Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.502
Filter
Add more filters

Publication year range
1.
Cell ; 187(18): 4926-4945.e22, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38986619

ABSTRACT

Posterior fossa group A (PFA) ependymoma is a lethal brain cancer diagnosed in infants and young children. The lack of driver events in the PFA linear genome led us to search its 3D genome for characteristic features. Here, we reconstructed 3D genomes from diverse childhood tumor types and uncovered a global topology in PFA that is highly reminiscent of stem and progenitor cells in a variety of human tissues. A remarkable feature exclusively present in PFA are type B ultra long-range interactions in PFAs (TULIPs), regions separated by great distances along the linear genome that interact with each other in the 3D nuclear space with surprising strength. TULIPs occur in all PFA samples and recur at predictable genomic coordinates, and their formation is induced by expression of EZHIP. The universality of TULIPs across PFA samples suggests a conservation of molecular principles that could be exploited therapeutically.


Subject(s)
Ependymoma , Ependymoma/genetics , Humans , Infratentorial Neoplasms/genetics , Infratentorial Neoplasms/pathology , Genome, Human , Infant , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Child , Male , Female
2.
Cell ; 187(13): 3338-3356.e30, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38810644

ABSTRACT

Suspended animation states allow organisms to survive extreme environments. The African turquoise killifish has evolved diapause as a form of suspended development to survive a complete drought. However, the mechanisms underlying the evolution of extreme survival states are unknown. To understand diapause evolution, we performed integrative multi-omics (gene expression, chromatin accessibility, and lipidomics) in the embryos of multiple killifish species. We find that diapause evolved by a recent remodeling of regulatory elements at very ancient gene duplicates (paralogs) present in all vertebrates. CRISPR-Cas9-based perturbations identify the transcription factors REST/NRSF and FOXOs as critical for the diapause gene expression program, including genes involved in lipid metabolism. Indeed, diapause shows a distinct lipid profile, with an increase in triglycerides with very-long-chain fatty acids. Our work suggests a mechanism for the evolution of complex adaptations and offers strategies to promote long-term survival by activating suspended animation programs in other species.


Subject(s)
Diapause , Animals , Biological Evolution , Diapause/genetics , Embryo, Nonmammalian/metabolism , Fundulidae/genetics , Fundulidae/metabolism , Gene Expression Regulation, Developmental , Killifishes/genetics , Killifishes/metabolism , Lipid Metabolism/genetics , Fish Proteins/genetics , Male , Female
3.
Cell ; 187(2): 446-463.e16, 2024 01 18.
Article in English | MEDLINE | ID: mdl-38242087

ABSTRACT

Treatment failure for the lethal brain tumor glioblastoma (GBM) is attributed to intratumoral heterogeneity and tumor evolution. We utilized 3D neuronavigation during surgical resection to acquire samples representing the whole tumor mapped by 3D spatial coordinates. Integrative tissue and single-cell analysis revealed sources of genomic, epigenomic, and microenvironmental intratumoral heterogeneity and their spatial patterning. By distinguishing tumor-wide molecular features from those with regional specificity, we inferred GBM evolutionary trajectories from neurodevelopmental lineage origins and initiating events such as chromothripsis to emergence of genetic subclones and spatially restricted activation of differential tumor and microenvironmental programs in the core, periphery, and contrast-enhancing regions. Our work depicts GBM evolution and heterogeneity from a 3D whole-tumor perspective, highlights potential therapeutic targets that might circumvent heterogeneity-related failures, and establishes an interactive platform enabling 360° visualization and analysis of 3D spatial patterns for user-selected genes, programs, and other features across whole GBM tumors.


Subject(s)
Brain Neoplasms , Glioblastoma , Models, Biological , Humans , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Epigenomics , Genomics , Glioblastoma/genetics , Glioblastoma/pathology , Single-Cell Analysis , Tumor Microenvironment , Genetic Heterogeneity
4.
Cell ; 185(23): 4448-4464.e17, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36272405

ABSTRACT

The recent development of spatial omics methods has enabled single-cell profiling of the transcriptome and 3D genome organization with high spatial resolution. Expanding the repertoire of spatial omics tools, a spatially resolved single-cell epigenomics method will accelerate understanding of the spatial regulation of cell and tissue functions. Here, we report a method for spatially resolved epigenomic profiling of single cells using in situ tagmentation and transcription followed by multiplexed imaging. We demonstrated the ability to profile histone modifications marking active promoters, putative enhancers, and silent promoters in individual cells, and generated high-resolution spatial atlas of hundreds of active promoters and putative enhancers in embryonic and adult mouse brains. Our results suggested putative promoter-enhancer pairs and enhancer hubs regulating developmentally important genes. We envision this approach will be generally applicable to spatial profiling of epigenetic modifications and DNA-binding proteins, advancing our understanding of how gene expression is spatiotemporally regulated by the epigenome.


Subject(s)
Epigenomics , Histone Code , Mice , Animals , Promoter Regions, Genetic , Epigenesis, Genetic , Transcriptome , Enhancer Elements, Genetic , Chromatin
5.
Cell ; 184(11): 3041-3055.e21, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33964211

ABSTRACT

cis-regulatory elements (CREs) encode the genomic blueprints of spatiotemporal gene expression programs enabling highly specialized cell functions. Using single-cell genomics in six maize organs, we determined the cis- and trans-regulatory factors defining diverse cell identities and coordinating chromatin organization by profiling transcription factor (TF) combinatorics, identifying TFs with non-cell-autonomous activity, and uncovering TFs underlying higher-order chromatin interactions. Cell-type-specific CREs were enriched for enhancer activity and within unmethylated long terminal repeat retrotransposons. Moreover, we found cell-type-specific CREs are hotspots for phenotype-associated genetic variants and were targeted by selection during modern maize breeding, highlighting the biological implications of this CRE atlas. Through comparison of maize and Arabidopsis thaliana developmental trajectories, we identified TFs and CREs with conserved and divergent chromatin dynamics, showcasing extensive evolution of gene regulatory networks. In addition to this rich dataset, we developed single-cell analysis software, Socrates, which can be used to understand cis-regulatory variation in any species.


Subject(s)
Gene Expression Regulation, Plant/genetics , Regulatory Elements, Transcriptional/genetics , Zea mays/genetics , Arabidopsis/genetics , Gene Expression/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Plant/physiology , Gene Regulatory Networks/genetics , Genome , Genomics , Regulatory Elements, Transcriptional/physiology , Single-Cell Analysis , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome/genetics
6.
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
7.
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
8.
Cell ; 181(2): 382-395.e21, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32246942

ABSTRACT

Multiple sclerosis (MS) is an autoimmune disease characterized by attack on oligodendrocytes within the central nervous system (CNS). Despite widespread use of immunomodulatory therapies, patients may still face progressive disability because of failure of myelin regeneration and loss of neurons, suggesting additional cellular pathologies. Here, we describe a general approach for identifying specific cell types in which a disease allele exerts a pathogenic effect. Applying this approach to MS risk loci, we pinpoint likely pathogenic cell types for 70%. In addition to T cell loci, we unexpectedly identified myeloid- and CNS-specific risk loci, including two sites that dysregulate transcriptional pause release in oligodendrocytes. Functional studies demonstrated inhibition of transcriptional elongation is a dominant pathway blocking oligodendrocyte maturation. Furthermore, pause release factors are frequently dysregulated in MS brain tissue. These data implicate cell-intrinsic aberrations outside of the immune system and suggest new avenues for therapeutic development. VIDEO ABSTRACT.


Subject(s)
Cell Communication/genetics , Disease/genetics , Oligodendroglia/metabolism , Animals , Brain/metabolism , Central Nervous System/metabolism , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Humans , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Multiple Sclerosis/physiopathology , Myelin Sheath/metabolism , Neurons/metabolism , Oligodendroglia/physiology , Risk Factors
9.
Cell ; 176(4): 897-912.e20, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30686579

ABSTRACT

A complete chart of cis-regulatory elements and their dynamic activity is necessary to understand the transcriptional basis of differentiation and function of an organ system. We generated matched epigenome and transcriptome measurements in 86 primary cell types that span the mouse immune system and its differentiation cascades. This breadth of data enable variance components analysis that suggests that genes fall into two distinct classes, controlled by either enhancer- or promoter-driven logic, and multiple regression that connects genes to the enhancers that regulate them. Relating transcription factor (TF) expression to the genome-wide accessibility of their binding motifs classifies them as predominantly openers or closers of local chromatin accessibility, pinpointing specific cis-regulatory elements where binding of given TFs is likely functionally relevant, validated by chromatin immunoprecipitation sequencing (ChIP-seq). Overall, this cis-regulatory atlas provides a trove of information on transcriptional regulation through immune differentiation and a foundational scaffold to define key regulatory events throughout the immunological genome.


Subject(s)
Immune System/immunology , Immune System/metabolism , Regulatory Elements, Transcriptional/genetics , Animals , Binding Sites/genetics , Chromatin , Chromatin Immunoprecipitation/methods , Enhancer Elements, Genetic/genetics , Epigenomics/methods , Gene Expression Regulation/genetics , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Transcription Factors/metabolism , Transcriptome/genetics
10.
Cell ; 176(1-2): 361-376.e17, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30580963

ABSTRACT

Here, we present Perturb-ATAC, a method that combines multiplexed CRISPR interference or knockout with genome-wide chromatin accessibility profiling in single cells based on the simultaneous detection of CRISPR guide RNAs and open chromatin sites by assay of transposase-accessible chromatin with sequencing (ATAC-seq). We applied Perturb-ATAC to transcription factors (TFs), chromatin-modifying factors, and noncoding RNAs (ncRNAs) in ∼4,300 single cells, encompassing more than 63 genotype-phenotype relationships. Perturb-ATAC in human B lymphocytes uncovered regulators of chromatin accessibility, TF occupancy, and nucleosome positioning and identified a hierarchy of TFs that govern B cell state, variation, and disease-associated cis-regulatory elements. Perturb-ATAC in primary human epidermal cells revealed three sequential modules of cis-elements that specify keratinocyte fate. Combinatorial deletion of all pairs of these TFs uncovered their epistatic relationships and highlighted genomic co-localization as a basis for synergistic interactions. Thus, Perturb-ATAC is a powerful strategy to dissect gene regulatory networks in development and disease.


Subject(s)
Epigenomics/methods , Gene Regulatory Networks/genetics , Single-Cell Analysis/methods , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly/physiology , Clustered Regularly Interspaced Short Palindromic Repeats/physiology , Gene Regulatory Networks/physiology , High-Throughput Nucleotide Sequencing/methods , Humans , Sequence Analysis, DNA/methods , Transcription Factors/metabolism
11.
Immunity ; 57(9): 2232-2250.e10, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39137779

ABSTRACT

Due to its stimulatory potential for immunomodulatory CD4+ regulatory T (Treg) cells, low-dose interleukin-2 (IL-2) immunotherapy has gained considerable attention for the treatment of autoimmune diseases. In this investigator-initiated single-arm non-placebo-controlled phase-2 clinical trial of low-dose IL-2 immunotherapy in systemic lupus erythematosus (SLE) patients, we generated a comprehensive atlas of in vivo human immune responses to low-dose IL-2. We performed an in-depth study of circulating and cutaneous immune cells by imaging mass cytometry, high-parameter flow cytometry, transcriptomics, and targeted serum proteomics. Low-dose IL-2 stimulated various circulating immune cells, including Treg cells with a skin-homing phenotype that appeared in the skin of SLE patients in close interaction with endothelial cells. Analysis of surface proteins and transcriptomes revealed different IL-2-driven Treg cell activation programs, including gut-homing CD38+, skin-homing HLA-DR+, and highly proliferative inflammation-homing CD38+ HLA-DR+ Treg cells. Collectively, these data define the distinct human Treg cell subsets that are responsive to IL-2 immunotherapy.


Subject(s)
Immunotherapy , Interleukin-2 , Lupus Erythematosus, Systemic , Skin , T-Lymphocytes, Regulatory , Humans , T-Lymphocytes, Regulatory/immunology , Interleukin-2/immunology , Skin/immunology , Immunotherapy/methods , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/therapy , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Lymphocyte Activation/immunology , Female , Adult , Male
12.
Immunity ; 57(1): 171-187.e14, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38198850

ABSTRACT

Immune responses are tightly regulated yet highly variable between individuals. To investigate human population variation of trained immunity, we immunized healthy individuals with Bacillus Calmette-Guérin (BCG). This live-attenuated vaccine induces not only an adaptive immune response against tuberculosis but also triggers innate immune activation and memory that are indicative of trained immunity. We established personal immune profiles and chromatin accessibility maps over a 90-day time course of BCG vaccination in 323 individuals. Our analysis uncovered genetic and epigenetic predictors of baseline immunity and immune response. BCG vaccination enhanced the innate immune response specifically in individuals with a dormant immune state at baseline, rather than providing a general boost of innate immunity. This study advances our understanding of BCG's heterologous immune-stimulatory effects and trained immunity in humans. Furthermore, it highlights the value of epigenetic cell states for connecting immune function with genotype and the environment.


Subject(s)
BCG Vaccine , Trained Immunity , Humans , Multiomics , Vaccination , Epigenesis, Genetic
13.
Cell ; 173(6): 1535-1548.e16, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29706549

ABSTRACT

Human hematopoiesis involves cellular differentiation of multipotent cells into progressively more lineage-restricted states. While the chromatin accessibility landscape of this process has been explored in defined populations, single-cell regulatory variation has been hidden by ensemble averaging. We collected single-cell chromatin accessibility profiles across 10 populations of immunophenotypically defined human hematopoietic cell types and constructed a chromatin accessibility landscape of human hematopoiesis to characterize differentiation trajectories. We find variation consistent with lineage bias toward different developmental branches in multipotent cell types. We observe heterogeneity within common myeloid progenitors (CMPs) and granulocyte-macrophage progenitors (GMPs) and develop a strategy to partition GMPs along their differentiation trajectory. Furthermore, we integrated single-cell RNA sequencing (scRNA-seq) data to associate transcription factors to chromatin accessibility changes and regulatory elements to target genes through correlations of expression and regulatory element accessibility. Overall, this work provides a framework for integrative exploration of complex regulatory dynamics in a primary human tissue at single-cell resolution.


Subject(s)
Chromatin/chemistry , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Single-Cell Analysis , Transcription Factors/metabolism , Animals , Cell Differentiation , Cell Lineage , Epigenesis, Genetic , Epigenomics , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Humans , Myeloid Progenitor Cells/cytology , Principal Component Analysis , Regulatory Sequences, Nucleic Acid , Sequence Analysis, RNA , Transcriptome
14.
Cell ; 174(5): 1309-1324.e18, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30078704

ABSTRACT

We applied a combinatorial indexing assay, sci-ATAC-seq, to profile genome-wide chromatin accessibility in ∼100,000 single cells from 13 adult mouse tissues. We identify 85 distinct patterns of chromatin accessibility, most of which can be assigned to cell types, and ∼400,000 differentially accessible elements. We use these data to link regulatory elements to their target genes, to define the transcription factor grammar specifying each cell type, and to discover in vivo correlates of heterogeneity in accessibility within cell types. We develop a technique for mapping single cell gene expression data to single-cell chromatin accessibility data, facilitating the comparison of atlases. By intersecting mouse chromatin accessibility with human genome-wide association summary statistics, we identify cell-type-specific enrichments of the heritability signal for hundreds of complex traits. These data define the in vivo landscape of the regulatory genome for common mammalian cell types at single-cell resolution.


Subject(s)
Chromatin/chemistry , Single-Cell Analysis/methods , Animals , Cluster Analysis , Epigenesis, Genetic , Epigenomics , Gene Expression Regulation , Genome, Human , Genome-Wide Association Study , Humans , Male , Mammals , Mice , Mice, Inbred C57BL , Transcription Factors
15.
Immunity ; 56(9): 2152-2171.e13, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37582369

ABSTRACT

Microglia phenotypes are highly regulated by the brain environment, but the transcriptional networks that specify the maturation of human microglia are poorly understood. Here, we characterized stage-specific transcriptomes and epigenetic landscapes of fetal and postnatal human microglia and acquired corresponding data in induced pluripotent stem cell (iPSC)-derived microglia, in cerebral organoids, and following engraftment into humanized mice. Parallel development of computational approaches that considered transcription factor (TF) co-occurrence and enhancer activity allowed prediction of shared and state-specific gene regulatory networks associated with fetal and postnatal microglia. Additionally, many features of the human fetal-to-postnatal transition were recapitulated in a time-dependent manner following the engraftment of iPSC cells into humanized mice. These data and accompanying computational approaches will facilitate further efforts to elucidate mechanisms by which human microglia acquire stage- and disease-specific phenotypes.


Subject(s)
Induced Pluripotent Stem Cells , Microglia , Humans , Mice , Animals , Gene Regulatory Networks , Brain , Gene Expression Regulation
16.
Cell ; 170(3): 522-533.e15, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28753427

ABSTRACT

Genome-wide association studies (GWASs) implicate the PHACTR1 locus (6p24) in risk for five vascular diseases, including coronary artery disease, migraine headache, cervical artery dissection, fibromuscular dysplasia, and hypertension. Through genetic fine mapping, we prioritized rs9349379, a common SNP in the third intron of the PHACTR1 gene, as the putative causal variant. Epigenomic data from human tissue revealed an enhancer signature at rs9349379 exclusively in aorta, suggesting a regulatory function for this SNP in the vasculature. CRISPR-edited stem cell-derived endothelial cells demonstrate rs9349379 regulates expression of endothelin 1 (EDN1), a gene located 600 kb upstream of PHACTR1. The known physiologic effects of EDN1 on the vasculature may explain the pattern of risk for the five associated diseases. Overall, these data illustrate the integration of genetic, phenotypic, and epigenetic analysis to identify the biologic mechanism by which a common, non-coding variant can distally regulate a gene and contribute to the pathogenesis of multiple vascular diseases.


Subject(s)
Coronary Artery Disease/genetics , Endothelin-1/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Vascular Diseases/genetics , Acetylation , Cells, Cultured , Chromatin/metabolism , Chromosome Mapping , Chromosomes, Human, Pair 6 , Endothelial Cells/cytology , Endothelin-1/blood , Epigenomics , Gene Editing , Gene Expression , Genome-Wide Association Study , Histones/metabolism , Humans , Muscle, Smooth, Vascular/cytology
17.
Immunity ; 55(8): 1402-1413.e4, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35882235

ABSTRACT

The differentiation of innate lymphoid cells (ILCs) from hematopoietic stem cells needs to go through several multipotent progenitor stages. However, it remains unclear whether the fates of multipotent progenitors are predefined by epigenetic states. Here, we report the identification of distinct accessible chromatin regions in all lymphoid progenitors (ALPs), EILPs, and ILC precursors (ILCPs). Single-cell MNase-seq analyses revealed that EILPs contained distinct subpopulations epigenetically primed toward either dendritic cell lineages or ILC lineages. We found that TCF-1 and GATA3 co-bound to the lineage-defining sites for ILCs (LDS-Is), whereas PU.1 binding was enriched in the LDSs for alternative dendritic cells (LDS-As). TCF-1 and GATA3 were indispensable for the epigenetic priming of LDSs at the EILP stage. Our results suggest that the multipotency of progenitor cells is defined by the existence of a heterogeneous population of cells epigenetically primed for distinct downstream lineages, which are regulated by key transcription factors.


Subject(s)
Immunity, Innate , Lymphocytes , Cell Differentiation , Cell Lineage , Epigenesis, Genetic , Hematopoietic Stem Cells
18.
Mol Cell ; 83(1): 121-138.e7, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36521490

ABSTRACT

Cell cycle (CC) facilitates cell division via robust, cyclical gene expression. Protective immunity requires the expansion of pathogen-responsive cell types, but whether CC confers unique gene expression programs that direct the subsequent immunological response remains unclear. Here, we demonstrate that single macrophages (MFs) adopt different plasticity states in CC, which leads to heterogeneous cytokine-induced polarization, priming, and repolarization programs. Specifically, MF plasticity to interferon gamma (IFNG) is substantially reduced during S-G2/M, whereas interleukin 4 (IL-4) induces S-G2/M-biased gene expression, mediated by CC-biased enhancers. Additionally, IL-4 polarization shifts the CC-phase distribution of MFs toward the G2/M phase, providing a subpopulation-specific mechanism for IL-4-induced, dampened IFNG responsiveness. Finally, we demonstrate CC-dependent MF responses in murine and human disease settings in vivo, including Th2-driven airway inflammation and pulmonary fibrosis, where MFs express an S-G2/M-biased tissue remodeling gene program. Therefore, MF inflammatory and regenerative responses are gated by CC in a cyclical, phase-dependent manner.


Subject(s)
Chromatin , Interleukin-4 , Humans , Mice , Animals , Interleukin-4/genetics , Interleukin-4/pharmacology , Chromatin/genetics , Chromatin/metabolism , Macrophages/metabolism , Interferon-gamma/genetics , Interferon-gamma/pharmacology , Cell Cycle/genetics , Cell Division
19.
Immunity ; 51(2): 241-257.e9, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31303399

ABSTRACT

Cytokine tumor necrosis factor (TNF)-mediated macrophage polarization is important for inflammatory disease pathogenesis, but the mechanisms regulating polarization are not clear. We performed transcriptomic and epigenomic analysis of the TNF response in primary human macrophages and revealed late-phase activation of SREBP2, the master regulator of cholesterol biosynthesis genes. TNF stimulation extended the genomic profile of SREBP2 occupancy to include binding to and activation of inflammatory and interferon response genes independently of its functions in sterol metabolism. Genetic ablation of SREBP function shifted the balance of macrophage polarization from an inflammatory to a reparative phenotype in peritonitis and skin wound healing models. Genetic ablation of SREBP activity in myeloid cells or topical pharmacological inhibition of SREBP improved skin wound healing under homeostatic and chronic inflammatory conditions. Our results identify a function and mechanism of action for SREBPs in augmenting TNF-induced macrophage activation and inflammation and open therapeutic avenues for promoting wound repair.


Subject(s)
Inflammation/metabolism , Macrophages/immunology , Peritonitis/metabolism , Receptors, G-Protein-Coupled/metabolism , Skin Diseases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Epigenomics , Female , Humans , Macrophage Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , RNA, Small Interfering/genetics , Receptors, G-Protein-Coupled/genetics , Transcriptome , Wound Healing
20.
Immunity ; 48(1): 75-90.e6, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29343442

ABSTRACT

The molecular basis of signal-dependent transcriptional activation has been extensively studied in macrophage polarization, but our understanding remains limited regarding the molecular determinants of repression. Here we show that IL-4-activated STAT6 transcription factor is required for the direct transcriptional repression of a large number of genes during in vitro and in vivo alternative macrophage polarization. Repression results in decreased lineage-determining transcription factor, p300, and RNA polymerase II binding followed by reduced enhancer RNA expression, H3K27 acetylation, and chromatin accessibility. The repressor function of STAT6 is HDAC3 dependent on a subset of IL-4-repressed genes. In addition, STAT6-repressed enhancers show extensive overlap with the NF-κB p65 cistrome and exhibit decreased responsiveness to lipopolysaccharide after IL-4 stimulus on a subset of genes. As a consequence, macrophages exhibit diminished inflammasome activation, decreased IL-1ß production, and pyroptosis. Thus, the IL-4-STAT6 signaling pathway establishes an alternative polarization-specific epigenenomic signature resulting in dampened macrophage responsiveness to inflammatory stimuli.


Subject(s)
Interleukin-4/metabolism , Macrophages/metabolism , STAT6 Transcription Factor/metabolism , Animals , Blotting, Western , Cell Line , Enhancer Elements, Genetic , Flow Cytometry , Gene Expression Regulation , Inflammasomes/metabolism , Laser Scanning Cytometry , Lipopolysaccharides/pharmacology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Pyroptosis/genetics , Signal Transduction/genetics , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL