Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.311
Filter
1.
Annu Rev Immunol ; 36: 579-601, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29677476

ABSTRACT

A fundamental question in developmental immunology is how bipotential thymocyte precursors generate both CD4+ helper and CD8+ cytotoxic T cell lineages. The MHC specificity of αß T cell receptors (TCRs) on precursors is closely correlated with cell fate-determining processes, prompting studies to characterize how variations in TCR signaling are linked with genetic programs establishing lineage-specific gene expression signatures, such as exclusive CD4 or CD8 expression. The key transcription factors ThPOK and Runx3 have been identified as mediating development of helper and cytotoxic T cell lineages, respectively. Together with increasing knowledge of epigenetic regulators, these findings have advanced our understanding of the transcription factor network regulating the CD4/CD8 dichotomy. It has also become apparent that CD4+ T cells retain developmental plasticity, allowing them to acquire cytotoxic activity in the periphery. Despite such advances, further studies are necessary to identify the molecular links between TCR signaling and the nuclear machinery regulating expression of ThPOK and Runx3.


Subject(s)
Cell Differentiation/immunology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , Animals , CD4 Antigens/genetics , CD4 Antigens/metabolism , CD8 Antigens/genetics , CD8 Antigens/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Lineage/immunology , Core Binding Factor Alpha 3 Subunit/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation , Humans , Immunomodulation/genetics , Immunomodulation/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Regulatory Sequences, Nucleic Acid , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Transcription Factors/genetics , Transcription, Genetic
2.
Cell ; 186(23): 5183-5199.e22, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37852258

ABSTRACT

Cellular lineage histories and their molecular states encode fundamental principles of tissue development and homeostasis. Current lineage-recording mouse models have insufficient barcode diversity and single-cell lineage coverage for profiling tissues composed of millions of cells. Here, we developed DARLIN, an inducible Cas9 barcoding mouse line that utilizes terminal deoxynucleotidyl transferase (TdT) and 30 CRISPR target sites. DARLIN is inducible, generates massive lineage barcodes across tissues, and enables the detection of edited barcodes in ∼70% of profiled single cells. Using DARLIN, we examined fate bias within developing hematopoietic stem cells (HSCs) and revealed unique features of HSC migration. Additionally, we established a protocol for joint transcriptomic and epigenomic single-cell measurements with DARLIN and found that cellular clonal memory is associated with genome-wide DNA methylation rather than gene expression or chromatin accessibility. DARLIN will enable the high-resolution study of lineage relationships and their molecular signatures in diverse tissues and physiological contexts.


Subject(s)
Epigenomics , Transcriptome , Animals , Mice , Transcriptome/genetics , Cell Lineage/genetics , Gene Expression Profiling , Disease Models, Animal , DNA
3.
Annu Rev Immunol ; 33: 607-42, 2015.
Article in English | MEDLINE | ID: mdl-25665079

ABSTRACT

The lymphocyte family has expanded significantly in recent years to include not only the adaptive lymphocytes (T cells, B cells) and NK cells, but also several additional innate lymphoid cell (ILC) types. ILCs lack clonally distributed antigen receptors characteristic of adaptive lymphocytes and instead respond exclusively to signaling via germline-encoded receptors. ILCs resemble T cells more closely than any other leukocyte lineage at the transcriptome level and express many elements of the core T cell transcriptional program, including Notch, Gata3, Tcf7, and Bcl11b. We present our current understanding of the shared and distinct transcriptional regulatory mechanisms involved in the development of adaptive T lymphocytes and closely related ILCs. We discuss the possibility that a core set of transcriptional regulators common to ILCs and T cells establish enhancers that enable implementation of closely aligned effector pathways. Studies of the transcriptional regulation of lymphopoiesis will support the development of novel therapeutic approaches to correct early lymphoid developmental defects and aberrant lymphocyte function.


Subject(s)
Adaptive Immunity/genetics , Cell Lineage/genetics , Gene Expression Regulation , Immunity, Innate/genetics , Lymphocytes/immunology , Lymphocytes/metabolism , Transcription, Genetic , Animals , Cell Differentiation , Humans , Lymphocytes/cytology , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism
4.
Cell ; 185(24): 4604-4620.e32, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36423582

ABSTRACT

Natural and induced somatic mutations that accumulate in the genome during development record the phylogenetic relationships of cells; whether these lineage barcodes capture the complex dynamics of progenitor states remains unclear. We introduce quantitative fate mapping, an approach to reconstruct the hierarchy, commitment times, population sizes, and commitment biases of intermediate progenitor states during development based on a time-scaled phylogeny of their descendants. To reconstruct time-scaled phylogenies from lineage barcodes, we introduce Phylotime, a scalable maximum likelihood clustering approach based on a general barcoding mutagenesis model. We validate these approaches using realistic in silico and in vitro barcoding experiments. We further establish criteria for the number of cells that must be analyzed for robust quantitative fate mapping and a progenitor state coverage statistic to assess the robustness. This work demonstrates how lineage barcodes, natural or synthetic, enable analyzing progenitor fate and dynamics long after embryonic development in any organism.


Subject(s)
Embryonic Development , Cell Lineage/genetics , Retrospective Studies , Phylogeny , Mutagenesis
5.
Cell ; 184(19): 5053-5069.e23, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34390642

ABSTRACT

Genetic perturbations of cortical development can lead to neurodevelopmental disease, including autism spectrum disorder (ASD). To identify genomic regions crucial to corticogenesis, we mapped the activity of gene-regulatory elements generating a single-cell atlas of gene expression and chromatin accessibility both independently and jointly. This revealed waves of gene regulation by key transcription factors (TFs) across a nearly continuous differentiation trajectory, distinguished the expression programs of glial lineages, and identified lineage-determining TFs that exhibited strong correlation between linked gene-regulatory elements and expression levels. These highly connected genes adopted an active chromatin state in early differentiating cells, consistent with lineage commitment. Base-pair-resolution neural network models identified strong cell-type-specific enrichment of noncoding mutations predicted to be disruptive in a cohort of ASD individuals and identified frequently disrupted TF binding sites. This approach illustrates how cell-type-specific mapping can provide insights into the programs governing human development and disease.


Subject(s)
Cerebral Cortex/embryology , Chromatin/metabolism , Gene Expression Regulation, Developmental , Single-Cell Analysis , Astrocytes/cytology , Cell Differentiation , Cell Lineage/genetics , Cluster Analysis , Deep Learning , Epigenesis, Genetic , Fuzzy Logic , Glutamates/metabolism , Humans , Mutation/genetics , Neurons/metabolism , Regulatory Sequences, Nucleic Acid/genetics
6.
Cell ; 184(11): 2843-2859.e20, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33991488

ABSTRACT

Since establishment of the first embryonic stem cells (ESCs), in vitro culture of totipotent cells functionally and molecularly comparable with in vivo blastomeres with embryonic and extraembryonic developmental potential has been a challenge. Here we report that spliceosomal repression in mouse ESCs drives a pluripotent-to-totipotent state transition. Using the splicing inhibitor pladienolide B, we achieve stable in vitro culture of totipotent ESCs comparable at molecular levels with 2- and 4-cell blastomeres, which we call totipotent blastomere-like cells (TBLCs). Mouse chimeric assays combined with single-cell RNA sequencing (scRNA-seq) demonstrate that TBLCs have a robust bidirectional developmental capability to generate multiple embryonic and extraembryonic cell lineages. Mechanically, spliceosomal repression causes widespread splicing inhibition of pluripotent genes, whereas totipotent genes, which contain few short introns, are efficiently spliced and transcriptionally activated. Our study provides a means for capturing and maintaining totipotent stem cells.


Subject(s)
Totipotent Stem Cells/cytology , Totipotent Stem Cells/metabolism , Animals , Blastomeres/cytology , Cell Differentiation/genetics , Cell Line , Cell Lineage/genetics , Embryo, Mammalian/cytology , Embryonic Stem Cells/cytology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mouse Embryonic Stem Cells/cytology , Totipotent Stem Cells/physiology
7.
Cell ; 184(3): 741-758.e17, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33484631

ABSTRACT

Both transcription and three-dimensional (3D) architecture of the mammalian genome play critical roles in neurodevelopment and its disorders. However, 3D genome structures of single brain cells have not been solved; little is known about the dynamics of single-cell transcriptome and 3D genome after birth. Here, we generated a transcriptome (3,517 cells) and 3D genome (3,646 cells) atlas of the developing mouse cortex and hippocampus by using our high-resolution multiple annealing and looping-based amplification cycles for digital transcriptomics (MALBAC-DT) and diploid chromatin conformation capture (Dip-C) methods and developing multi-omic analysis pipelines. In adults, 3D genome "structure types" delineate all major cell types, with high correlation between chromatin A/B compartments and gene expression. During development, both transcriptome and 3D genome are extensively transformed in the first post-natal month. In neurons, 3D genome is rewired across scales, correlated with gene expression modules, and independent of sensory experience. Finally, we examine allele-specific structure of imprinted genes, revealing local and chromosome (chr)-wide differences. These findings uncover an unknown dimension of neurodevelopment.


Subject(s)
Brain/growth & development , Genome , Sensation/genetics , Transcription, Genetic , Alleles , Animals , Animals, Newborn , Cell Lineage/genetics , Chromatin/metabolism , Gene Expression Regulation, Developmental , Gene Ontology , Gene Regulatory Networks , Genetic Loci , Genomic Imprinting , Mice , Multigene Family , Neuroglia/metabolism , Neurons/metabolism , Transcriptome/genetics , Visual Cortex/metabolism
8.
Cell ; 184(11): 3056-3074.e21, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33932339

ABSTRACT

The choroid plexus (ChP) in each brain ventricle produces cerebrospinal fluid (CSF) and forms the blood-CSF barrier. Here, we construct a single-cell and spatial atlas of each ChP in the developing, adult, and aged mouse brain. We delineate diverse cell types, subtypes, cell states, and expression programs in epithelial and mesenchymal cells across ages and ventricles. In the developing ChP, we predict a common progenitor pool for epithelial and neuronal cells, validated by lineage tracing. Epithelial and fibroblast cells show regionalized expression by ventricle, starting at embryonic stages and persisting with age, with a dramatic transcriptional shift with maturation, and a smaller shift in each aged cell type. With aging, epithelial cells upregulate host-defense programs, and resident macrophages upregulate interleukin-1ß (IL-1ß) signaling genes. Our atlas reveals cellular diversity, architecture and signaling across ventricles during development, maturation, and aging of the ChP-brain barrier.


Subject(s)
Choroid Plexus/embryology , Choroid Plexus/metabolism , Age Factors , Aging/physiology , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Brain/physiology , Brain Diseases/genetics , Brain Diseases/physiopathology , Cell Differentiation/genetics , Cell Lineage/genetics , Choroid Plexus/physiology , Epithelial Cells/metabolism , Female , Male , Mice/embryology , Mice, Inbred C57BL , Signal Transduction , Single-Cell Analysis
9.
Nat Immunol ; 24(10): 1698-1710, 2023 10.
Article in English | MEDLINE | ID: mdl-37592014

ABSTRACT

In development, pioneer transcription factors access silent chromatin to reveal lineage-specific gene programs. The structured DNA-binding domains of pioneer factors have been well characterized, but whether and how intrinsically disordered regions affect chromatin and control cell fate is unclear. Here, we report that deletion of an intrinsically disordered region of the pioneer factor TCF-1 (termed L1) leads to an early developmental block in T cells. The few T cells that develop from progenitors expressing TCF-1 lacking L1 exhibit lineage infidelity distinct from the lineage diversion of TCF-1-deficient cells. Mechanistically, L1 is required for activation of T cell genes and repression of GATA2-driven genes, normally reserved to the mast cell and dendritic cell lineages. Underlying this lineage diversion, L1 mediates binding of TCF-1 to its earliest target genes, which are subject to repression as T cells develop. These data suggest that the intrinsically disordered N terminus of TCF-1 maintains T cell lineage fidelity.


Subject(s)
T-Lymphocytes , Transcription Factors , Transcription Factors/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , T-Lymphocytes/metabolism , T Cell Transcription Factor 1/genetics , Chromatin/metabolism
10.
Annu Rev Immunol ; 31: 31-50, 2013.
Article in English | MEDLINE | ID: mdl-23121398

ABSTRACT

The generation of the TCRαß lineage of T cells occurs in the thymus through a series of orchestrated developmental events that result in a carefully selected population of CD4 or CD8 lineage-committed TCR(+) thymocytes capable of recognizing foreign antigen in the context of self MHC. T cells first exit the thymus in a phenotypically and functionally immature state and require an approximately 3-week period of post-thymic maturation before transitioning into the mature T cell compartment. A greater understanding of recent thymic emigrant biology has come with the development of methods to exclusively identify and isolate this population for further characterization. I now review current knowledge about the phenotype and function of this key but understudied population of peripheral T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Lineage/immunology , Cell Movement/immunology , Cellular Senescence/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Movement/genetics , Cellular Senescence/genetics , Humans , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Thymus Gland/metabolism
11.
Cell ; 182(6): 1384-1400, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32946781

ABSTRACT

Hematopoiesis has long served as a paradigm of stem cell biology and tissue homeostasis. In the past decade, the genomics revolution has ushered in powerful new methods for investigating the hematopoietic system that have provided transformative insights into its biology. As part of the advances in genomics, increasingly accurate deep sequencing and novel methods of cell tracking have revealed hematopoiesis to be more of a continuous and less of a discrete and punctuated process than originally envisioned. In part, this continuous nature of hematopoiesis is made possible by the emergent outcomes of vast, interconnected regulatory networks that influence cell fates and lineage commitment. It is also becoming clear how these mechanisms are modulated by genetic variation present throughout the population. This review describes how these recently uncovered complexities are reshaping our concept of tissue development and homeostasis while opening up a more comprehensive future understanding of hematopoiesis.


Subject(s)
Cell Lineage , Gene Expression Regulation, Developmental , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Animals , Cell Lineage/genetics , Cell Lineage/physiology , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Genomics , Homeostasis , Human Genetics , Humans , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Cell ; 183(3): 702-716.e14, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33125890

ABSTRACT

The cellular complexity and scale of the early liver have constrained analyses examining its emergence during organogenesis. To circumvent these issues, we analyzed 45,334 single-cell transcriptomes from embryonic day (E)7.5, when endoderm progenitors are specified, to E10.5 liver, when liver parenchymal and non-parenchymal cell lineages emerge. Our data detail divergence of vascular and sinusoidal endothelia, including a distinct transcriptional profile for sinusoidal endothelial specification by E8.75. We characterize two distinct mesothelial cell types as well as early hepatic stellate cells and reveal distinct spatiotemporal distributions for these populations. We capture transcriptional profiles for hepatoblast specification and migration, including the emergence of a hepatomesenchymal cell type and evidence for hepatoblast collective cell migration. Further, we identify cell-cell interactions during the organization of the primitive sinusoid. This study provides a comprehensive atlas of liver lineage establishment from the endoderm and mesoderm through to the organization of the primitive sinusoid at single-cell resolution.


Subject(s)
Cell Lineage/genetics , Liver/cytology , Liver/metabolism , Single-Cell Analysis , Transcriptome/genetics , Animals , Cell Movement , Embryo, Mammalian/cytology , Endothelium/cytology , Mesoderm/cytology , Mice , Signal Transduction , Stem Cells/cytology
13.
Cell ; 183(4): 1103-1116.e20, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33098772

ABSTRACT

Cell differentiation and function are regulated across multiple layers of gene regulation, including modulation of gene expression by changes in chromatin accessibility. However, differentiation is an asynchronous process precluding a temporal understanding of regulatory events leading to cell fate commitment. Here we developed simultaneous high-throughput ATAC and RNA expression with sequencing (SHARE-seq), a highly scalable approach for measurement of chromatin accessibility and gene expression in the same single cell, applicable to different tissues. Using 34,774 joint profiles from mouse skin, we develop a computational strategy to identify cis-regulatory interactions and define domains of regulatory chromatin (DORCs) that significantly overlap with super-enhancers. During lineage commitment, chromatin accessibility at DORCs precedes gene expression, suggesting that changes in chromatin accessibility may prime cells for lineage commitment. We computationally infer chromatin potential as a quantitative measure of chromatin lineage-priming and use it to predict cell fate outcomes. SHARE-seq is an extensible platform to study regulatory circuitry across diverse cells in tissues.


Subject(s)
Chromatin/metabolism , Gene Expression Profiling , RNA/genetics , Single-Cell Analysis , Animals , Cell Differentiation/genetics , Cell Line , Cell Lineage/genetics , Enhancer Elements, Genetic/genetics , Female , Gene Expression Regulation , Histones/metabolism , Mice, Inbred C57BL , Protein Processing, Post-Translational , RNA/metabolism
14.
Cell ; 181(4): 922-935.e21, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32315617

ABSTRACT

Single-cell RNA sequencing (scRNA-seq) provides a leap forward in resolving cellular diversity and developmental trajectories but fails to comprehensively delineate the spatial organization and precise cellular makeup of individual embryos. Here, we reconstruct from scRNA-seq and light sheet imaging data a canonical digital embryo that captures the genome-wide gene expression trajectory of every single cell at every cell division in the 18 lineages up to gastrulation in the ascidian Phallusia mammillata. By using high-coverage scRNA-seq, we devise a computational framework that stratifies single cells of individual embryos into cell types without prior knowledge. Unbiased transcriptome data analysis mapped each cell's physical position and lineage history, yielding the complete history of gene expression at the genome-wide level for every single cell in a developing embryo. A comparison of individual embryos reveals both extensive reproducibility between symmetric embryo sides and a large inter-embryonic variability due to small differences in embryogenesis timing.


Subject(s)
Gene Expression Profiling/methods , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Animals , Cell Lineage/genetics , Chordata/genetics , Computational Biology/methods , Gastrulation/genetics , Gene Expression Regulation, Developmental/genetics , Reproducibility of Results , Transcriptome/genetics , Urochordata/genetics
15.
Cell ; 181(6): 1410-1422.e27, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32413320

ABSTRACT

Tracing the lineage history of cells is key to answering diverse and fundamental questions in biology. Coupling of cell ancestry information with other molecular readouts represents an important goal in the field. Here, we describe the CRISPR array repair lineage tracing (CARLIN) mouse line and corresponding analysis tools that can be used to simultaneously interrogate the lineage and transcriptomic information of single cells in vivo. This model exploits CRISPR technology to generate up to 44,000 transcribed barcodes in an inducible fashion at any point during development or adulthood, is compatible with sequential barcoding, and is fully genetically defined. We have used CARLIN to identify intrinsic biases in the activity of fetal liver hematopoietic stem cell (HSC) clones and to uncover a previously unappreciated clonal bottleneck in the response of HSCs to injury. CARLIN also allows the unbiased identification of transcriptional signatures associated with HSC activity without cell sorting.


Subject(s)
CRISPR-Cas Systems/genetics , Cell Lineage/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Transcriptome/genetics , Animals , Cell Line , Female , Flow Cytometry/methods , Hematopoietic Stem Cells/physiology , Male , Mice , Transduction, Genetic/methods
16.
Cell ; 181(3): 604-620.e22, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32259486

ABSTRACT

During embryonic and postnatal development, organs and tissues grow steadily to achieve their final size at the end of puberty. However, little is known about the cellular dynamics that mediate postnatal growth. By combining in vivo clonal lineage tracing, proliferation kinetics, single-cell transcriptomics, and in vitro micro-pattern experiments, we resolved the cellular dynamics taking place during postnatal skin epidermis expansion. Our data revealed that harmonious growth is engineered by a single population of developmental progenitors presenting a fixed fate imbalance of self-renewing divisions with an ever-decreasing proliferation rate. Single-cell RNA sequencing revealed that epidermal developmental progenitors form a more uniform population compared with adult stem and progenitor cells. Finally, we found that the spatial pattern of cell division orientation is dictated locally by the underlying collagen fiber orientation. Our results uncover a simple design principle of organ growth where progenitors and differentiated cells expand in harmony with their surrounding tissues.


Subject(s)
Epidermal Cells/metabolism , Epidermis/growth & development , Skin/growth & development , Animals , Animals, Outbred Strains , Cell Differentiation/physiology , Cell Division/physiology , Cell Lineage/genetics , Cell Proliferation/physiology , Cells, Cultured , Epidermal Cells/pathology , Epidermis/metabolism , Female , Male , Mice , Mice, Transgenic , Stem Cells/cytology
17.
Nat Immunol ; 23(4): 505-517, 2022 04.
Article in English | MEDLINE | ID: mdl-35354960

ABSTRACT

Intrinsic and extrinsic cues determine developmental trajectories of hematopoietic stem cells (HSCs) towards erythroid, myeloid and lymphoid lineages. Using two newly generated transgenic mice that report and trace the expression of terminal deoxynucleotidyl transferase (TdT), transient induction of TdT was detected on a newly identified multipotent progenitor (MPP) subset that lacked self-renewal capacity but maintained multilineage differentiation potential. TdT induction on MPPs reflected a transcriptionally dynamic but uncommitted stage, characterized by low expression of lineage-associated genes. Single-cell CITE-seq indicated that multipotency in the TdT+ MPPs is associated with expression of the endothelial cell adhesion molecule ESAM. Stable and progressive upregulation of TdT defined the lymphoid developmental trajectory. Collectively, we here identify a new multipotent progenitor within the MPP4 compartment. Specification and commitment are defined by downregulation of ESAM which marks the progressive loss of alternative fates along all lineages.


Subject(s)
DNA Nucleotidylexotransferase , Hematopoietic Stem Cells , Multipotent Stem Cells , Animals , Cell Differentiation , Cell Lineage/genetics , DNA Nucleotidylexotransferase/genetics , DNA Nucleotidylexotransferase/metabolism , Hematopoietic Stem Cells/physiology , Mice , Mice, Transgenic
18.
Nat Immunol ; 23(11): 1628-1643, 2022 11.
Article in English | MEDLINE | ID: mdl-36316479

ABSTRACT

T cell differentiation requires Notch1 signaling. In the present study, we show that an enhancer upstream of Notch1 active in double-negative (DN) mouse thymocytes is responsible for raising Notch1 signaling intrathymically. This enhancer is required to expand multipotent progenitors intrathymically while delaying early differentiation until lineage restrictions have been established. Early thymic progenitors lacking the enhancer show accelerated differentiation through the DN stages and increased frequency of B, innate lymphoid (IL) and natural killer (NK) cell differentiation. Transcription regulators for T cell lineage restriction and commitment are expressed normally, but IL and NK cell gene expression persists after T cell lineage commitment and T cell receptor ß VDJ recombination, Cd3 expression and ß-selection have been impaired. This Notch1 enhancer is inactive in double-positive (DP) thymocytes. Its aberrant reactivation at this stage in Ikaros mutants is required for leukemogenesis. Thus, the DN-specific Notch1 enhancer harnesses the regulatory architecture of DN and DP thymocytes to achieve carefully orchestrated changes in Notch1 signaling required for early lineage restrictions and normal T cell differentiation.


Subject(s)
Immunity, Innate , Thymocytes , Mice , Animals , Thymocytes/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Lymphocytes/metabolism , Thymus Gland , Cell Differentiation/genetics , Cell Lineage/genetics
19.
Nat Immunol ; 23(5): 731-742, 2022 05.
Article in English | MEDLINE | ID: mdl-35523960

ABSTRACT

T cell specificity and function are linked during development, as MHC-II-specific TCR signals generate CD4 helper T cells and MHC-I-specific TCR signals generate CD8 cytotoxic T cells, but the basis remains uncertain. We now report that switching coreceptor proteins encoded by Cd4 and Cd8 gene loci functionally reverses the T cell immune system, generating CD4 cytotoxic and CD8 helper T cells. Such functional reversal reveals that coreceptor proteins promote the helper-lineage fate when encoded by Cd4, but promote the cytotoxic-lineage fate when encoded in Cd8-regardless of the coreceptor proteins each locus encodes. Thus, T cell lineage fate is determined by cis-regulatory elements in coreceptor gene loci and is not determined by the coreceptor proteins they encode, invalidating coreceptor signal strength as the basis of lineage fate determination. Moreover, we consider that evolution selected the particular coreceptor proteins that Cd4 and Cd8 gene loci encode to avoid generating functionally reversed T cells because they fail to promote protective immunity against environmental pathogens.


Subject(s)
CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Cell Differentiation , Cell Lineage/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Thymus Gland/metabolism
20.
Annu Rev Immunol ; 30: 707-31, 2012.
Article in English | MEDLINE | ID: mdl-22224760

ABSTRACT

T helper cell differentiation occurs in the context of the extracellular cytokine milieu evoked by diverse microbes and other pathogenic stimuli along with T cell receptor stimulation. The culmination of these signals results in specification of T helper lineages, which occurs through the combinatorial action of multiple transcription factors that establish distinctive transcriptomes. In this manner, inducible, but constitutively active, master regulators work in conjunction with factors such as the signal transducer and activator of transcriptions (STATs) that sense the extracellular environment. The acquisition of a distinctive transcriptome also depends on chromatin modifications that impact key cis elements as well as the changes in global genomic organization. Thus, signal transduction and epigenetics are linked in these processes of differentiation. In this review, recent advances in understanding T helper lineage specification and deciphering the action of transcription factors are summarized with emphasis on comprehensive views of the dynamic T cell epigenome.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Transcription, Genetic , Animals , Autoimmune Diseases/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Epigenomics , Humans , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Regulatory Elements, Transcriptional , T-Lymphocytes, Helper-Inducer/cytology , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL