Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Diabetologia ; 67(10): 2275-2288, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38958700

ABSTRACT

AIMS/HYPOTHESIS: Strategies to augment functional beta cell mass include directed differentiation of stem cells towards a beta cell fate, which requires extensive knowledge of transcriptional programs governing endocrine progenitor cell differentiation in vivo. We aimed to study the contributions of the Brahma-related gene-1 (BRG1) and Brahma (BRM) ATPase subunits of the SWI/SNF chromatin remodelling complex to endocrine cell development. METHODS: We generated mice with endocrine progenitor-specific Neurog3-Cre BRG1 removal in the presence of heterozygous (Brg1Δendo;Brm+/-) or homozygous (double knockout: DKOΔendo) BRM deficiency. Whole-body metabolic phenotyping, islet function characterisation, islet quantitative PCR and histological characterisation were performed on animals and tissues postnatally. To test the mechanistic actions of SWI/SNF in controlling gene expression during endocrine cell development, single-cell RNA-seq was performed on flow-sorted endocrine-committed cells from embryonic day 15.5 control and mutant embryos. RESULTS: Brg1Δendo;Brm+/- mice exhibit severe glucose intolerance, hyperglycaemia and hypoinsulinaemia, resulting, in part, from reduced islet number; diminished alpha, beta and delta cell mass; compromised islet insulin secretion; and altered islet gene expression programs, including reductions in MAFA and urocortin 3 (UCN3). DKOΔendo mice were not recovered at weaning; however, postnatal day 6 DKOΔendo mice were severely hyperglycaemic with reduced serum insulin levels and beta cell area. Single-cell RNA-seq of embryonic day 15.5 lineage-labelled cells revealed endocrine progenitor, alpha and beta cell populations from SWI/SNF mutants have reduced expression of Mafa, Gcg, Ins1 and Ins2, suggesting limited differentiation capacity. Reduced Neurog3 transcripts were discovered in DKOΔendo endocrine progenitor clusters, and the proliferative capacity of neurogenin 3 (NEUROG3)+ cells was reduced in Brg1Δendo;Brm+/- and DKOΔendo mutants. CONCLUSIONS/INTERPRETATION: Loss of BRG1 from developing endocrine progenitor cells has a severe postnatal impact on glucose homeostasis, and loss of both subunits impedes animal survival, with both groups exhibiting alterations in hormone transcripts embryonically. Taken together, these data highlight the critical role SWI/SNF plays in governing gene expression programs essential for endocrine cell development and expansion. DATA AVAILABILITY: Raw and processed data for scRNA-seq have been deposited into the NCBI Gene Expression Omnibus (GEO) database under the accession number GSE248369.


Subject(s)
Cell Differentiation , Chromatin Assembly and Disassembly , DNA Helicases , Transcription Factors , Animals , Mice , Cell Differentiation/physiology , Transcription Factors/metabolism , Transcription Factors/genetics , DNA Helicases/metabolism , DNA Helicases/genetics , Mice, Knockout , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/cytology , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Islets of Langerhans/metabolism , Islets of Langerhans/cytology , Male , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Cell Proliferation/genetics , SMARCB1 Protein
2.
J Tissue Eng ; 14: 20417314231185858, 2023.
Article in English | MEDLINE | ID: mdl-37435573

ABSTRACT

Induced pluripotent stem cells (iPSCs) have enormous potential in producing human tissues endlessly. We previously reported that type V collagen (COL5), a pancreatic extracellular matrix protein, promotes islet development and maturation from iPSCs. In this study, we identified a bioactive peptide domain of COL5, WWASKS, through bioinformatic analysis of decellularized pancreatic ECM (dpECM)-derived collagens. RNA-sequencing suggests that WWASKS induces the formation of pancreatic endocrine progenitors while suppressing the development of other types of organs. The expressions of hypoxic genes were significantly downregulated in the endocrine progenitors formed under peptide stimulation. Furthermore, we unveiled an enhancement of iPSC-derived islets' (i-islets) glucose sensitivity under peptide stimulation. These i-islets secrete insulin in a glucose responsive manner. They were comprised of α, ß, δ, and γ cells and were assembled into a tissue architecture similar to that of human islets. Mechanistically, the peptide is able to activate the canonical Wnt signaling pathway, permitting the translocation of ß-catenin from the cytoplasm to the nucleus for pancreatic progenitor development. Collectively, for the first time, we demonstrated that an ECM-derived peptide dictates iPSC fate toward the generation of endocrine progenitors and subsequent islet organoids.

3.
Cell Rep ; 40(9): 111260, 2022 08 30.
Article in English | MEDLINE | ID: mdl-36044861

ABSTRACT

Hematopoiesis was considered a hierarchical stepwise process but was revised to a continuous process following single-cell RNA sequencing. However, the uncertainty or fluctuation of single-cell transcriptome dynamics during differentiation was not considered, and the dendritic cell (DC) pathway in the lymphoid context remains unclear. Here, we identify human B-plasmacytoid DC (pDC) bifurcation as large fluctuating transcriptome dynamics in the putative B/NK progenitor region by dry and wet methods. By converting splicing kinetics into diffusion dynamics in a deep generative model, our original computational methodology reveals strong fluctuation at B/pDC bifurcation in IL-7Rα+ regions, and LFA-1 fluctuates positively in the pDC direction at the bifurcation. These expectancies are validated by the presence of B/pDC progenitors in the IL-7Rα+ fraction and preferential expression of LFA-1 in pDC-biased progenitors with a niche-like culture system. We provide a model of fluctuation-based differentiation, which reconciles continuous and discrete models and is applicable to other developmental systems.


Subject(s)
Cell Differentiation , Dendritic Cells , Lymphocyte Function-Associated Antigen-1 , Cell Differentiation/genetics , Dendritic Cells/metabolism , Hematopoiesis , Humans , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Transcriptome/genetics
4.
Front Endocrinol (Lausanne) ; 12: 736286, 2021.
Article in English | MEDLINE | ID: mdl-34566896

ABSTRACT

NEUROGENIN3+ (NEUROG3+) cells are considered to be pancreatic endocrine progenitors. Our current knowledge on the molecular program of NEUROG3+ cells in humans is largely extrapolated from studies in mice. We hypothesized that single-cell RNA-seq enables in-depth exploration of the rare NEUROG3+ cells directly in humans. We aligned four large single-cell RNA-seq datasets from postnatal human pancreas. Our integrated analysis revealed 10 NEUROG3+ epithelial cells from a total of 11,174 pancreatic cells. Noticeably, human NEUROG3+ cells clustered with mature pancreatic cells and epsilon cells displayed the highest frequency of NEUROG3 positivity. We confirmed the co-expression of NEUROG3 with endocrine markers and the high percentage of NEUROG3+ cells among epsilon cells at the protein level based on immunostaining on pancreatic tissue sections. We further identified unique genetic signatures of the NEUROG3+ cells. Regulatory network inference revealed novel transcription factors including Prospero homeobox protein 1 (PROX1) may act jointly with NEUROG3. As NEUROG3 plays a central role in endocrine differentiation, knowledge gained from our study will accelerate the development of beta cell regeneration therapies to treat diabetes.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Endocrine Cells/metabolism , Nerve Tissue Proteins/genetics , Pancreas/metabolism , Stem Cells/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Humans , Nerve Tissue Proteins/metabolism
5.
G3 (Bethesda) ; 11(11)2021 10 19.
Article in English | MEDLINE | ID: mdl-34534285

