Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Annu Rev Immunol ; 37: 497-519, 2019 04 26.
Article in English | MEDLINE | ID: mdl-31026413

ABSTRACT

During development innate lymphoid cells and specialized lymphocyte subsets colonize peripheral tissues, where they contribute to organogenesis and later constitute the first line of protection while maintaining tissue homeostasis. A few of these subsets are produced only during embryonic development and remain in the tissues throughout life. They are generated through a unique developmental program initiated in lympho-myeloid-primed progenitors, which lose myeloid and B cell potential. They either differentiate into innate lymphoid cells or migrate to the thymus to give rise to embryonic T cell receptor-invariant T cells. At later developmental stages, adaptive T lymphocytes are derived from lympho-myeloid progenitors that colonize the thymus, while lymphoid progenitors become specialized in the production of B cells. This sequence of events highlights the requirement for stratification in the establishment of immune functions that determine efficient seeding of peripheral tissues by a limited number of cells.


Subject(s)
B-Lymphocytes/immunology , Lymphocytes/physiology , Lymphoid Progenitor Cells/physiology , Natural Killer T-Cells/immunology , Thymus Gland/immunology , Animals , Cell Differentiation , Cell Lineage , Cellular Microenvironment , Cytokines/metabolism , Humans , Immunity, Innate , Lymphocyte Activation , Paracrine Communication , Transcriptome
2.
Development ; 145(16)2018 08 15.
Article in English | MEDLINE | ID: mdl-30042179

ABSTRACT

To decipher the populations of cells present in the human fetal pancreas and their lineage relationships, we developed strategies to isolate pancreatic progenitors, endocrine progenitors and endocrine cells. Transcriptome analysis of the individual populations revealed a large degree of conservation among vertebrates in the drivers of gene expression changes that occur at different steps of differentiation, although notably, sometimes, different members of the same gene family are expressed. The transcriptome analysis establishes a resource to identify novel genes and pathways involved in human pancreas development. Single-cell profiling further captured intermediate stages of differentiation and enabled us to decipher the sequence of transcriptional events occurring during human endocrine differentiation. Furthermore, we evaluate how well individual pancreatic cells derived in vitro from human pluripotent stem cells mirror the natural process occurring in human fetuses. This comparison uncovers a few differences at the progenitor steps, a convergence at the steps of endocrine induction, and the current inability to fully resolve endocrine cell subtypes in vitro.


Subject(s)
Fetus/embryology , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation, Developmental/physiology , Pancreas/embryology , Transcription, Genetic/physiology , Fetus/cytology , Humans , Pancreas/cytology , Pluripotent Stem Cells/metabolism
3.
Nat Immunol ; 18(10): 1139-1149, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28825702

ABSTRACT

The molecular events that initiate lymphoid-lineage specification remain unidentified because the stages of differentiation during which lineage commitment occurs are difficult to characterize. We isolated fetal liver progenitor cells undergoing restriction of their differentiation potential toward the T cell-innate lymphoid cell lineage or the B cell lineage. Transcripts that defined the molecular signatures of these two subsets were sequentially upregulated in lympho-myeloid precursor cells and in common lymphoid progenitor cells, respectively, and this preceded lineage restriction; this indicates that T cell-versus-B cell commitment is not a binary fate 'decision'. The T cell-bias and B cell-bias transcriptional programs were frequently co-expressed in common lymphoid progenitor cells and were segregated in subsets biased toward T cell differentiation or B cell differentiation, after interleukin 7 (IL-7) signaling that controlled the number of progenitor cells engaging in T cell differentiation versus B cell differentiation.


Subject(s)
B-Lymphocytes/cytology , Cell Lineage , Liver/cytology , Lymphopoiesis , T-Lymphocytes/cytology , Animals , B-Lymphocytes/metabolism , Biomarkers , Cell Differentiation/genetics , Cell Lineage/genetics , Cluster Analysis , Fetus , Gene Expression Profiling , Gene Expression Regulation, Developmental , Immunophenotyping , Interleukin-7/metabolism , Liver/embryology , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Lymphopoiesis/genetics , Mice , Mice, Transgenic , Signal Transduction , T-Lymphocytes/metabolism , Transcriptome
4.
Elife ; 62017 07 21.
Article in English | MEDLINE | ID: mdl-28731406

ABSTRACT

Information remains scarce on human development compared to animal models. Here, we reconstructed human fetal pancreatic differentiation using cell surface markers. We demonstrate that at 7weeks of development, the glycoprotein 2 (GP2) marks a multipotent cell population that will differentiate into the acinar, ductal or endocrine lineages. Development towards the acinar lineage is paralleled by an increase in GP2 expression. Conversely, a subset of the GP2+ population undergoes endocrine differentiation by down-regulating GP2 and CD142 and turning on NEUROG3, a marker of endocrine differentiation. Endocrine maturation progresses by up-regulating SUSD2 and lowering ECAD levels. Finally, in vitro differentiation of pancreatic endocrine cells derived from human pluripotent stem cells mimics key in vivo events. Our work paves the way to extend our understanding of the origin of mature human pancreatic cell types and how such lineage decisions are regulated.


Subject(s)
Biomarkers/metabolism , Cell Differentiation , Cell Lineage , Fetus/cytology , Gene Expression Regulation, Developmental , Pancreas/cytology , Acinar Cells/cytology , Acinar Cells/metabolism , Cells, Cultured , Endocrine Cells/cytology , Endocrine Cells/metabolism , Female , Fetus/metabolism , Humans , Pancreas/metabolism , Pancreatic Ducts/cytology , Pancreatic Ducts/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Transcriptome
5.
Cell Rep ; 19(1): 36-49, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28380361

ABSTRACT

Stem cell-based therapy for type 1 diabetes would benefit from implementation of a cell purification step at the pancreatic endoderm stage. This would increase the safety of the final cell product, allow the establishment of an intermediate-stage stem cell bank, and provide a means for upscaling ß cell manufacturing. Comparative gene expression analysis revealed glycoprotein 2 (GP2) as a specific cell surface marker for isolating pancreatic endoderm cells (PECs) from differentiated hESCs and human fetal pancreas. Isolated GP2+ PECs efficiently differentiated into glucose responsive insulin-producing cells in vitro. We found that in vitro PEC proliferation declines due to enhanced expression of the cyclin-dependent kinase (CDK) inhibitors CDKN1A and CDKN2A. However, we identified a time window when reducing CDKN1A or CDKN2A expression increased proliferation and yield of GP2+ PECs. Altogether, our results contribute tools and concepts toward the isolation and use of PECs as a source for the safe production of hPSC-derived ß cells.


Subject(s)
Embryonic Stem Cells/metabolism , GPI-Linked Proteins/metabolism , Glucose/metabolism , Insulin-Secreting Cells/cytology , Biomarkers/metabolism , Cell Differentiation , Cell Proliferation , Cell Separation/methods , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinase Inhibitor p18/genetics , Cyclin-Dependent Kinase Inhibitor p18/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Diabetes Mellitus, Type 1/therapy , Embryonic Stem Cells/cytology , Endoderm/cytology , GPI-Linked Proteins/genetics , Gene Expression Regulation , Genome-Wide Association Study , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism
6.
Hum Mol Genet ; 26(3): 599-610, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28025328

ABSTRACT

Congenital hypothyroidism is the most common neonatal endocrine disorder and is primarily caused by developmental abnormalities otherwise known as thyroid dysgenesis (TD). We performed whole exome sequencing (WES) in a consanguineous family with TD and subsequently sequenced a cohort of 134 probands with TD to identify genetic factors predisposing to the disease. We identified the novel missense mutations p.S148F, p.R114Q and p.L177W in the BOREALIN gene in TD-affected families. Borealin is a major component of the Chromosomal Passenger Complex (CPC) with well-known functions in mitosis. Further analysis of the missense mutations showed no apparent effects on mitosis. In contrast, expression of the mutants in human thyrocytes resulted in defects in adhesion and migration with corresponding changes in gene expression suggesting others functions for this mitotic protein. These results were well correlated with the same gene expression pattern analysed in the thyroid tissue of the patient with BOREALIN-p.R114W. These studies open new avenues in the genetics of TD in humans.


Subject(s)
Cell Cycle Proteins/genetics , Genetic Predisposition to Disease , Mutation, Missense/genetics , Thyroid Dysgenesis/genetics , Cell Cycle Proteins/biosynthesis , Cell Movement/genetics , Exome/genetics , Female , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans , Mitosis/genetics , Pedigree , Thyroid Dysgenesis/pathology
7.
Sci Rep ; 6: 25765, 2016 05 11.
Article in English | MEDLINE | ID: mdl-27166427

ABSTRACT

Acute or chronic metabolic complications such as diabetic ketoacidosis are often associated with extracellular acidification and pancreatic ß-cell dysfunction. However, the mechanisms by which human ß-cells sense and respond to acidic pH remain elusive. In this study, using the recently developed human ß-cell line EndoC-ßH2, we demonstrate that ß-cells respond to extracellular acidification through GPR68, which is the predominant proton sensing receptor of human ß-cells. Using gain- and loss-of-function studies, we provide evidence that the ß-cell enriched transcription factor RFX6 is a major regulator of GPR68. Further, we show that acidic pH stimulates the production and secretion of the chemokine IL-8 by ß-cells through NF-кB activation. Blocking of GPR68 or NF-кB activity severely attenuated acidification induced IL-8 production. Thus, we provide mechanistic insights into GPR68 mediated ß-cell response to acidic microenvironment, which could be a new target to protect ß-cell against acidosis induced inflammation.


Subject(s)
Acids/metabolism , Extracellular Space/chemistry , Insulin-Secreting Cells/metabolism , Interleukin-8/biosynthesis , Receptors, G-Protein-Coupled/metabolism , Cell Line , Cyclic AMP/biosynthesis , Humans , Hydrogen-Ion Concentration , Inflammation Mediators/metabolism , Inositol Phosphates/metabolism , NF-kappa B/metabolism , Neutrophils/metabolism , Protons , Regulatory Factor X Transcription Factors/metabolism
8.
J Vis Exp ; (100): e52795, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26131754

ABSTRACT

Characterizing thymic settling progenitors is important to understand the pre-thymic stages of T cell development, essential to devise strategies for T cell replacement in lymphopenic patients. We studied thymic settling progenitors from murine embryonic day 13 and 18 thymi by two complementary in vitro and in vivo techniques, both based on the "hanging drop" method. This method allowed colonizing irradiated fetal thymic lobes with E13 and/or E18 thymic progenitors distinguished by CD45 allotypic markers and thus following their progeny. Colonization with mixed populations allows analyzing cell autonomous differences in biologic properties of the progenitors while colonization with either population removes possible competitive selective pressures. The colonized thymic lobes can also be grafted in immunodeficient male recipient mice allowing the analysis of the mature T cell progeny in vivo, such as population dynamics of the peripheral immune system and colonization of different tissues and organs. Fetal thymic organ cultures revealed that E13 progenitors developed rapidly into all mature CD3(+) cells and gave rise to the canonical γδ T cell subset, known as dendritic epithelial T cells. In comparison, E18 progenitors have a delayed differentiation and were unable to generate dendritic epithelial T cells. The monitoring of peripheral blood of thymus-grafted CD3(-/-) mice further showed that E18 thymic settling progenitors generate, with time, larger numbers of mature T cells than their E13 counterparts, a feature that could not be appreciated in the short term fetal thymic organ cultures.


Subject(s)
Embryonic Stem Cells/cytology , Organ Culture Techniques/methods , Thymus Gland/cytology , Animals , Embryonic Stem Cells/metabolism , Female , Flow Cytometry , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Pregnancy , Receptors, Antigen, T-Cell/biosynthesis , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Thymus Gland/embryology , Thymus Gland/metabolism
9.
Diabetologia ; 57(11): 2348-56, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25186293

ABSTRACT

AIM/HYPOTHESIS: Different studies have linked hypoxia to embryonic development. Specifically, when embryonic pancreases are cultured ex vivo under hypoxic conditions (3% O2), beta cell development is impaired. Different cellular signalling pathways are involved in adaptation to hypoxia, including the ubiquitous hypoxia-inducible-factor 1-α (HIF1-α) pathway. We aimed to analyse the effects of HIF1-α stabilisation on fetal pancreas development in vivo. METHODS: We deleted the Vhl gene, which encodes von Hippel-Lindau protein (pVHL), a factor necessary for HIF1-α degradation, by crossing Vhl-floxed mice with Sox9-Cre mice. RESULTS: HIF1-α was stabilised in pancreatic progenitor cells in which the HIF pathway was induced. The number of neurogenin-3 (NGN3)-expressing cells was reduced and consequently endocrine development was altered in Vhl knockout pancreases. HIF1-α stabilisation induced Vegfa upregulation, leading to increased vascularisation. To investigate the impact of increased vascularisation on NGN3 expression, we used a bioassay in which Vhl mutant pancreases were cultured with or without vascular endothelial growth factor (VEGF) receptor 2 (VEGF-R2) inhibitors (e.g. Ki8751). Ex vivo analysis showed that Vhl knockout pancreases developed fewer NGN3-positive cells compared with controls. Interestingly, this effect was blocked when vascularisation was inhibited in the presence of VEGF-R2 inhibitors. CONCLUSIONS/INTERPRETATION: Our data demonstrate that HIF1-α negatively controls beta cell differentiation in vivo by regulating NGN3 expression, and that this effect is mediated by signals from blood vessels.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Pancreas/cytology , Vascular Endothelial Growth Factor A/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Female , Flow Cytometry , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pancreas/embryology , Pregnancy , Vascular Endothelial Growth Factor A/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics
10.
Nat Immunol ; 15(1): 27-35, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24317038

ABSTRACT

The generation of T cells depends on the migration of hematopoietic progenitor cells to the thymus throughout life. The identity of the thymus-settling progenitor cells has been a matter of considerable debate. Here we found that thymopoiesis was initiated by a first wave of T cell lineage-restricted progenitor cells with limited capacity for population expansion but accelerated differentiation into mature T cells. They gave rise to αß and γδ T cells that constituted Vγ3(+) dendritic epithelial T cells. Thymopoiesis was subsequently maintained by less-differentiated progenitor cells that retained the potential to develop into B cells and myeloid cells. In that second wave, which started before birth, progenitor cells had high proliferative capacity but delayed differentiation capacity and no longer gave rise to embryonic γδ T cells. Our work reconciles conflicting hypotheses on the nature of thymus-settling progenitor cells.


Subject(s)
Cell Differentiation/genetics , Hematopoietic Stem Cells/metabolism , T-Lymphocytes/metabolism , Thymus Gland/metabolism , Animals , Animals, Newborn , Cell Line , Cell Lineage/genetics , Cells, Cultured , Flow Cytometry , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/cytology , Interleukin-7 Receptor alpha Subunit/genetics , Interleukin-7 Receptor alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/cytology , Thymocytes/cytology , Thymocytes/metabolism , Thymus Gland/cytology , Thymus Gland/embryology , Time Factors , Transcriptome , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
11.
J Immunol ; 189(8): 3822-30, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22972921

ABSTRACT

Lineage commitment is regulated during hematopoiesis, with stepwise loss of differentiation potential ultimately resulting in lineage commitment. In this study we describe a novel population of B/NK bipotent precursors among common lymphoid progenitors in the fetal liver and the bone marrow. The absence of T cell precursor potential, both in vivo and in vitro, is due to low Notch1 expression and secondary to inhibition of E2A activity by members of the inhibitor of DNA binding (Id) protein family. Our results demonstrate a new, Id protein-dependent, molecular mechanism of Notch1 repression, operative in both fetal and adult common lymphoid progenitors, where T cell potential is selectively inhibited without affecting either the B or NK programs. This study identifies Id proteins as negative regulators of T cell specification, before B and NK commitment, and provides important insights into the transcriptional networks orchestrating hematopoiesis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Cell Differentiation/immunology , DNA-Binding Proteins/antagonists & inhibitors , Down-Regulation/immunology , Receptor, Notch1/antagonists & inhibitors , Stem Cells/immunology , T-Lymphocyte Subsets/immunology , fms-Like Tyrosine Kinase 3/deficiency , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Lineage/immunology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Down-Regulation/genetics , Inhibitor of Differentiation Protein 2/deficiency , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Proteins/deficiency , Inhibitor of Differentiation Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Multigene Family/genetics , Multigene Family/immunology , Receptor, Notch1/biosynthesis , Receptor, Notch1/genetics , Stem Cells/cytology , Stem Cells/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , fms-Like Tyrosine Kinase 3/biosynthesis , fms-Like Tyrosine Kinase 3/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...