Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Nat Immunol ; 20(11): 1456-1468, 2019 11.
Article in English | MEDLINE | ID: mdl-31636466

ABSTRACT

T cell specification and commitment require Notch signaling. Although the requirement for Notch signaling during intrathymic T cell development is known, it is still unclear whether the onset of T cell priming can occur in a prethymic niche and whether RBPJ-dependent Notch signaling has a role during this event. Here, we established an Rbpj-inducible system that allowed temporal and tissue-specific control of the responsiveness to Notch in all hematopoietic cells. Using this system, we found that Notch signaling was required before the early T cell progenitor stage in the thymus. Lymphoid-primed multipotent progenitors in the bone marrow underwent Notch signaling with Rbpj induction, which inhibited development towards the myeloid lineage in thymus-seeding progenitors. Thus, our results indicated that the onset of T cell differentiation occurred in a prethymic setting, and that Notch played an important role during this event.


Subject(s)
Cell Differentiation/immunology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Precursor Cells, T-Lymphoid/physiology , Receptors, Notch/metabolism , T-Lymphocyte Subsets/immunology , Animals , Cell Lineage/immunology , Cell Separation , Female , Flow Cytometry , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Male , Mice , Mice, Transgenic , Primary Cell Culture , Signal Transduction/immunology , T-Lymphocyte Subsets/metabolism , Thymus Gland/cytology , Thymus Gland/immunology
2.
Methods Mol Biol ; 1884: 87-117, 2019.
Article in English | MEDLINE | ID: mdl-30465197

ABSTRACT

This chapter will describe the current methodologies to isolate and expand NK cells from Peripheral Blood (PB) or tissues for "in vitro" studies, including NK cell antitumor immune function. In addition, methods to induce NK cell maturation, differentiation, and expansion from CD34+ precursors will also be described. Finally, it will also be treated the topical issue of the characterization of new functionally and phenotypically defined NK cell subsets.


Subject(s)
Cell Separation/methods , Flow Cytometry/methods , Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , Neoplasms/immunology , Antigens, CD34/metabolism , Cell Differentiation/immunology , Cell Separation/instrumentation , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Fetal Blood/cytology , Flow Cytometry/instrumentation , Fluorescent Dyes/chemistry , Humans , Immunologic Surveillance , Killer Cells, Natural/metabolism , Lymphocyte Subsets/metabolism , Precursor Cells, T-Lymphoid/physiology , Primary Cell Culture/instrumentation , Primary Cell Culture/methods
3.
J Immunol ; 200(6): 2046-2056, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29436414

ABSTRACT

The PU.1 transcription factor plays a critical role in the regulation of T cell development, so a report that it is dispensable for fetal thymopoiesis is puzzling. To understand this paradox, we examined the requirement for PU.1, encoded by Spi1, during fetal, neonatal, and adult thymopoiesis in a PU.1 hypomorphic mouse generated by deletion of the Spi1 14-kb upstream regulatory element and by analysis of patterns of gene expression in fetal and adult T cell progenitors. Our data demonstrate that the initiation of thymopoiesis during early gestation is less dependent on PU.1 compared with T cell differentiation in adults and that fetal T cell progenitors express lower levels of Spi1 compared with their adult counterparts. We also show that expression of the core network of T lineage transcription factors regulated by PU.1 differs in fetal and adult T cell progenitors. In particular, PU.1-regulated genes that promote T cell differentiation are differentially expressed in fetal versus adult early T lineage progenitors. These results indicate that the transcriptional differences between the fetal and adult T cell developmental programs are driven in part by differential levels of PU.1 expression and that this likely underlies the differences in the properties of fetal and adult T cell progenitors.


Subject(s)
Cell Lineage/physiology , Fetus/metabolism , Fetus/physiology , Proto-Oncogene Proteins/metabolism , T-Lymphocytes/physiology , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation/physiology , Gene Expression/physiology , Mice , Mice, Inbred C57BL , Precursor Cells, T-Lymphoid/physiology
4.
Mucosal Immunol ; 11(2): 333-344, 2018 03.
Article in English | MEDLINE | ID: mdl-28745324

ABSTRACT

The intestine is a major immune organ with several specialized lymphoid structures and immune cells. Among these are thymus-derived natural intraepithelial lymphocytes (IELs) that lack expression of the classical co-receptors CD4 or CD8αß (double negative (DN)). Natural IELs are both αß+ and γδ+ T cells that play important roles in the maintenance of the epithelial barrier at steady state and during inflammation. The transcription factor T-bet is essential for the peripheral development of natural IELs, but its role during thymic development has remained less clear. Here we show that a T-bet gradient in DN TCRαß+NK1.1- thymocytes (IEL precursors (IELPs)) determines IEL fate in natural TCRαß+ IELs. Employing T-bet ZsGreen reporter mice in in vitro cultures and in vivo transfer experiments, we demonstrate that with increasing expression of T-bet, DN TCRαß+NK1.1- thymocytes are gradually restricted to a DN IEL fate. Furthermore, we show that the natural TCRαß+ IELs seed the intestine within the first month of life. This in turn is preceded by the appearance of T-bet- and T-bet+ IELPs that egress from the thymus in a sphingosine-1-phosphate (S1P)-dependent manner. In summary, the use of T-bet reporter mice has enabled us to identify and refine an immediate and clearly committed postselection precursor of natural TCRαß+ IELs.


Subject(s)
Intestines/immunology , Intraepithelial Lymphocytes/physiology , Precursor Cells, T-Lymphoid/physiology , T-Lymphocytes/physiology , Thymus Gland/physiology , Animals , Cell Differentiation , Cells, Cultured , Clonal Selection, Antigen-Mediated , Lysophospholipids/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
5.
J Immunol ; 199(8): 2758-2766, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28877990

ABSTRACT

Themis is a new component of the TCR signaling machinery that plays a critical role during T cell development. The positive selection of immature CD4+CD8+ double-positive thymocytes and their commitment to the CD4+CD8- single-positive stage are impaired in Themis-/- mice, suggesting that Themis might be important to sustain TCR signals during these key developmental processes. However, the analysis of Themis mRNA levels revealed that Themis gene expression is rapidly extinguished during positive selection. We show in this article that Themis protein expression is increased in double-positive thymocytes undergoing positive selection and is sustained in immature single-positive thymocytes, despite the strong decrease in Themis mRNA levels in these subsets. We found that Themis stability is controlled by the ubiquitin-specific protease USP9X, which removes ubiquitin K48-linked chains on Themis following TCR engagement. Biochemical analyses indicate that USP9X binds directly to the N-terminal CABIT domain of Themis and indirectly to the adaptor protein Grb2, with the latter interaction enabling recruitment of Themis/USP9X complexes to LAT, thereby sustaining Themis expression following positive selection. Together, these data suggest that TCR-mediated signals enhance Themis stability upon T cell development and identify USP9X as a key regulator of Themis protein turnover.


Subject(s)
Endopeptidases/metabolism , Precursor Cells, T-Lymphoid/physiology , Proteins/metabolism , T-Lymphocytes/physiology , Thymus Gland/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Differentiation , Cells, Cultured , Clonal Selection, Antigen-Mediated , GRB2 Adaptor Protein/metabolism , Intercellular Signaling Peptides and Proteins , Lymphocyte Activation , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins/metabolism , Protein Binding , Protein Stability , Proteins/genetics , Receptors, Antigen, T-Cell/metabolism , Ubiquitin Thiolesterase
6.
J Immunol ; 199(8): 2767-2776, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28893952

ABSTRACT

Early thymic progenitors (ETPs) are endowed with diverse potencies and can give rise to myeloid and lymphoid lineage progenitors. How the thymic environment guides ETP commitment and maturation toward a specific lineage remains obscure. We have previously shown that ETPs expressing the heteroreceptor (HR) comprising IL-4Rα and IL-13Rα1 give rise to myeloid cells but not T cells. In this article, we show that signaling through the HR inhibits ETP maturation to the T cell lineage but enacts commitment toward the myeloid cells. Indeed, HR+ ETPs, but not HR- ETPs, exhibit activated STAT6 transcription factor, which parallels with downregulation of Notch1, a critical factor for T cell development. Meanwhile, the myeloid-specific transcription factor C/EBPα, usually under the control of Notch1, is upregulated. Furthermore, in vivo inhibition of STAT6 phosphorylation restores Notch1 expression in HR+ ETPs, which regain T lineage potential. In addition, upon stimulation with IL-4 or IL-13, HR- ETPs expressing virally transduced HR also exhibit STAT6 phosphorylation and downregulation of Notch1, leading to inhibition of lymphoid, but not myeloid, lineage potential. These observations indicate that environmental cytokines play a role in conditioning ETP lineage choice, which would impact T cell development.


Subject(s)
Interleukin-13 Receptor alpha1 Subunit/metabolism , Interleukin-13/metabolism , Interleukin-4/metabolism , Precursor Cells, T-Lymphoid/physiology , Receptors, Cell Surface/metabolism , T-Lymphocytes/physiology , Thymus Gland/immunology , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation , Cell Lineage , Cells, Cultured , Interleukin-13 Receptor alpha1 Subunit/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/physiology , Receptors, Cell Surface/genetics , STAT6 Transcription Factor/metabolism , Signal Transduction
7.
Nat Immunol ; 18(2): 173-183, 2017 02.
Article in English | MEDLINE | ID: mdl-27992401

ABSTRACT

Most Foxp3+ regulatory T (Treg) cells develop in the thymus as a functionally mature T cell subpopulation specialized for immune suppression. Their cell fate appears to be determined before Foxp3 expression; yet molecular events that prime Foxp3- Treg precursor cells are largely obscure. We found that Treg cell-specific super-enhancers (Treg-SEs), which were associated with Foxp3 and other Treg cell signature genes, began to be activated in Treg precursor cells. T cell-specific deficiency of the genome organizer Satb1 impaired Treg-SE activation and the subsequent expression of Treg signature genes, causing severe autoimmunity due to Treg cell deficiency. These results suggest that Satb1-dependent Treg-SE activation is crucial for Treg cell lineage specification in the thymus and that its perturbation is causative of autoimmune and other immunological diseases.


Subject(s)
Cell Differentiation/immunology , Forkhead Transcription Factors/metabolism , Matrix Attachment Region Binding Proteins/metabolism , T-Lymphocytes, Regulatory/physiology , Transcriptional Activation/immunology , Animals , Autoimmunity , Cell Lineage , Cells, Cultured , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Forkhead Transcription Factors/genetics , Immune Tolerance , Male , Matrix Attachment Region Binding Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity , Precursor Cells, T-Lymphoid/physiology
8.
Nat Immunol ; 18(1): 45-53, 2017 01.
Article in English | MEDLINE | ID: mdl-27869820

ABSTRACT

TET proteins oxidize 5-methylcytosine in DNA to 5-hydroxymethylcytosine and other oxidation products. We found that simultaneous deletion of Tet2 and Tet3 in mouse CD4+CD8+ double-positive thymocytes resulted in dysregulated development and proliferation of invariant natural killer T cells (iNKT cells). Tet2-Tet3 double-knockout (DKO) iNKT cells displayed pronounced skewing toward the NKT17 lineage, with increased DNA methylation and impaired expression of genes encoding the key lineage-specifying factors T-bet and ThPOK. Transfer of purified Tet2-Tet3 DKO iNKT cells into immunocompetent recipient mice resulted in an uncontrolled expansion that was dependent on the nonclassical major histocompatibility complex (MHC) protein CD1d, which presents lipid antigens to iNKT cells. Our data indicate that TET proteins regulate iNKT cell fate by ensuring their proper development and maturation and by suppressing aberrant proliferation mediated by the T cell antigen receptor (TCR).


Subject(s)
Cell Differentiation , DNA-Binding Proteins/metabolism , Natural Killer T-Cells/physiology , Precursor Cells, T-Lymphoid/physiology , Proto-Oncogene Proteins/metabolism , Animals , Antigens, CD1d/genetics , Antigens, CD1d/metabolism , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Cell Lineage , Cell Proliferation , Cells, Cultured , DNA Methylation/genetics , DNA-Binding Proteins/genetics , Dioxygenases , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/genetics , Receptors, Antigen, T-Cell/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Trends Immunol ; 38(2): 128-139, 2017 02.
Article in English | MEDLINE | ID: mdl-27842955

ABSTRACT

T cells are continually generated in the thymus in a highly dynamic process comprising discrete steps of lineage commitment, T cell receptor (TCR) gene rearrangement, and selection. These steps are linked to distinct rates of proliferation, survival, and cell death, but a quantitative picture of T cell development is only beginning to emerge. Here we summarize recent technical advances, including genetic fate mapping, barcoding, and molecular timers, that have allowed the implementation of computational models to quantify developmental dynamics in the thymus. Coupling new techniques with mathematical models has recently resulted in the emergence of new paradigms in early hematopoiesis and might similarly open new perspectives on T cell development.


Subject(s)
Cell Differentiation , Models, Theoretical , Precursor Cells, T-Lymphoid/physiology , T-Lymphocytes/physiology , Thymus Gland/immunology , Animals , Cell Lineage , Hematopoiesis , Humans , Immunoassay , Lymphocyte Activation , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism
10.
Proc Natl Acad Sci U S A ; 113(48): E7788-E7797, 2016 11 29.
Article in English | MEDLINE | ID: mdl-27849617

ABSTRACT

Adoptive immunotherapy retargeting T cells to CD19 via a chimeric antigen receptor (CAR) is an investigational treatment capable of inducing complete tumor regression of B-cell malignancies when there is sustained survival of infused cells. T-memory stem cells (TSCM) retain superior potential for long-lived persistence, but challenges exist in manufacturing this T-cell subset because they are rare among circulating lymphocytes. We report a clinically relevant approach to generating CAR+ T cells with preserved TSCM potential using the Sleeping Beauty platform. Because IL-15 is fundamental to T-cell memory, we incorporated its costimulatory properties by coexpressing CAR with a membrane-bound chimeric IL-15 (mbIL15). The mbIL15-CAR T cells signaled through signal transducer and activator of transcription 5 to yield improved T-cell persistence independent of CAR signaling, without apparent autonomous growth or transformation, and achieved potent rejection of CD19+ leukemia. Long-lived T cells were CD45ROnegCCR7+CD95+, phenotypically most similar to TSCM, and possessed a memory-like transcriptional profile. Overall, these results demonstrate that CAR+ T cells can develop long-term persistence with a memory stem-cell phenotype sustained by signaling through mbIL15. This observation warrants evaluation in clinical trials.


Subject(s)
Interleukin-15/metabolism , Neoplasms, Experimental/therapy , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/physiology , Animals , Antigens, CD19/metabolism , Humans , Immunotherapy, Adoptive , Lymphocyte Activation , Mice , Precursor Cells, T-Lymphoid/physiology , Recombinant Fusion Proteins/metabolism , Signal Transduction
11.
Nat Immunol ; 17(10): 1206-1215, 2016 10.
Article in English | MEDLINE | ID: mdl-27548434

ABSTRACT

Thymic epithelial cell differentiation, growth and function depend on the expression of the transcription factor Foxn1; however, its target genes have never been physically identified. Using static and inducible genetic model systems and chromatin studies, we developed a genome-wide map of direct Foxn1 target genes for postnatal thymic epithelia and defined the Foxn1 binding motif. We determined the function of Foxn1 in these cells and found that, in addition to the transcriptional control of genes involved in the attraction and lineage commitment of T cell precursors, Foxn1 regulates the expression of genes involved in antigen processing and thymocyte selection. Thus, critical events in thymic lympho-stromal cross-talk and T cell selection are indispensably choreographed by Foxn1.


Subject(s)
Epithelial Cells/physiology , Forkhead Transcription Factors/metabolism , Precursor Cells, T-Lymphoid/physiology , T-Lymphocytes/physiology , Thymus Gland/physiology , Animals , Antigen Presentation/genetics , Cell Communication , Cell Differentiation/genetics , Cell Lineage/genetics , Cells, Cultured , Clonal Selection, Antigen-Mediated/genetics , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Genome/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic
12.
J Immunol ; 197(6): 2280-9, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27489283

ABSTRACT

Although ribosomal proteins (RP) are thought to primarily facilitate biogenesis of the ribosome and its ability to synthesize protein, emerging evidence suggests that individual RP can perform critical regulatory functions that control developmental processes. We showed previously that despite the ubiquitous expression of the RP ribosomal protein L22 (Rpl22), germline ablation of Rpl22 in mice causes a selective, p53-dependent block in the development of αß, but not γδ, T cell progenitors. Nevertheless, the basis by which Rpl22 loss selectively induces p53 in αß T cell progenitors remained unclear. We show in this study that Rpl22 regulates the development of αß T cells by restraining endoplasmic reticulum (ER) stress responses. In the absence of Rpl22, ER stress is exacerbated in αß, but not γδ, T cell progenitors. The exacerbated ER stress in Rpl22-deficient αß T lineage progenitors is responsible for selective induction of p53 and their arrest, as pharmacological induction of stress is sufficient to induce p53 and replicate the selective block of αß T cells, and attenuation of ER stress signaling by knockdown of protein kinase R-like ER kinase, an ER stress sensor, blunts p53 induction and rescues development of Rpl22-deficient αß T cell progenitors. Rpl22 deficiency appears to exacerbate ER stress by interfering with the ability of ER stress signals to block new protein synthesis. Our finding that Rpl22 deficiency exacerbates ER stress responses and induces p53 in αß T cell progenitors provides insight into how a ubiquitously expressed RP can perform regulatory functions that are selectively required by some cell lineages but not others.


Subject(s)
Endoplasmic Reticulum Stress , Gene Expression Regulation , Precursor Cells, T-Lymphoid/physiology , RNA-Binding Proteins/physiology , Receptors, Antigen, T-Cell, alpha-beta , Ribosomal Proteins/physiology , Signal Transduction , T-Lymphocyte Subsets/physiology , Animals , Cell Differentiation , Cell Lineage/physiology , Mice , Ribosomal Proteins/deficiency , T-Lymphocyte Subsets/immunology , Tumor Suppressor Protein p53/metabolism
13.
Adv Immunol ; 129: 109-74, 2016.
Article in English | MEDLINE | ID: mdl-26791859

ABSTRACT

T-lymphocyte development branches off from other lymphoid developmental programs through its requirement for sustained environmental signals through the Notch pathway. In the thymus, Notch signaling induces a succession of T-lineage regulatory factors that collectively create the T-cell identity through distinct steps. This process involves both the staged activation of T-cell identity genes and the staged repression of progenitor-cell-inherited regulatory genes once their roles in self-renewal and population expansion are no longer needed. With the recent characterization of innate lymphoid cells (ILCs) that share transcriptional regulation programs extensively with T-cell subsets, T-cell identity can increasingly be seen as defined in modular terms, as the processes selecting and actuating effector function are potentially detachable from the processes generating and selecting clonally unique T-cell receptor structures. The developmental pathways of different classes of T cells and ILCs are distinguished by the numbers of prerequisites of gene rearrangement, selection, and antigen contact before the cells gain access to nearly common regulatory mechanisms for choosing effector function. Here, the major classes of transcription factors that interact with Notch signals during T-lineage specification are discussed in terms of their roles in these programs, the evidence for their spectra of target genes at different stages, and their cross-regulatory and cooperative actions with each other. Specific topics include Notch modulation of PU.1 and GATA-3, PU.1-Notch competition, the relationship between PU.1 and GATA-3, and the roles of E proteins, Bcl11b, and GATA-3 in guiding acquisition of T-cell identity while avoiding redirection to an ILC fate.


Subject(s)
Gene Expression Regulation/immunology , Precursor Cells, T-Lymphoid/physiology , Receptors, Notch/metabolism , T-Lymphocyte Subsets/physiology , Transcription Factors/metabolism , Transcription, Genetic/immunology , Animals , Cell Differentiation , Cellular Microenvironment , Humans , Receptors, Notch/immunology , Signal Transduction , Transcription Factors/immunology
14.
Eur J Immunol ; 46(3): 531-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26763078

ABSTRACT

T-cell development occurs in multipotent progenitors arriving in the thymus, which provides a highly specialized microenvironment. Specification and sequential commitment processes to T cells begin in early thymic progenitors upon receiving thymus-specific environmental cues, resulting in the activation of the genetically programmed transcriptional cascade that includes turning on and off numerous transcription factors in a precise manner. Thus, early thymocyte differentiation has been an excellent model system to study cell differentiation processes. This review summarizes recent advances in our knowledge on thymic T-cell development from newly arrived multipotent T-cell progenitors to fully committed T-cell precursors, from the transcriptional regulation perspective.


Subject(s)
Gene Expression Regulation , T-Lymphocytes/immunology , Thymus Gland/immunology , Transcription Factors/metabolism , Transcriptional Activation , Cell Differentiation/genetics , Cell Lineage , Lymphocyte Activation , Precursor Cells, T-Lymphoid/physiology , T-Lymphocytes/physiology , Thymus Gland/cytology , Thymus Gland/physiology
15.
Nat Immunol ; 16(6): 628-34, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25939024

ABSTRACT

Most T lymphocytes, including regulatory T cells (Treg cells), differentiate in the thymus. The age-dependent involution of this organ leads to decreasing production of T cells. Here we found that the output of new Treg cells from the thymus decreased substantially more than that of conventional T cells. Peripheral mouse and human Treg cells recirculated back to the thymus, where they constituted a large proportion of the pool of Treg cells and displayed an activated and differentiated phenotype. In the thymus, the recirculating cells exerted their regulatory function by inhibiting interleukin 2 (IL-2)-dependent de novo differentiation of Treg cells. Thus, Treg cell development is controlled by a negative feedback loop in which mature progeny cells return to the thymus and restrain development of precursors of Treg cells.


Subject(s)
Precursor Cells, T-Lymphoid/physiology , T-Lymphocyte Subsets/physiology , T-Lymphocytes, Regulatory/physiology , Thymus Gland/immunology , Aging/immunology , Animals , Blood Circulation , Cell Differentiation/genetics , Cells, Cultured , Child , Feedback, Physiological , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Immune Tolerance , Interleukin-2/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic
16.
Nat Cell Biol ; 17(5): 580-91, 2015 May.
Article in English | MEDLINE | ID: mdl-25915127

ABSTRACT

The generation of haematopoietic stem cells (HSCs) from human pluripotent stem cells (hPSCs) will depend on the accurate recapitulation of embryonic haematopoiesis. In the early embryo, HSCs develop from the haemogenic endothelium (HE) and are specified in a Notch-dependent manner through a process named endothelial-to-haematopoietic transition (EHT). As HE is associated with arteries, it is assumed that it represents a subpopulation of arterial vascular endothelium (VE). Here we demonstrate at a clonal level that hPSC-derived HE and VE represent separate lineages. HE is restricted to the CD34(+)CD73(-)CD184(-) fraction of day 8 embryoid bodies and it undergoes a NOTCH-dependent EHT to generate RUNX1C(+) cells with multilineage potential. Arterial and venous VE progenitors, in contrast, segregate to the CD34(+)CD73(med)CD184(+) and CD34(+)CD73(hi)CD184(-) fractions, respectively. Together, these findings identify HE as distinct from VE and provide a platform for defining the signalling pathways that regulate their specification to functional HSCs.


Subject(s)
Arteries/physiology , Cell Differentiation , Cell Lineage , Endothelial Progenitor Cells/physiology , Hematopoietic Stem Cells/physiology , Multipotent Stem Cells/physiology , Pluripotent Stem Cells/physiology , 5'-Nucleotidase/deficiency , Antigens, CD34/metabolism , Arteries/cytology , Arteries/metabolism , Biomarkers/metabolism , Cell Line , Cell Separation/methods , Coculture Techniques , Core Binding Factor Alpha 2 Subunit/metabolism , Endothelial Progenitor Cells/metabolism , GPI-Linked Proteins/deficiency , Hematopoietic Stem Cells/metabolism , Humans , Microscopy, Video , Multipotent Stem Cells/metabolism , Phenotype , Pluripotent Stem Cells/metabolism , Precursor Cells, T-Lymphoid/physiology , Receptors, CXCR5/deficiency , Receptors, Notch/metabolism , Signal Transduction , Time Factors , Veins/cytology , Veins/physiology
17.
Cell Stem Cell ; 16(4): 348-9, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25842973

ABSTRACT

Mutations causing X-linked severe combined immunodeficiency (SCID-X1) reduce immune cell populations and function and may be amenable to targeted gene correction strategies. Now in Cell Stem Cell, Menon et al. (2015) correct SCID-X1-related blood differentiation defects by TALEN-mediated genome editing in patient-derived iPSCs, suggesting a possible strategy for autologous cell therapy of SCID-X1.


Subject(s)
Genetic Therapy/methods , Immunotherapy, Adoptive , Induced Pluripotent Stem Cells/physiology , Killer Cells, Natural/physiology , Precursor Cells, T-Lymphoid/physiology , Regeneration , Regenerative Medicine , X-Linked Combined Immunodeficiency Diseases/therapy , Humans
18.
Cell Stem Cell ; 16(4): 367-72, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25772073

ABSTRACT

X-linked Severe Combined Immunodeficiency (SCID-X1) is a genetic disease that leaves newborns at high risk of serious infection and a predicted life span of less than 1 year in the absence of a matched bone marrow donor. The disease pathogenesis is due to mutations in the gene encoding the Interleukin-2 receptor gamma chain (IL-2Rγ), leading to a lack of functional lymphocytes. With the leukemogenic concerns of viral gene therapy there is a need to explore alternative therapeutic options. We have utilized induced pluripotent stem cell (iPSC) technology and genome editing mediated by TALENs to generate isogenic subject-specific mutant and gene-corrected iPSC lines. While the subject-derived mutant iPSCs have the capacity to generate hematopoietic precursors and myeloid cells, only wild-type and gene-corrected iPSCs can additionally generate mature NK cells and T cell precursors expressing the correctly spliced IL-2Rγ. This study highlights the potential for the development of autologous cell therapy for SCID-X1 subjects.


Subject(s)
Genetic Therapy/methods , Immunotherapy, Adoptive , Induced Pluripotent Stem Cells/physiology , Killer Cells, Natural/physiology , Precursor Cells, T-Lymphoid/physiology , Regeneration , Regenerative Medicine , X-Linked Combined Immunodeficiency Diseases/therapy , Antigens, CD/metabolism , Bacterial Proteins/metabolism , Cell Differentiation/genetics , Cell Line , DNA Repair , DNA Repair Enzymes/metabolism , Humans , Induced Pluripotent Stem Cells/transplantation , Infant , Interleukin Receptor Common gamma Subunit/genetics , Killer Cells, Natural/transplantation , Mutation/genetics , Precursor Cells, T-Lymphoid/transplantation , X-Linked Combined Immunodeficiency Diseases/genetics
19.
Proc Natl Acad Sci U S A ; 111(10): 3805-10, 2014 Mar 11.
Article in English | MEDLINE | ID: mdl-24567410

ABSTRACT

mTOR is an evolutionarily conserved kinase that plays a critical role in sensing and responding to environmental determinants. Recent studies have shown that fine-tuning of the activity of mTOR complexes contributes to organogenesis and tumorigenesis. Although rapamycin, an allosteric mTOR inhibitor, is an effective immunosuppressant, the precise roles of mTOR complexes in early T-cell development remain unclear. Here we show that mTORC1 plays a critical role in the development of both early T-cell progenitors and leukemia. Deletion of Raptor, an essential component of mTORC1, produced defects in the earliest development of T-cell progenitors in vivo and in vitro. Deficiency of Raptor resulted in cell cycle abnormalities in early T-cell progenitors that were associated with instability of the Cyclin D2/D3-CDK6 complexes; deficiency of Rictor, an mTORC2 component, did not have the same effect, indicating that mTORC1 and -2 control T-cell development in different ways. In a model of myeloproliferative neoplasm and T-cell acute lymphoblastic leukemia (T-ALL) evoked by Kras activation, Raptor deficiency dramatically inhibited the cell cycle in oncogenic Kras-expressing T-cell progenitors, but not myeloid progenitors, and specifically prevented the development of T-ALL. Although rapamycin treatment significantly prolonged the survival of recipient mice bearing T-ALL cells, rapamycin-insensitive leukemia cells continued to propagate in vivo. In contrast, Raptor deficiency in the T-ALL model resulted in cell cycle arrest and efficient eradication of leukemia. Thus, understanding the cell-context-dependent role of mTORC1 illustrates the potential importance of mTOR signals as therapeutic targets.


Subject(s)
Lymphopoiesis/physiology , Models, Immunological , Multiprotein Complexes/physiology , Precursor Cells, T-Lymphoid/physiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/physiology , Adaptor Proteins, Signal Transducing/deficiency , Animals , Carrier Proteins/metabolism , Cell Cycle/immunology , Cell Cycle/physiology , DNA Primers , Flow Cytometry , Gene Expression Profiling , Immunoblotting , Immunohistochemistry , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Multiprotein Complexes/deficiency , Rapamycin-Insensitive Companion of mTOR Protein , Regulatory-Associated Protein of mTOR , Signal Transduction/physiology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/deficiency
20.
PLoS Pathog ; 8(2): e1002544, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22383879

ABSTRACT

The elderly are particularly susceptible to influenza A virus infections, with increased occurrence, disease severity and reduced vaccine efficacy attributed to declining immunity. Experimentally, the age-dependent decline in influenza-specific CD8(+) T cell responsiveness reflects both functional compromise and the emergence of 'repertoire holes' arising from the loss of low frequency clonotypes. In this study, we asked whether early priming limits the time-related attrition of immune competence. Though primary responses in aged mice were compromised, animals vaccinated at 6 weeks then challenged >20 months later had T-cell responses that were normal in magnitude. Both functional quality and the persistence of 'preferred' TCR clonotypes that expand in a characteristic immunodominance hierarchy were maintained following early priming. Similar to the early priming, vaccination at 22 months followed by challenge retained a response magnitude equivalent to young mice. However, late priming resulted in reduced TCRß diversity in comparison with vaccination earlier in life. Thus, early priming was critical to maintaining individual and population-wide TCRß diversity. In summary, early exposure leads to the long-term maintenance of memory T cells and thus preserves optimal, influenza-specific CD8(+) T-cell responsiveness and protects against the age-related attrition of naïve T-cell precursors. Our study supports development of vaccines that prime CD8(+) T-cells early in life to elicit the broadest possible spectrum of CD8(+) T-cell memory and preserve the magnitude, functionality and TCR usage of responding populations. In addition, our study provides the most comprehensive analysis of the aged (primary, secondary primed-early and secondary primed-late) TCR repertoires published to date.


Subject(s)
Aging/immunology , Antigen Presentation/physiology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/physiology , Immunologic Memory/physiology , Age Factors , Animals , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Epitopes, T-Lymphocyte/immunology , Female , Mice , Mice, Inbred C57BL , Precursor Cells, T-Lymphoid/immunology , Precursor Cells, T-Lymphoid/physiology , Receptors, Antigen, T-Cell/immunology , Validation Studies as Topic
SELECTION OF CITATIONS
SEARCH DETAIL
...