Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cell Res ; 31(8): 886-903, 2021 08.
Article in English | MEDLINE | ID: mdl-33692492

ABSTRACT

The pancreatic islet contains multiple hormone+ endocrine lineages (α, ß, δ, PP and ε cells), but the developmental processes that underlie endocrinogenesis are poorly understood. Here, we generated novel mouse lines and combined them with various genetic tools to enrich all types of hormone+ cells for well-based deep single-cell RNA sequencing (scRNA-seq), and gene coexpression networks were extracted from the generated data for the optimization of high-throughput droplet-based scRNA-seq analyses. These analyses defined an entire endocrinogenesis pathway in which different states of endocrine progenitor (EP) cells sequentially differentiate into specific endocrine lineages in mice. Subpopulations of the EP cells at the final stage (EP4early and EP4late) show different potentials for distinct endocrine lineages. ε cells and an intermediate cell population were identified as distinct progenitors that independently generate both α and PP cells. Single-cell analyses were also performed to delineate the human pancreatic endocrinogenesis process. Although the developmental trajectory of pancreatic lineages is generally conserved between humans and mice, clear interspecies differences, including differences in the proportions of cell types and the regulatory networks associated with the differentiation of specific lineages, have been detected. Our findings support a model in which sequential transient progenitor cell states determine the differentiation of multiple cell lineages and provide a blueprint for directing the generation of pancreatic islets in vitro.


Subject(s)
Islets of Langerhans , Pancreas , Animals , Cell Differentiation , Cell Lineage , Humans , Mice , Stem Cells
2.
Cell Res ; 31(3): 326-344, 2021 03.
Article in English | MEDLINE | ID: mdl-33106598

ABSTRACT

Defining the precise regionalization of specified definitive endoderm progenitors is critical for understanding the mechanisms underlying the generation and regeneration of respiratory and digestive organs, yet the patterning of endoderm progenitors remains unresolved, particularly in humans. We performed single-cell RNA sequencing on endoderm cells during the early somitogenesis stages in mice and humans. We developed molecular criteria to define four major endoderm regions (foregut, lip of anterior intestinal portal, midgut, and hindgut) and their developmental pathways. We identified the cell subpopulations in each region and their spatial distributions and characterized key molecular features along the body axes. Dorsal and ventral pancreatic progenitors appear to originate from the midgut population and follow distinct pathways to develop into an identical cell type. Finally, we described the generally conserved endoderm patterning in humans and clear differences in dorsal cell distribution between species. Our study comprehensively defines single-cell endoderm patterning and provides novel insights into the spatiotemporal process that drives establishment of early endoderm domains.


Subject(s)
Body Patterning/genetics , Embryo, Mammalian/cytology , Endoderm/cytology , Intestines/cytology , Lip/cytology , Animals , Cells, Cultured , Female , Gene Expression , Gene Expression Regulation, Developmental , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , RNA-Seq/methods , Single-Cell Analysis/methods
3.
Cell Rep ; 33(10): 108455, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33296648

ABSTRACT

The ever-increasing therapeutic and pharmaceutical demand for liver cells calls for systems that enable mass production of hepatic cells. Here we describe a large-scale suspension system that uses human endoderm stem cells (hEnSCs) as precursors to generate functional and transplantable hepatocytes (E-heps) or cholangiocytes (E-chos). hEnSC-derived hepatic populations are characterized by single-cell transcriptomic analyses and compared with hESC-derived counterparts, in-vitro-maintained or -expanded primary hepatocytes and adult cells, which reveals that hepatic differentiation of hEnSCs recapitulates in vivo development and that the heterogeneities of the resultant populations can be manipulated by regulating the EGF and MAPK signaling pathways. Functional assessments demonstrate that E-heps and E-chos possess properties comparable with adult counterparts and that, when transplanted intraperitoneally, encapsulated E-heps were able to rescue rats with acute liver failure. Our study lays the foundation for cell-based therapeutic agents and in vitro applications for liver diseases.


Subject(s)
Cell Culture Techniques/methods , Endoderm/cytology , Hepatocytes/cytology , Human Embryonic Stem Cells/cytology , Bile Ducts/cytology , Bile Ducts/metabolism , Cell Differentiation/physiology , Endoderm/metabolism , Endoderm/transplantation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Hepatocytes/metabolism , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/transplantation , Humans , Liver/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/transplantation
4.
Mol Metab ; 37: 100982, 2020 07.
Article in English | MEDLINE | ID: mdl-32247924

ABSTRACT

OBJECTIVES: The streptozotocin (STZ) model is widely used in diabetes research. However, the cellular and molecular states of pancreatic endocrine cells in this model remain unclear. This study explored the molecular characteristics of islet cells treated with STZ and re-evaluated ß-cell dysfunction and regeneration in the STZ model. METHODS: We performed single-cell RNA sequencing of pancreatic endocrine cells from STZ-treated mice. High-quality sequencing data from 2,999 cells were used to identify clusters via Louvain clustering analysis. Principal component analysis (PCA), t-distributed stochastic neighbor embedding (t-SNE), uniform manifold approximation and projection (UMAP), force-directed layout (FDL), and differential expression analysis were used to define the heterogeneity and transcriptomic changes in islet cells. In addition, qPCR and immunofluorescence staining were used to confirm findings from the sequencing data. RESULTS: Untreated ß-cells were divided into two populations at the transcriptomic level, a large high-Glut2 expression (Glut2high) population and a small low-Glut2 expression (Glut2low) population. At the transcriptomic level, Glut2low ß-cells in adult mice did not represent a developmentally immature state, although a fraction of genes associated with ß-cell maturation and function were downregulated in Glut2low cells. After a single high-dose STZ treatment, most Glut2high cells were killed, but Glut2low cells survived and over time changed to a distinct cell state. We did not observe conversion of Glut2low to Glut2high ß-cells up to 9 months after STZ treatment. In addition, we did not detect transcriptomic changes in the non-ß endocrine cells or a direct trans-differentiation pathway from the α-cell lineage to the ß-cell lineage in the STZ model. CONCLUSIONS: We identified the heterogeneity of ß-cells in both physiological and pathological conditions. However, we did not observe conversion of Glut2low to Glut2high ß-cells, transcriptomic changes in the non-ß endocrine cells, or direct trans-differentiation from the α-cell lineage to the ß-cell lineage in the STZ model. Our results clearly define the states of islet cells treated with STZ and allow us to re-evaluate the STZ model widely used in diabetes studies.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Insulin-Secreting Cells/physiology , Islets of Langerhans/metabolism , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/physiopathology , Gene Expression/genetics , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , Glucagon-Secreting Cells/metabolism , Glucose/metabolism , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/metabolism , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/physiology , Male , Mice , Mice, Transgenic , Single-Cell Analysis/methods , Streptozocin/pharmacology , Transcriptome/genetics
5.
EMBO J ; 38(8)2019 04 15.
Article in English | MEDLINE | ID: mdl-30737258

ABSTRACT

The generation of terminally differentiated cell lineages during organogenesis requires multiple, coordinated cell fate choice steps. However, this process has not been clearly delineated, especially in complex solid organs such as the pancreas. Here, we performed single-cell RNA-sequencing in pancreatic cells sorted from multiple genetically modified reporter mouse strains at embryonic stages E9.5-E17.5. We deciphered the developmental trajectories and regulatory strategies of the exocrine and endocrine pancreatic lineages as well as intermediate progenitor populations along the developmental pathways. Notably, we discovered previously undefined programs representing the earliest events in islet α- and ß-cell lineage allocation as well as the developmental pathway of the "first wave" of α-cell generation. Furthermore, we demonstrated that repressing ERK pathway activity is essential for inducing both α- and ß-lineage differentiation. This study provides key insights into the regulatory mechanisms underlying cell fate choice and stepwise cell fate commitment and can be used as a resource to guide the induction of functional islet lineage cells from stem cells in vitro.


Subject(s)
Cell Differentiation , Cell Lineage , Gene Expression Regulation, Developmental , Organogenesis , Pancreas/metabolism , Single-Cell Analysis/methods , Animals , Female , Mice , Pancreas/cytology
6.
J Vis Exp ; (139)2018 09 30.
Article in English | MEDLINE | ID: mdl-30320740

ABSTRACT

Pancreatic endocrine cells, which are clustered in islets, regulate blood glucose stability and energy metabolism. The distinct cell types in islets, including insulin-secreting ß cells, are differentiated from common endocrine progenitors during the embryonic stage. Immature endocrine cells expand via cell proliferation and mature during a long postnatal developmental period. However, the mechanisms underlying these processes are not clearly defined. Single-cell RNA-sequencing is a promising approach for the characterization of distinct cell populations and tracing cell lineage differentiation pathways. Here, we describe a method for the single-cell RNA-sequencing of isolated pancreatic ß cells from embryonic, neonatal and postnatal pancreases.


Subject(s)
Endocrine Cells/metabolism , Gene Expression Profiling/methods , Insulin-Secreting Cells/cytology , Single-Cell Analysis/methods , Animals , Cell Differentiation , Cell Proliferation , Glucose/metabolism , Insulin/metabolism , Mice , Sequence Analysis, RNA
7.
EMBO Rep ; 19(10)2018 10.
Article in English | MEDLINE | ID: mdl-30065074

ABSTRACT

The pancreas of vertebrates is separately derived from both the dorsal and ventral endodermal domains. However, the difference between these two programs has been unclear. Here, using a pancreatic determination gene, Pdx1, driven GFP transgenic mouse strain, we identified Pdx1-GFP highly expressing cells (Pdx1high) and Pdx1-GFP lowly expressing cells (Pdx1low) in both embryonic dorsal Pdx1-expressing region (DPR) and ventral Pdx1-expressing region (VPR). We analyzed the transcriptomes of single Pdx1low and Pdx1high cells from the DPR and VPR. In the VPR, Pdx1low cells have an intermediate progenitor identity and can generate hepatoblasts, extrahepatobiliary cells, and Pdx1high pancreatic progenitor cells. In the DPR, Pdx1high cells are directly specified as pancreatic progenitors, whereas Pdx1low cells are precocious endocrine cells. Therefore, our study defines distinct road maps for dorsal and ventral pancreatic progenitor specification. The findings provide guidance for optimization of current ß-cell induction protocols by following the in vivo dorsal pancreatic specification program.


Subject(s)
Homeodomain Proteins/genetics , Pancreas/growth & development , Stem Cells/metabolism , Trans-Activators/genetics , Transcriptome/genetics , Animals , Cell Lineage/genetics , Gene Expression Regulation, Developmental , Insulin-Secreting Cells/metabolism , Mice , Mice, Transgenic/genetics , Pancreas/embryology , Pancreas/metabolism , Single-Cell Analysis
8.
Development ; 145(6)2018 03 20.
Article in English | MEDLINE | ID: mdl-29559448

ABSTRACT

Pancreatic endocrine lineages are derived from pancreatic progenitors that undergo a cell fate transition requiring a switch from low to high Ngn3 expression. However, the underlying chromatin regulatory mechanisms are unclear. Here, we performed epigenomic analysis of gene regulatory loci featuring histone marks in cells with low or high level of Ngn3 expression. In combination with transcriptomic analysis, we discovered that in Ngn3-high cells, the removal of H3K27me3 was associated with the activation of key transcription factors and the establishment of primed and active enhancers. Deletion of Jmjd3, a histone demethylase for H3K27me3, at the pancreatic progenitor stage impaired the efficiency of endocrine cell fate transition and thereafter islet formation. Curiously, single-cell RNA-seq revealed that the transcriptome and developmental pathway of Ngn3-high cells were not affected by the deletion of Jmjd3 Our study indicates sequential chromatin events and identifies a crucial role for Jmjd3 in regulating the efficiency of the transition from Ngn3-low to Ngn3-high cells.


Subject(s)
Chromatin/metabolism , Endocrine Cells/metabolism , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Pancreas/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Cell Differentiation , Endocrine Cells/cytology , Epigenomics , Fluorescent Antibody Technique , Mice , Nerve Tissue Proteins/metabolism , Polymerase Chain Reaction , Transcriptome
10.
Hepatology ; 66(5): 1387-1401, 2017 11.
Article in English | MEDLINE | ID: mdl-28681484

ABSTRACT

How bipotential hepatoblasts differentiate into hepatocytes and cholangiocytes remains unclear. Here, using single-cell transcriptomic analysis of hepatoblasts, hepatocytes, and cholangiocytes sorted from embryonic day 10.5 (E10.5) to E17.5 mouse embryos, we found that hepatoblast-to-hepatocyte differentiation occurred gradually and followed a linear default pathway. As more cells became fully differentiated hepatocytes, the number of proliferating cells decreased. Surprisingly, proliferating and quiescent hepatoblasts exhibited homogeneous differentiation states at a given developmental stage. This unique feature enabled us to combine single-cell and bulk-cell analyses to define the precise timing of the hepatoblast-to-hepatocyte transition, which occurs between E13.5 and E15.5. In contrast to hepatocyte development at almost all levels, hepatoblast-to-cholangiocyte differentiation underwent a sharp detour from the default pathway. New cholangiocyte generation occurred continuously between E11.5 and E14.5, but their maturation states at a given developmental stage were heterogeneous. Even more surprising, the number of proliferating cells increased as more progenitor cells differentiated into mature cholangiocytes. Based on an observation from the single-cell analysis, we also discovered that the protein kinase C/mitogen-activated protein kinase signaling pathway promoted cholangiocyte maturation. CONCLUSION: Our studies have defined distinct pathways for hepatocyte and cholangiocyte development in vivo, which are critically important for understanding basic liver biology and developing effective strategies to induce stem cells to differentiate toward specific hepatic cell fates in vitro. (Hepatology 2017;66:1387-1401).


Subject(s)
Cell Differentiation , Hepatocytes/cytology , Liver/embryology , Animals , Hepatocytes/metabolism , Liver/cytology , MAP Kinase Signaling System , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Single-Cell Analysis , Transcriptome
11.
Cell Metab ; 25(5): 1194-1205.e4, 2017 May 02.
Article in English | MEDLINE | ID: mdl-28467935

ABSTRACT

Pancreatic ß and α cells play essential roles in maintaining glucose homeostasis. However, the mechanisms by which these distinct cell populations are generated, expand, and mature during pancreas development remain unclear. In this study, we addressed this critical question by performing a single-cell transcriptomic analysis of mouse ß and α cells sorted from fetal to adult stages. We discovered that ß and α cells use different regulatory strategies for their maturation and that cell proliferation peaks at different developmental times. However, the quiescent and proliferative cells in both the ß lineage and α lineage are synchronous in their maturation states. The heterogeneity of juvenile ß cells reflects distinct cell-cycling phases, origins, and maturation states, whereas adult ß cells are relatively homogeneous at the transcriptomic level. These analyses provide not only a high-resolution roadmap for islet lineage development but also insights into the mechanisms of cellular heterogeneity, cell number expansion, and maturation of both ß and α cells.


Subject(s)
Glucagon-Secreting Cells/cytology , Insulin-Secreting Cells/cytology , Animals , Cell Cycle , Cell Differentiation , Cell Proliferation , Cells, Cultured , Female , Glucagon-Secreting Cells/metabolism , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Signal Transduction , Single-Cell Analysis , Transcriptome
12.
Genome Biol ; 16: 277, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26653294

ABSTRACT

BACKGROUND: Patterns of genetic and genomic variance are informative in inferring population history for human, model species and endangered populations. RESULTS: Here the genome sequence of wild-born African cheetahs reveals extreme genomic depletion in SNV incidence, SNV density, SNVs of coding genes, MHC class I and II genes, and mitochondrial DNA SNVs. Cheetah genomes are on average 95 % homozygous compared to the genomes of the outbred domestic cat (24.08 % homozygous), Virunga Mountain Gorilla (78.12 %), inbred Abyssinian cat (62.63 %), Tasmanian devil, domestic dog and other mammalian species. Demographic estimators impute two ancestral population bottlenecks: one >100,000 years ago coincident with cheetah migrations out of the Americas and into Eurasia and Africa, and a second 11,084-12,589 years ago in Africa coincident with late Pleistocene large mammal extinctions. MHC class I gene loss and dramatic reduction in functional diversity of MHC genes would explain why cheetahs ablate skin graft rejection among unrelated individuals. Significant excess of non-synonymous mutations in AKAP4 (p<0.02), a gene mediating spermatozoon development, indicates cheetah fixation of five function-damaging amino acid variants distinct from AKAP4 homologues of other Felidae or mammals; AKAP4 dysfunction may cause the cheetah's extremely high (>80 %) pleiomorphic sperm. CONCLUSIONS: The study provides an unprecedented genomic perspective for the rare cheetah, with potential relevance to the species' natural history, physiological adaptations and unique reproductive disposition.


Subject(s)
Acinonyx/genetics , Genome , Animals , Cats , Dogs , Genetic Variation , Genomics , Male , Multigene Family
SELECTION OF CITATIONS
SEARCH DETAIL
...