drug discovery, disease etiology, and the impact of genetic variation on human biology.


Subject(s)
Induced Pluripotent Stem Cells , Drug Development , Drug Discovery/methods , Genomics
8.
Development ; 149(3)2022 02 01.
Article in English | MEDLINE | ID: mdl-35037942

ABSTRACT

Generating comprehensive image maps, while preserving spatial three-dimensional (3D) context, is essential in order to locate and assess quantitatively specific cellular features and cell-cell interactions during organ development. Despite recent advances in 3D imaging approaches, our current knowledge of the spatial organization of distinct cell types in the embryonic pancreatic tissue is still largely based on two-dimensional histological sections. Here, we present a light-sheet fluorescence microscopy approach to image the pancreas in three dimensions and map tissue interactions at key time points in the mouse embryo. We demonstrate the utility of the approach by providing volumetric data, 3D distribution of three main cellular components (epithelial, mesenchymal and endothelial cells) within the developing pancreas, and quantification of their relative cellular abundance within the tissue. Interestingly, our 3D images show that endocrine cells are constantly and increasingly in contact with endothelial cells forming small vessels, whereas the interactions with mesenchymal cells decrease over time. These findings suggest distinct cell-cell interaction requirements for early endocrine cell specification and late differentiation. Lastly, we combine our image data in an open-source online repository (referred to as the Pancreas Embryonic Cell Atlas).


Subject(s)
Imaging, Three-Dimensional/methods , Pancreas/anatomy & histology , Animals , Embryo, Mammalian/anatomy & histology , Embryonic Development , Endothelial Cells/cytology , Endothelial Cells/metabolism , Epithelium/anatomy & histology , Homeobox Protein Nkx-2.5/deficiency , Homeobox Protein Nkx-2.5/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence
9.
Dev Cell ; 56(19): 2703-2711.e5, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34499867

ABSTRACT

Glucose homeostasis depends on regulated insulin secretion from pancreatic ß cells, which acquire their mature phenotype postnatally. The functional maturation of ß cells is regulated by a combination of cell-autonomous and exogenous factors; the identity of the latter is mostly unknown. Here, we identify BMP4 as a critical component through which the pancreatic microenvironment regulates ß cell function. By combining transgenic mouse models and human iPSCs, we show that BMP4 promotes the expression of core ß cell genes and is required for proper insulin production and secretion. We identified pericytes as the primary pancreatic source of BMP4, which start producing this ligand midway through the postnatal period, at the age ß cells mature. Overall, our findings show that the islet niche directly promotes ß cell functional maturation through the timely production of BMP4. Our study highlights the need to recapitulate the physiological postnatal islet niche for generating fully functional stem-cell-derived ß cells for cell replacement therapy for diabetes.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Insulin-Secreting Cells/metabolism , Pancreas/metabolism , Animals , Animals, Newborn , Bone Morphogenetic Protein 4/physiology , Cell Differentiation/genetics , Gene Expression/genetics , Gene Expression Regulation/genetics , Glucose/metabolism , Homeodomain Proteins/metabolism , Homeostasis , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organogenesis , Pancreas/physiology , Pericytes/metabolism , Trans-Activators/metabolism
10.
Nature ; 597(7874): 87-91, 2021 09.
Article in English | MEDLINE | ID: mdl-34433966

ABSTRACT

Studies based on single cells have revealed vast cellular heterogeneity in stem cell and progenitor compartments, suggesting continuous differentiation trajectories with intermixing of cells at various states of lineage commitment and notable degrees of plasticity during organogenesis1-5. The hepato-pancreato-biliary organ system relies on a small endoderm progenitor compartment that gives rise to a variety of different adult tissues, including the liver, pancreas, gall bladder and extra-hepatic bile ducts6,7. Experimental manipulation of various developmental signals in the mouse embryo has underscored important cellular plasticity in this embryonic territory6. This is reflected in the existence of human genetic syndromes as well as congenital malformations featuring multi-organ phenotypes in liver, pancreas and gall bladder6. Nevertheless, the precise lineage hierarchy and succession of events leading to the segregation of an endoderm progenitor compartment into hepatic, biliary and pancreatic structures have not yet been established. Here we combine computational modelling approaches with genetic lineage tracing to accurately reconstruct the hepato-pancreato-biliary lineage tree. We show that a multipotent progenitor subpopulation persists in the pancreato-biliary organ rudiment, contributing cells not only to the pancreas and gall bladder but also to the liver. Moreover, using single-cell RNA sequencing and functional experiments we define a specialized niche that supports this subpopulation in a multipotent state for an extended time during development. Together these findings indicate sustained plasticity underlying hepato-pancreato-biliary development that might also explain the rapid expansion of the liver while attenuating pancreato-biliary growth.


Subject(s)
Biliary Tract/cytology , Cell Lineage , Liver/cytology , Pancreas/cytology , Stem Cell Niche , Animals , Biliary Tract/embryology , Biliary Tract/metabolism , Cell Lineage/genetics , Cell Tracking , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Liver/embryology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Biological , Pancreas/embryology , Pancreas/metabolism , RNA-Seq , Signal Transduction , Single-Cell Analysis , Stem Cell Niche/genetics
11.
Development ; 148(14)2021 07 15.
Article in English | MEDLINE | ID: mdl-34251450

ABSTRACT

The second EMBO-EMBL Symposium 'Synthetic Morphogenesis: From Gene Circuits to Tissue Architecture' was held virtually in March 2021, with participants from all over the world joining from the comfort of their sofas to discuss synthetic morphogenesis at large. Leading scientists from a range of disciplines, including developmental biology, physics, chemistry and computer science, covered a gamut of topics from the principles of cell and tissue organization, patterning and gene regulatory networks, to synthetic approaches for exploring evolutionary and developmental biology principles. Here, we describe some of the high points.


Subject(s)
Developmental Biology , Gene Regulatory Networks , Genetic Engineering , Morphogenesis/genetics , Biological Evolution , Humans , Signal Transduction , Synthetic Biology , Systems Biology
12.
Curr Opin Genet Dev ; 70: 32-39, 2021 10.
Article in English | MEDLINE | ID: mdl-34062490

ABSTRACT

Diabetes is a group of metabolic disorders, which results from insufficient functional pancreatic ß-cell mass either due to the autoimmune destruction of insulin producing ß-cells, or their death or de-differentiation as compensation for insulin resistance. The ability to reprogram cell types within close developmental proximity to ß-cells offers a strategy to replenish ß-cell mass and a future possible treatment of diabetes. Here, we review recent advances in the fields of pancreas development and lineage reprogramming. We also probe the possibility of using reprogrammed cells as an approach by which to further understand developmental mechanisms, in particular roadblocks to changing cell identity. Finally, we highlight fundamental challenges that need to be overcome to advance lineage reprogramming for generating pancreatic cells.


Subject(s)
Cellular Reprogramming/physiology , Pancreas/cytology , Animals , Cell Lineage , Cell Plasticity , Cellular Reprogramming Techniques/methods , Gene Expression Regulation , Humans , Pancreas/embryology , Pancreas/growth & development , Transcription Factors/genetics , Transcription Factors/metabolism
13.
EMBO J ; 40(10): e106785, 2021 05 17.
Article in English | MEDLINE | ID: mdl-33934382

ABSTRACT

The interplay between extrinsic signaling and downstream gene networks controls the establishment of cell identity during development and its maintenance in adult life. Advances in next-generation sequencing and single-cell technologies have revealed additional layers of complexity in cell identity. Here, we review our current understanding of transcription factor (TF) networks as key determinants of cell identity. We discuss the concept of the core regulatory circuit as a set of TFs and interacting factors that together define the gene expression profile of the cell. We propose the core regulatory circuit as a comprehensive conceptual framework for defining cellular identity and discuss its connections to cell function in different contexts.


Subject(s)
Regenerative Medicine/methods , Humans , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Dev Cell ; 55(2): 150-162.e6, 2020 10 26.
Article in English | MEDLINE | ID: mdl-32857951

ABSTRACT

The interplay between pancreatic epithelium and the surrounding microenvironment is pivotal for pancreas formation and differentiation as well as adult organ homeostasis. The mesenchyme is the main component of the embryonic pancreatic microenvironment, yet its cellular identity is broadly defined, and whether it comprises functionally distinct cell subsets is not known. Using genetic lineage tracing, transcriptome, and functional studies, we identified mesenchymal populations with different roles during pancreatic development. Moreover, we showed that Pbx transcription factors act within the mouse pancreatic mesenchyme to define a pro-endocrine specialized niche. Pbx directs differentiation of endocrine progenitors into insulin- and glucagon-positive cells through non-cell-autonomous regulation of ECM-integrin interactions and soluble molecules. Next, we measured functional conservation between mouse and human pancreatic mesenchyme by testing identified mesenchymal factors in an iPSC-based differentiation model. Our findings provide insights into how lineage-specific crosstalk between epithelium and neighboring mesenchymal cells underpin the generation of different pancreatic cell types.


Subject(s)
Cell Differentiation/physiology , Mesenchymal Stem Cells/metabolism , Mesoderm/metabolism , Pancreas/metabolism , Animals , Endocrine System , Epithelium/metabolism , Gene Expression Regulation, Developmental/genetics , Humans , Mice, Transgenic , Organogenesis/physiology , Pancreas/pathology
15.
Article in English | MEDLINE | ID: mdl-31767653

ABSTRACT

Direct lineage reprogramming of abundant and accessible cells into therapeutically useful cell types holds tremendous potential in regenerative medicine. To date, a number of different cell types have been generated by lineage reprogramming methods, including cells from the neural, cardiac, hepatic, and pancreatic lineages. The success of this strategy relies on developmental biology and the knowledge of cell-fate-defining transcriptional networks. Hepatocytes represent a prime target for ß cell conversion for numerous reasons, including close developmental origin, accessibility, and regenerative potential. We present here an overview of pancreatic and hepatic development, with a particular focus on the mechanisms underlying the divergence between the two cell lineages. Additionally, we discuss to what extent this lineage relationship can be exploited in efforts to reprogram one cell type into the other and whether such an approach may provide a suitable strategy for regenerative therapies of diabetes.


Subject(s)
Cell Lineage , Cell Plasticity , Cellular Reprogramming , Liver/physiology , Pancreas/physiology , Animals , Cell Differentiation , Diabetes Mellitus/physiopathology , Hepatocytes/metabolism , Humans , Insulin-Secreting Cells/metabolism , Ligands , Liver/embryology , Mice , Pancreas/embryology , Regeneration , Transcription Factors/metabolism
16.
Development ; 146(14)2019 07 24.
Article in English | MEDLINE | ID: mdl-31142539

ABSTRACT

An early step in pancreas development is marked by the expression of the transcription factor Pdx1 within the pancreatic endoderm, where it is required for the specification of all endocrine cell types. Subsequently, Pdx1 expression becomes restricted to the ß-cell lineage, where it plays a central role in ß-cell function. This pivotal role of Pdx1 at various stages of pancreas development makes it an attractive target to enhance pancreatic ß-cell differentiation and increase ß-cell function. In this study, we used a newly generated zebrafish reporter to screen over 8000 small molecules for modulators of pdx1 expression. We found four hit compounds and validated their efficacy at different stages of pancreas development. Notably, valproic acid treatment increased pancreatic endoderm formation, while inhibition of TGFß signaling led to α-cell to ß-cell transdifferentiation. HC toxin, another HDAC inhibitor, enhances ß-cell function in primary mouse and human islets. Thus, using a whole organism screening strategy, this study identified new pdx1 expression modulators that can be used to influence different steps in pancreas and ß-cell development.


Subject(s)
Drug Evaluation, Preclinical/methods , Islets of Langerhans/embryology , Models, Animal , Organogenesis/drug effects , Small Molecule Libraries/analysis , Zebrafish , Animals , Animals, Genetically Modified , COS Cells , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Transdifferentiation/drug effects , Cell Transdifferentiation/genetics , Cells, Cultured , Chlorocebus aethiops , Embryo, Nonmammalian , Gene Expression Regulation, Developmental/drug effects , Histone Deacetylase Inhibitors/isolation & purification , Histone Deacetylase Inhibitors/pharmacology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Islets of Langerhans/drug effects , Islets of Langerhans/growth & development , Islets of Langerhans/metabolism , Mice , Mice, Inbred C57BL , Organogenesis/genetics , Small Molecule Libraries/isolation & purification , Trans-Activators/genetics , Trans-Activators/metabolism , Valproic Acid/isolation & purification , Valproic Acid/pharmacology , Zebrafish/embryology , Zebrafish/genetics
17.
Curr Top Dev Biol ; 132: 221-256, 2019.
Article in English | MEDLINE | ID: mdl-30797510

ABSTRACT

During embryonic development, pancreatic epithelial cells engage in concomitant morphogenetic and fate specification events that will give rise to the final organ architecture and functions. Cues from the surrounding microenvironment are known to influence the behavior of epithelial progenitors and orchestrate these concomitant events throughout pancreas development. Nevertheless, the composition of the pancreatic microenvironment remains elusive; also, the interplay between components of the surrounding microenvironment and the epithelium is poorly characterized. We present here a comprehensive overview of the pancreatic microenvironment and what is known regarding distinct cell types, signaling molecules, ECM, that constitute it. We focus on the molecular circuits governing cell-cell interactions, which are at play in the developing pancreas, controlling pancreatic progenitor proliferation, morphogenesis, and differentiation. Finally, open questions and implication of future research in this field are discussed in the context of pancreatic diseases, such as diabetes and cancer, as well as therapeutic approaches for these diseases.


Subject(s)
Epithelial Cells/metabolism , Epithelium/embryology , Organogenesis , Pancreas/embryology , Animals , Cell Differentiation , Cellular Microenvironment , Humans , Intrinsic Factor/metabolism , Pancreas/cytology , Stem Cells/metabolism
18.
Nature ; 564(7734): 50-51, 2018 12.
Article in English | MEDLINE | ID: mdl-30510228
19.
Nat Commun ; 9(1): 5082, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30504829

ABSTRACT

A complex interplay of intrinsic factors and extrinsic signalling pathways controls both cell lineage commitment and maintenance of cell identity. Loss of defined cellular states is the cause of many different cancers, including pancreatic cancer. Recent findings suggest a clinical role for the conserved SLIT/ROBO signalling pathway in pancreatic cancer. However, whilst this pathway has been extensively studied in many processes, a role for Slit and Robo genes in pancreas cell identity and plasticity has not been established yet. Here, we identify Slit/Robo signalling as a key regulator of pancreatic progenitor identity. We find that Robo1 and Robo2 are required for preserving pancreatic cell identity shortly after fate induction and, subsequently, for expansion of the pancreatic progenitor pool in the mouse. Furthermore, we show that Robo receptors control the expression of Tead transcription factors as well as its downstream transcriptional activity. Our work identifies an interplay between Slit/Robo pathway and Tead intrinsic regulators, functioning as gatekeeper of pancreatic cell identity.


Subject(s)
Pancreas/cytology , Pancreas/metabolism , Signal Transduction/physiology , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dyneins/genetics , Dyneins/metabolism , Immunohistochemistry , In Situ Hybridization , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Reverse Transcriptase Polymerase Chain Reaction , TEA Domain Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism
20.
Mol Metab ; 8: 96-105, 2018 02.
Article in English | MEDLINE | ID: mdl-29310936

ABSTRACT

OBJECTIVE: Actin cytoskeleton remodeling is necessary for glucose-stimulated insulin secretion in pancreatic ß-cells. A mechanistic understanding of actin dynamics in the islet is paramount to a better comprehension of ß-cell dysfunction in diabetes. Here, we investigate the Rho GTPase regulator Stard13 and its role in F-actin cytoskeleton organization and islet function in adult mice. METHODS: We used Lifeact-EGFP transgenic animals to visualize actin cytoskeleton organization and dynamics in vivo in the mouse islets. Furthermore, we applied this model to study actin cytoskeleton and insulin secretion in mutant mice deleted for Stard13 selectively in pancreatic cells. We isolated transgenic islets for 3D-imaging and perifusion studies to measure insulin secretion dynamics. In parallel, we performed histological and morphometric analyses of the pancreas and used in vivo approaches to study glucose metabolism in the mouse. RESULTS: In this study, we provide the first genetic evidence that Stard13 regulates insulin secretion in response to glucose. Postnatally, Stard13 expression became restricted to the mouse pancreatic islets. We showed that Stard13 deletion results in a marked increase in actin polymerization in islet cells, which is accompanied by severe reduction of insulin secretion in perifusion experiments. Consistently, Stard13-deleted mice displayed impaired glucose tolerance and reduced glucose-stimulated insulin secretion. CONCLUSIONS: Taken together, our results suggest a previously unappreciated role for the RhoGAP protein Stard13 in the interplay between actin cytoskeletal remodeling and insulin secretion.


Subject(s)
Actins/metabolism , GTPase-Activating Proteins/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cells, Cultured , GTPase-Activating Proteins/genetics , Glucose/metabolism , Insulin-Secreting Cells/cytology , Mice , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...