ABSTRACT

Insm1, Neurod1, and Pax6 are essential for the formation and function of pancreatic endocrine cells. Here, we report comparative immunohistochemical, transcriptomic, functional enrichment, and RNA splicing analyses of these genes using gene knock-out mice. Quantitative immunohistochemical analysis confirmed that elimination of each of these three factors variably impairs the proliferation, survival, and differentiation of endocrine cells. Transcriptomic analysis revealed that each factor contributes uniquely to the transcriptome although their effects were overlapping. Functional enrichment analysis revealed that genes downregulated by the elimination of Insm1, Neurod1, and Pax6 are commonly involved in mRNA metabolism, chromatin organization, secretion, and cell cycle regulation, and upregulated genes are associated with protein degradation, autophagy, and apoptotic process. Elimination of Insm1, Neurod1, and Pax6 impaired expression of many RNA-binding proteins thereby altering RNA splicing events, including for Syt14 and Snap25, two genes required for insulin secretion. All three factors are necessary for normal splicing of Syt14, and both Insm1 and Pax6 are necessary for the processing of Snap25. Collectively, these data provide new insights into how Insm1, Neurod1, and Pax6 contribute to the formation of functional pancreatic endocrine cells.


Subject(s)
Endocrine Cells , Transcription Factors , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Endocrine Cells/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Mice , PAX6 Transcription Factor/genetics , RNA , RNA Splicing , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics
6.
Genes Cells ; 25(5): 302-311, 2020 May.
Article in English | MEDLINE | ID: mdl-32065490

ABSTRACT

Transplantation of pancreatic islets is an effective therapy for severe type 1 diabetes. As donor shortage is a major problem for this therapy, attempts have been made to produce a large number of pancreatic islets from human pluripotent stem cells (hPSCs). However, as the differentiation of hPSCs to pancreatic islets requires multiple and lengthy processes using various expensive cytokines, the process is variable, low efficiency and costly. Therefore, it would be beneficial if islet progenitors could be expanded. Neurogenin3 (NGN3)-expressing pancreatic endocrine progenitor (EP) cells derived from hPSCs exhibited the ability to differentiate into pancreatic islets while their cell cycle was arrested. By using a lentivirus vector, we introduced several growth-promoting genes into NGN3-expressing EP cells. We found that SV40LT expression induced proliferation of the EP cells but reduced the expression of endocrine lineage-commitment factors, NGN3, NEUROD1 and NKX2.2, resulting in the suppression of islet differentiation. By using the Cre-loxP system, we removed SV40LT after the expansion, leading to re-expression of endocrine-lineage commitment genes and differentiation into functional pancreatic islets. Thus, our findings will pave a way to generate a large quantity of functional pancreatic islets through the expansion of EP cells from hPSCs.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Homeodomain Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Islets of Langerhans/metabolism , Nerve Tissue Proteins/metabolism , Zebrafish Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Islets of Langerhans/cytology , Nerve Tissue Proteins/genetics , Nuclear Proteins , Transcription Factors , Zebrafish Proteins/genetics
7.
Development ; 146(12)2019 06 17.
Article in English | MEDLINE | ID: mdl-31160421

ABSTRACT

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.


Subject(s)
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
8.
J Theor Biol ; 430: 32-44, 2017 10 07.
Article in English | MEDLINE | ID: mdl-28652000

ABSTRACT

During pancreas development, Neurog3 positive endocrine progenitors are specified by Delta/Notch (D/N) mediated lateral inhibition in the growing ducts. During neurogenesis, genes that determine the transition from the proneural state to neuronal or glial lineages are oscillating before their expression is sustained. Although the basic gene regulatory network is very similar, cycling gene expression in pancreatic development was not investigated yet, and previous simulations of lateral inhibition in pancreas development excluded by design the possibility of oscillations. To explore this possibility, we developed a dynamic model of a growing duct that results in an oscillatory phase before the determination of endocrine progenitors by lateral inhibition. The basic network (D/N + Hes1 + Neurog3) shows scattered, stable Neurog3 expression after displaying transient expression. Furthermore, we included the Hes1 negative feedback as previously discussed in neurogenesis and show the consequences for Neurog3 expression in pancreatic duct development. Interestingly, a weakened HES1 action on the Hes1 promoter allows the coexistence of stable patterning and oscillations. In conclusion, cycling gene expression and lateral inhibition are not mutually exclusive. In this way, we argue for a unified mode of D/N mediated lateral inhibition in neurogenic and pancreatic progenitor specification.


Subject(s)
Models, Biological , Neurogenesis , Pancreas/growth & development , Receptors, Notch/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/physiology , Body Patterning , Cell Lineage , Endocrine System/cytology , Feedback, Physiological , Gene Expression Regulation, Developmental , Mice , Nerve Tissue Proteins/physiology , Oscillometry , Pancreas/innervation , Transcription Factor HES-1/physiology
9.
J Mol Endocrinol ; 56(2): 77-90, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26576643

ABSTRACT

The generation of therapeutic ß-cells from human pluripotent stem cells relies on the identification of growth factors that faithfully mimic pancreatic ß-cell development in vitro. In this context, the aim of the study was to determine the expression and function of the glial cell line derived neurotrophic factor receptor alpha 3 (GFRα3) and its ligand artemin (Artn) in islet cell development and function. GFRα3 and Artn expression were characterized by in situ hybridization, immunochemistry, and qRT-PCR. We used GFRα3-deficient mice to study GFRα3 function and generated transgenic mice overexpressing Artn in the embryonic pancreas to study Artn function. We found that GFRα3 is expressed at the surface of a subset of Ngn3-positive endocrine progenitors as well as of embryonic α- and ß-cells, while Artn is found in the pancreatic mesenchyme. Adult ß-cells lack GFRα3 but α-cells express the receptor. GFRα3 was also found in parasympathetic and sympathetic intra-islet neurons as well as in glial cells in the embryonic and adult pancreas. The loss of GFRα3 or overexpression of Artn has no impact on Ngn3 and islet cell formation and maintenance in the embryo. Islet organization and innervation as well as glucose homeostasis is normal in GFRα3-deficient mice suggesting functional redundancy.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Islets of Langerhans/metabolism , Animals , Gene Expression , Glucose/metabolism , Homeostasis , Islets of Langerhans/cytology , Islets of Langerhans/growth & development , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pancreas/cytology , Pancreas/growth & development , Pancreas/metabolism
10.
Development ; 141(15): 2939-49, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25053427

ABSTRACT

Insulinoma associated 1 (Insm1) plays an important role in regulating the development of cells in the central and peripheral nervous systems, olfactory epithelium and endocrine pancreas. To better define the role of Insm1 in pancreatic endocrine cell development we generated mice with an Insm1(GFPCre) reporter allele and used them to study Insm1-expressing and null populations. Endocrine progenitor cells lacking Insm1 were less differentiated and exhibited broad defects in hormone production, cell proliferation and cell migration. Embryos lacking Insm1 contained greater amounts of a non-coding Neurog3 mRNA splice variant and had fewer Neurog3/Insm1 co-expressing progenitor cells, suggesting that Insm1 positively regulates Neurog3. Moreover, endocrine progenitor cells that express either high or low levels of Pdx1, and thus may be biased towards the formation of specific cell lineages, exhibited cell type-specific differences in the genes regulated by Insm1. Analysis of the function of Ripply3, an Insm1-regulated gene enriched in the Pdx1-high cell population, revealed that it negatively regulates the proliferation of early endocrine cells. Taken together, these findings indicate that in developing pancreatic endocrine cells Insm1 promotes the transition from a ductal progenitor to a committed endocrine cell by repressing a progenitor cell program and activating genes essential for RNA splicing, cell migration, controlled cellular proliferation, vasculogenesis, extracellular matrix and hormone secretion.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA-Binding Proteins/physiology , Endocrine Cells/cytology , Gene Expression Regulation, Developmental , Nerve Tissue Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/physiology , Alleles , Alternative Splicing , Animals , Cell Differentiation , Cell Lineage , Cell Movement , Cell Proliferation , Cell Separation , Extracellular Matrix/metabolism , Flow Cytometry , Gene Regulatory Networks , Genes, Reporter , Green Fluorescent Proteins/metabolism , Mice , Mice, Knockout , Pancreas/embryology , RNA/metabolism , RNA Splicing , Stem Cells/cytology , Time Factors , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL