Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 186(24): 5269-5289.e22, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37995656

ABSTRACT

A generic level of chromatin organization generated by the interplay between cohesin and CTCF suffices to limit promiscuous interactions between regulatory elements, but a lineage-specific chromatin assembly that supersedes these constraints is required to configure the genome to guide gene expression changes that drive faithful lineage progression. Loss-of-function approaches in B cell precursors show that IKAROS assembles interactions across megabase distances in preparation for lymphoid development. Interactions emanating from IKAROS-bound enhancers override CTCF-imposed boundaries to assemble lineage-specific regulatory units built on a backbone of smaller invariant topological domains. Gain of function in epithelial cells confirms IKAROS' ability to reconfigure chromatin architecture at multiple scales. Although the compaction of the Igκ locus required for genome editing represents a function of IKAROS unique to lymphocytes, the more general function to preconfigure the genome to support lineage-specific gene expression and suppress activation of extra-lineage genes provides a paradigm for lineage restriction.


Subject(s)
Chromatin , Genome , B-Lymphocytes/metabolism , CCCTC-Binding Factor/metabolism , Chromatin/metabolism , Chromatin Assembly and Disassembly , Humans , Animals , Mice
2.
Cell ; 183(5): 1340-1353.e16, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33096020

ABSTRACT

The contribution of CD4+ T cells to protective or pathogenic immune responses to SARS-CoV-2 infection remains unknown. Here, we present single-cell transcriptomic analysis of >100,000 viral antigen-reactive CD4+ T cells from 40 COVID-19 patients. In hospitalized patients compared to non-hospitalized patients, we found increased proportions of cytotoxic follicular helper cells and cytotoxic T helper (TH) cells (CD4-CTLs) responding to SARS-CoV-2 and reduced proportion of SARS-CoV-2-reactive regulatory T cells (TREG). Importantly, in hospitalized COVID-19 patients, a strong cytotoxic TFH response was observed early in the illness, which correlated negatively with antibody levels to SARS-CoV-2 spike protein. Polyfunctional TH1 and TH17 cell subsets were underrepresented in the repertoire of SARS-CoV-2-reactive CD4+ T cells compared to influenza-reactive CD4+ T cells. Together, our analyses provide insights into the gene expression patterns of SARS-CoV-2-reactive CD4+ T cells in distinct disease severities.


Subject(s)
COVID-19/immunology , SARS-CoV-2/genetics , T Follicular Helper Cells/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Transcriptome , Adult , Aged , Aged, 80 and over , Antibodies, Viral/blood , Antibodies, Viral/immunology , CD4 Lymphocyte Count , COVID-19/epidemiology , COVID-19/virology , Cohort Studies , England/epidemiology , Female , Humans , Male , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Severity of Illness Index , Single-Cell Analysis/methods , Spike Glycoprotein, Coronavirus/immunology
3.
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
4.
Cell ; 176(4): 816-830.e18, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30595451

ABSTRACT

The temporal order of DNA replication (replication timing [RT]) is highly coupled with genome architecture, but cis-elements regulating either remain elusive. We created a series of CRISPR-mediated deletions and inversions of a pluripotency-associated topologically associating domain (TAD) in mouse ESCs. CTCF-associated domain boundaries were dispensable for RT. CTCF protein depletion weakened most TAD boundaries but had no effect on RT or A/B compartmentalization genome-wide. By contrast, deletion of three intra-TAD CTCF-independent 3D contact sites caused a domain-wide early-to-late RT shift, an A-to-B compartment switch, weakening of TAD architecture, and loss of transcription. The dispensability of TAD boundaries and the necessity of these "early replication control elements" (ERCEs) was validated by deletions and inversions at additional domains. Our results demonstrate that discrete cis-regulatory elements orchestrate domain-wide RT, A/B compartmentalization, TAD architecture, and transcription, revealing fundamental principles linking genome structure and function.


Subject(s)
DNA Replication Timing/physiology , DNA Replication/genetics , DNA Replication/physiology , Animals , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Chromatin , DNA/genetics , DNA Replication Timing/genetics , Embryonic Stem Cells , Enhancer Elements, Genetic/genetics , Mammals/genetics , Mammals/metabolism , Mice , Repressor Proteins/metabolism , Spatio-Temporal Analysis
5.
Nat Immunol ; 22(4): 485-496, 2021 04.
Article in English | MEDLINE | ID: mdl-33767426

ABSTRACT

Evasion of host immunity is a hallmark of cancer; however, mechanisms linking oncogenic mutations and immune escape are incompletely understood. Through loss-of-function screening of 1,001 tumor suppressor genes, we identified death-associated protein kinase 3 (DAPK3) as a previously unrecognized driver of anti-tumor immunity through the stimulator of interferon genes (STING) pathway of cytosolic DNA sensing. Loss of DAPK3 expression or kinase activity impaired STING activation and interferon (IFN)-ß-stimulated gene induction. DAPK3 deficiency in IFN-ß-producing tumors drove rapid growth and reduced infiltration of CD103+CD8α+ dendritic cells and cytotoxic lymphocytes, attenuating the response to cancer chemo-immunotherapy. Mechanistically, DAPK3 coordinated post-translational modification of STING. In unstimulated cells, DAPK3 inhibited STING K48-linked poly-ubiquitination and proteasome-mediated degradation. After cGAMP stimulation, DAPK3 was required for STING K63-linked poly-ubiquitination and STING-TANK-binding kinase 1 interaction. Comprehensive phospho-proteomics uncovered a DAPK3-specific phospho-site on the E3 ligase LMO7, critical for LMO7-STING interaction and STING K63-linked poly-ubiquitination. Thus, DAPK3 is an essential kinase for STING activation that drives tumor-intrinsic innate immunity and tumor immune surveillance.


Subject(s)
Death-Associated Protein Kinases/metabolism , Human Umbilical Vein Endothelial Cells/enzymology , Immunity, Innate , Interferon-beta/metabolism , Membrane Proteins/metabolism , Neoplasms/enzymology , Tumor Escape , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Death-Associated Protein Kinases/genetics , Female , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunity, Innate/drug effects , Interferon-beta/genetics , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/immunology , Phosphorylation , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Escape/drug effects , Ubiquitination
6.
Nat Immunol ; 22(8): 1052-1063, 2021 08.
Article in English | MEDLINE | ID: mdl-34168370

ABSTRACT

Immune-checkpoint blockade (ICB) has shown remarkable clinical success in boosting antitumor immunity. However, the breadth of its cellular targets and specific mode of action remain elusive. We find that tumor-infiltrating follicular regulatory T (TFR) cells are prevalent in tumor tissues of several cancer types. They are primarily located within tertiary lymphoid structures and exhibit superior suppressive capacity and in vivo persistence as compared with regulatory T cells, with which they share a clonal and developmental relationship. In syngeneic tumor models, anti-PD-1 treatment increases the number of tumor-infiltrating TFR cells. Both TFR cell deficiency and the depletion of TFR cells with anti-CTLA-4 before anti-PD-1 treatment improve tumor control in mice. Notably, in a cohort of 271 patients with melanoma, treatment with anti-CTLA-4 followed by anti-PD-1 at progression was associated with better a survival outcome than monotherapy with anti-PD-1 or anti-CTLA-4, anti-PD-1 followed by anti-CTLA-4 at progression or concomitant combination therapy.


Subject(s)
CTLA-4 Antigen/antagonists & inhibitors , Immune Checkpoint Inhibitors/therapeutic use , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes, Regulatory/immunology , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred C57BL , T Follicular Helper Cells/immunology , Tumor Microenvironment/immunology
7.
Nature ; 605(7911): 741-746, 2022 05.
Article in English | MEDLINE | ID: mdl-35508656

ABSTRACT

Phosphoinositide 3-kinase δ (PI3Kδ) has a key role in lymphocytes, and inhibitors that target this PI3K have been approved for treatment of B cell malignancies1-3. Although studies in mouse models of solid tumours have demonstrated that PI3Kδ inhibitors (PI3Kδi) can induce anti-tumour immunity4,5, its effect on solid tumours in humans remains unclear. Here we assessed the effects of the PI3Kδi AMG319 in human patients with head and neck cancer in a neoadjuvant, double-blind, placebo-controlled randomized phase II trial (EudraCT no. 2014-004388-20). PI3Kδ inhibition decreased the number of tumour-infiltrating regulatory T (Treg) cells and enhanced the cytotoxic potential of tumour-infiltrating T cells. At the tested doses of AMG319, immune-related adverse events (irAEs) required treatment to be discontinued in 12 out of 21 of patients treated with AMG319, suggestive of systemic effects on Treg cells. Accordingly, in mouse models, PI3Kδi decreased the number of Treg cells systemically and caused colitis. Single-cell RNA-sequencing analysis revealed a PI3Kδi-driven loss of tissue-resident colonic ST2 Treg cells, accompanied by expansion of pathogenic T helper 17 (TH17) and type 17 CD8+ T (TC17) cells, which probably contributed to toxicity; this points towards a specific mode of action for the emergence of irAEs. A modified treatment regimen with intermittent dosing of PI3Kδi in mouse models led to a significant decrease in tumour growth without inducing pathogenic T cells in colonic tissue, indicating that alternative dosing regimens might limit toxicity.


Subject(s)
Antineoplastic Agents , Head and Neck Neoplasms , Adenosine/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Disease Models, Animal , Head and Neck Neoplasms/drug therapy , Humans , Immunotherapy , Mice , Phosphatidylinositol 3-Kinases , Quinolines/therapeutic use , T-Lymphocytes, Regulatory
8.
Mol Cell ; 71(6): 956-972.e9, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30146317

ABSTRACT

Gene regulation requires selective targeting of DNA regulatory enhancers over megabase distances. Here we show that Evf2, a cloud-forming Dlx5/6 ultraconserved enhancer (UCE) lncRNA, simultaneously localizes to activated (Umad1, 1.6 Mb distant) and repressed (Akr1b8, 27 Mb distant) chr6 target genes, precisely regulating UCE-gene distances and cohesin binding in mouse embryonic forebrain GABAergic interneurons (INs). Transgene expression of Evf2 activates Lsm8 (12 Mb distant) but fails to repress Akr1b8, supporting trans activation and long-range cis repression. Through both short-range (Dlx6 antisense) and long-range (Akr1b8) repression, the Evf2-5'UCE links homeodomain and mevalonate pathway-regulated enhancers to IN diversity. The Evf2-3' end is required for long-range activation but dispensable for RNA cloud localization, functionally dividing the RNA into 3'-activator and 5'UCE repressor and targeting regions. Together, these results support that Evf2 selectively regulates UCE interactions with multi-megabase distant genes through complex effects on chromosome topology, linking lncRNA-dependent topological and transcriptional control with interneuron diversity and seizure susceptibility.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/genetics , Prosencephalon/embryology , Alcohol Oxidoreductases/genetics , Animals , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Conserved Sequence , Enhancer Elements, Genetic/genetics , Homeodomain Proteins/physiology , Interneurons/physiology , Mice , Neurogenesis/genetics , Neurogenesis/physiology , RNA, Long Noncoding/genetics , Transcription Factors , Cohesins
9.
Genome Res ; 31(5): 834-851, 2021 05.
Article in English | MEDLINE | ID: mdl-33906962

ABSTRACT

Toxoplasma gondii is a useful model for intracellular parasitism given its ease of culture in the laboratory and genomic resources. However, as for many other eukaryotes, the T. gondii genome contains hundreds of sequence gaps owing to repetitive and/or unclonable sequences that disrupt the assembly process. Here, we use the Oxford Nanopore Minion platform to generate near-complete de novo genome assemblies for multiple strains of T. gondii and its near relative, N. caninum We significantly improved T. gondii genome contiguity (average N50 of ∼6.6 Mb) and added ∼2 Mb of newly assembled sequence. For all of the T. gondii strains that we sequenced (RH, ME49, CTG, II×III progeny clones CL13, S27, S21, S26, and D3X1), the largest contig ranged in size between 11.9 and 12.1 Mb in size, which is larger than any previously reported T. gondii chromosome, and found to be due to a consistent fusion of Chromosomes VIIb and VIII. These data were validated by mapping existing T. gondii ME49 Hi-C data to our assembly, providing parallel lines of evidence that the T. gondii karyotype consists of 13, rather than 14, chromosomes. By using this technology, we also resolved hundreds of tandem repeats of varying lengths, including in well-known host-targeting effector loci like rhoptry protein 5 (ROP5) and ROP38 Finally, when we compared T. gondii with N. caninum, we found that although the 13-chromosome karyotype was conserved, extensive, previously unappreciated chromosome-scale rearrangements had occurred in T. gondii and N. caninum since their most recent common ancestry.


Subject(s)
Toxoplasma , DNA Copy Number Variations , Genome , Karyotype , Sequence Analysis, DNA , Toxoplasma/genetics
10.
Genome Res ; 31(4): 659-676, 2021 04.
Article in English | MEDLINE | ID: mdl-33674349

ABSTRACT

Systemic lupus erythematosus (SLE) is an incurable autoimmune disease disproportionately affecting women. A major obstacle in finding targeted therapies for SLE is its remarkable heterogeneity in clinical manifestations as well as in the involvement of distinct cell types. To identify cell-specific targets as well as cross-correlation relationships among expression programs of different cell types, we here analyze six major circulating immune cell types from SLE patient blood. Our results show that presence of an interferon response signature stratifies patients into two distinct groups (IFNneg vs. IFNpos). Comparing these two groups using differential gene expression and differential gene coexpression analysis, we prioritize a relatively small list of genes from classical monocytes including two known immune modulators: TNFSF13B/BAFF (target of belimumab, an approved therapeutic for SLE) and IL1RN (the basis of anakinra, a therapeutic for rheumatoid arthritis). We then develop a multi-cell type extension of the weighted gene coexpression network analysis (WGCNA) framework, termed mWGCNA. Applying mWGCNA to RNA-seq data from six sorted immune cell populations (15 SLE, 10 healthy donors), we identify a coexpression module with interferon-stimulated genes (ISGs) among all cell types and a cross-cell type correlation linking expression of specific T helper cell markers to B cell response as well as to TNFSF13B expression from myeloid cells, all of which in turn correlates with disease severity of IFNpos patients. Our results demonstrate the power of a hypothesis-free and data-driven approach to discover drug targets and to reveal novel cross-correlation across cell types in SLE with implications for other autoimmune diseases.


Subject(s)
Gene Regulatory Networks , Interferons , Lupus Erythematosus, Systemic , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Humans , Interferons/genetics , Interferons/immunology , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Monocytes/immunology , Monocytes/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
11.
Development ; 148(6)2021 03 15.
Article in English | MEDLINE | ID: mdl-33593819

ABSTRACT

The Evf2 long non-coding RNA directs Dlx5/6 ultraconserved enhancer(UCE)-intrachromosomal interactions, regulating genes across a 27 Mb region on chromosome 6 in mouse developing forebrain. Here, we show that Evf2 long-range gene repression occurs through multi-step mechanisms involving the transcription factor Sox2. Evf2 directly interacts with Sox2, antagonizing Sox2 activation of Dlx5/6UCE, and recruits Sox2 to the Dlx5/6eii shadow enhancer and key Dlx5/6UCE interaction sites. Sox2 directly interacts with Dlx1 and Smarca4, as part of the Evf2 ribonucleoprotein complex, forming spherical subnuclear domains (protein pools, PPs). Evf2 targets Sox2 PPs to one long-range repressed target gene (Rbm28), at the expense of another (Akr1b8). Evf2 and Sox2 shift Dlx5/6UCE interactions towards Rbm28, linking Evf2/Sox2 co-regulated topological control and gene repression. We propose a model that distinguishes Evf2 gene repression mechanisms at Rbm28 (Dlx5/6UCE position) and Akr1b8 (limited Sox2 availability). Genome-wide control of RNPs (Sox2, Dlx and Smarca4) shows that co-recruitment influences Sox2 DNA binding. Together, these data suggest that Evf2 organizes a Sox2 PP subnuclear domain and, through Sox2-RNP sequestration and recruitment, regulates chromosome 6 long-range UCE targeting and activity with genome-wide consequences.


Subject(s)
Chromosomes, Mammalian/genetics , Gene Expression Regulation, Developmental , Prosencephalon/metabolism , RNA, Long Noncoding/genetics , SOXB1 Transcription Factors/genetics , Animals , DNA Helicases/genetics , DNA Helicases/metabolism , Enhancer Elements, Genetic/genetics , Fluorescent Antibody Technique/methods , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , In Situ Hybridization, Fluorescence/methods , Mice, Knockout , Mice, Transgenic , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Prosencephalon/embryology , Protein Binding , RNA, Long Noncoding/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , SOXB1 Transcription Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Bioinformatics ; 39(10)2023 10 03.
Article in English | MEDLINE | ID: mdl-37740957

ABSTRACT

MOTIVATION: With the wide availability of single-cell RNA-seq (scRNA-seq) technology, population-scale scRNA-seq datasets across multiple individuals and time points are emerging. While the initial investigations of these datasets tend to focus on standard analysis of clustering and differential expression, leveraging the power of scRNA-seq data at the personalized dynamic gene co-expression network level has the potential to unlock subject and/or time-specific network-level variation, which is critical for understanding phenotypic differences. Community detection from co-expression networks of multiple time points or conditions has been well-studied; however, none of the existing settings included networks from multiple subjects and multiple time points simultaneously. To address this, we develop Multi-subject Dynamic Community Detection (MuDCoD) for multi-subject community detection in personalized dynamic gene networks from scRNA-seq. MuDCoD builds on the spectral clustering framework and promotes information sharing among the networks of the subjects as well as networks at different time points. It clusters genes in the personalized dynamic gene networks and reveals gene communities that are variable or shared not only across time but also among subjects. RESULTS: Evaluation and benchmarking of MuDCoD against existing approaches reveal that MuDCoD effectively leverages apparent shared signals among networks of the subjects at individual time points, and performs robustly when there is no or little information sharing among the networks. Applications to population-scale scRNA-seq datasets of human-induced pluripotent stem cells during dopaminergic neuron differentiation and CD4+ T cell activation indicate that MuDCoD enables robust inference for identifying time-varying personalized gene modules. Our results illustrate how personalized dynamic community detection can aid in the exploration of subject-specific biological processes that vary across time. AVAILABILITY AND IMPLEMENTATION: MuDCoD is publicly available at https://github.com/bo1929/MuDCoD as a Python package. Implementation includes simulation and real-data experiments together with extensive documentation.


Subject(s)
Gene Expression Profiling , Gene Regulatory Networks , Humans , Gene Expression Profiling/methods , Software , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Cluster Analysis
13.
J Autoimmun ; 144: 103177, 2024 04.
Article in English | MEDLINE | ID: mdl-38368767

ABSTRACT

Psoriasis (PS) and atopic dermatitis (AD) are common skin inflammatory diseases characterized by hyper-responsive keratinocytes. Although, some cytokines have been suggested to be specific for each disease, other cytokines might be central to both diseases. Here, we show that Tumor necrosis factor superfamily member 14 (TNFSF14), known as LIGHT, is required for experimental PS, similar to its requirement in experimental AD. Mice devoid of LIGHT, or deletion of either of its receptors, lymphotoxin ß receptor (LTßR) and herpesvirus entry mediator (HVEM), in keratinocytes, were protected from developing imiquimod-induced psoriatic features, including epidermal thickening and hyperplasia, and expression of PS-related genes. Correspondingly, in single cell RNA-seq analysis of PS patient biopsies, LTßR transcripts were found strongly expressed with HVEM in keratinocytes, and LIGHT was upregulated in T cells. Similar transcript expression profiles were also seen in AD biopsies, and LTßR deletion in keratinocytes also protected mice from allergen-induced AD features. Moreover, in vitro, LIGHT upregulated a broad spectrum of genes in human keratinocytes that are clinical features of both PS and AD skin lesions. Our data suggest that agents blocking LIGHT activity might be useful for therapeutic intervention in PS as well as in AD.


Subject(s)
Dermatitis, Atopic , Psoriasis , Humans , Mice , Animals , Receptors, Tumor Necrosis Factor, Member 14/genetics , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Dermatitis, Atopic/genetics , Dermatitis, Atopic/metabolism , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Keratinocytes/metabolism , Cytokines/metabolism , Psoriasis/genetics , Psoriasis/metabolism , Inflammation/metabolism
14.
J Allergy Clin Immunol ; 151(4): 976-990.e5, 2023 04.
Article in English | MEDLINE | ID: mdl-36473503

ABSTRACT

BACKGROUND: Dysregulation of airway smooth muscle cells (ASM) is central to the severity of asthma. Which molecules dominantly control ASM in asthma is unclear. High levels of the cytokine LIGHT (aka TNFSF14) have been linked to asthma severity and lower baseline predicted FEV1 percentage, implying that signals through its receptors might directly control ASM dysfunction. OBJECTIVE: Our study sought to determine whether signaling via lymphotoxin beta receptor (LTßR) or herpesvirus entry mediator from LIGHT dominantly drives ASM hyperreactivity induced by allergen. METHODS: Conditional knockout mice deficient for LTßR or herpesvirus entry mediator in smooth muscle cells were used to determine their role in ASM deregulation and airway hyperresponsiveness (AHR) in vivo. Human ASM were used to study signals induced by LTßR. RESULTS: LTßR was strongly expressed in ASM from normal and asthmatic subjects compared to several other receptors implicated in smooth muscle deregulation. Correspondingly, conditional deletion of LTßR only in smooth muscle cells in smMHCCreLTßRfl/fl mice minimized changes in their numbers and mass as well as AHR induced by house dust mite allergen in a model of severe asthma. Intratracheal LIGHT administration independently induced ASM hypertrophy and AHR in vivo dependent on direct LTßR signals to ASM. LIGHT promoted contractility, hypertrophy, and hyperplasia of human ASM in vitro. Distinguishing LTßR from the receptors for IL-13, TNF, and IL-17, which have also been implicated in smooth muscle dysregulation, LIGHT promoted NF-κB-inducing kinase-dependent noncanonical nuclear factor kappa-light-chain enhancer of activated B cells in ASM in vitro, leading to sustained accumulation of F-actin, phosphorylation of myosin light chain kinase, and contractile activity. CONCLUSIONS: LTßR signals directly and dominantly drive airway smooth muscle hyperresponsiveness relevant for pathogenesis of airway remodeling in severe asthma.


Subject(s)
Asthma , Receptors, Tumor Necrosis Factor, Member 14 , Humans , Mice , Animals , Lymphotoxin beta Receptor/genetics , Asthma/pathology , Muscle, Smooth , Myocytes, Smooth Muscle/pathology , Mice, Knockout , Allergens , Lung/pathology
15.
Genome Res ; 29(9): 1415-1428, 2019 09.
Article in English | MEDLINE | ID: mdl-31434679

ABSTRACT

DNA replication occurs in a defined temporal order known as the replication timing (RT) program and is regulated during development, coordinated with 3D genome organization and transcriptional activity. However, transcription and RT are not sufficiently coordinated to predict each other, suggesting an indirect relationship. Here, we exploit genome-wide RT profiles from 15 human cell types and intermediate differentiation stages derived from human embryonic stem cells to construct different types of RT regulatory networks. First, we constructed networks based on the coordinated RT changes during cell fate commitment to create highly complex RT networks composed of thousands of interactions that form specific functional subnetwork communities. We also constructed directional regulatory networks based on the order of RT changes within cell lineages, and identified master regulators of differentiation pathways. Finally, we explored relationships between RT networks and transcriptional regulatory networks (TRNs) by combining them into more complex circuitries of composite and bipartite networks. Results identified novel trans interactions linking transcription factors that are core to the regulatory circuitry of each cell type to RT changes occurring in those cell types. These core transcription factors were found to bind cooperatively to sites in the affected replication domains, providing provocative evidence that they constitute biologically significant directional interactions. Our findings suggest a regulatory link between the establishment of cell-type-specific TRNs and RT control during lineage specification.


Subject(s)
DNA Replication Timing , Embryonic Stem Cells/cytology , Transcription Factors/metabolism , Cell Differentiation , Cell Lineage , Cells, Cultured , DNA/metabolism , Embryonic Stem Cells/chemistry , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Humans , Transcription, Genetic
16.
Genome Res ; 29(9): 1521-1532, 2019 09.
Article in English | MEDLINE | ID: mdl-31315906

ABSTRACT

Long noncoding RNAs (lncRNAs) can regulate the activity of target genes by participating in the organization of chromatin architecture. We have devised a "chromatin-RNA in situ reverse transcription sequencing" (CRIST-seq) approach to profile the lncRNA interaction network in gene regulatory elements by combining the simplicity of RNA biotin labeling with the specificity of the CRISPR/Cas9 system. Using gene-specific gRNAs, we describe a pluripotency-specific lncRNA interacting network in the promoters of Sox2 and Pou5f1, two critical stem cell factors that are required for the maintenance of pluripotency. The promoter-interacting lncRNAs were specifically activated during reprogramming into pluripotency. Knockdown of these lncRNAs caused the stem cells to exit from pluripotency. In contrast, overexpression of the pluripotency-associated lncRNA activated the promoters of core stem cell factor genes and enhanced fibroblast reprogramming into pluripotency. These CRIST-seq data suggest that the Sox2 and Pou5f1 promoters are organized within a unique lncRNA interaction network that determines the fate of pluripotency during reprogramming. This CRIST approach may be broadly used to map lncRNA interaction networks at target loci across the genome.


Subject(s)
Chromatin/genetics , Octamer Transcription Factor-3/genetics , RNA, Long Noncoding/genetics , SOXB1 Transcription Factors/genetics , Sequence Analysis, RNA/methods , Animals , CRISPR-Cas Systems , Cell Line , Cellular Reprogramming , Fibroblasts/cytology , Fibroblasts/metabolism , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid
17.
Bioinformatics ; 37(20): 3412-3420, 2021 Oct 25.
Article in English | MEDLINE | ID: mdl-34014317

ABSTRACT

MOTIVATION: Access to large-scale genomics and transcriptomics data from various tissues and cell lines allowed the discovery of wide-spread alternative splicing events and alternative promoter usage in mammalians. Between human and mouse, gene-level orthology is currently present for nearly 16k protein-coding genes spanning a diverse repertoire of over 200k total transcript isoforms. RESULTS: Here, we describe a novel method, ExTraMapper, which leverages sequence conservation between exons of a pair of organisms and identifies a fine-scale orthology mapping at the exon and then transcript level. ExTraMapper identifies more than 350k exon mappings, as well as 30k transcript mappings between human and mouse using only sequence and gene annotation information. We demonstrate that ExTraMapper identifies a larger number of exon and transcript mappings compared to previous methods. Further, it identifies exon fusions, splits and losses due to splice site mutations, and finds mappings between microexons that are previously missed. By reanalysis of RNA-seq data from 13 matched human and mouse tissues, we show that ExTraMapper improves the correlation of transcript-specific expression levels suggesting a more accurate mapping of human and mouse transcripts. We also applied the method to detect conserved exon and transcript pairs between human and rhesus macaque genomes to highlight the point that ExTraMapper is applicable to any pair of organisms that have orthologous gene pairs. AVAILABILITY AND IMPLEMENTATION: The source code and the results are available at https://github.com/ay-lab/ExTraMapper and http://ay-lab-tools.lji.org/extramapper. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.

18.
Proc Natl Acad Sci U S A ; 116(8): 3183-3192, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30723152

ABSTRACT

The positioning of chromosomes in the nucleus of a eukaryotic cell is highly organized and has a complex and dynamic relationship with gene expression. In the human malaria parasite Plasmodium falciparum, the clustering of a family of virulence genes correlates with their coordinated silencing and has a strong influence on the overall organization of the genome. To identify conserved and species-specific principles of genome organization, we performed Hi-C experiments and generated 3D genome models for five Plasmodium species and two related apicomplexan parasites. Plasmodium species mainly showed clustering of centromeres, telomeres, and virulence genes. In P. falciparum, the heterochromatic virulence gene cluster had a strong repressive effect on the surrounding nuclear space, while this was less pronounced in Plasmodium vivax and Plasmodium berghei, and absent in Plasmodium yoelii In Plasmodium knowlesi, telomeres and virulence genes were more dispersed throughout the nucleus, but its 3D genome showed a strong correlation with gene expression. The Babesia microti genome showed a classical Rabl organization with colocalization of subtelomeric virulence genes, while the Toxoplasma gondii genome was dominated by clustering of the centromeres and lacked virulence gene clustering. Collectively, our results demonstrate that spatial genome organization in most Plasmodium species is constrained by the colocalization of virulence genes. P. falciparum and P. knowlesi, the only two Plasmodium species with gene families involved in antigenic variation, are unique in the effect of these genes on chromosome folding, indicating a potential link between genome organization and gene expression in more virulent pathogens.


Subject(s)
Genome, Protozoan/genetics , Heterochromatin/genetics , Malaria, Falciparum/genetics , Plasmodium falciparum/genetics , Animals , Centromere/genetics , Gene Expression Regulation/genetics , Genomics , Humans , Malaria, Falciparum/parasitology , Plasmodium berghei/genetics , Plasmodium berghei/pathogenicity , Plasmodium falciparum/pathogenicity , Plasmodium knowlesi/genetics , Plasmodium knowlesi/pathogenicity , Plasmodium vivax/genetics , Plasmodium vivax/pathogenicity , Telomere/genetics , Toxoplasma/genetics , Toxoplasma/pathogenicity
19.
Semin Cell Dev Biol ; 90: 104-113, 2019 06.
Article in English | MEDLINE | ID: mdl-30017907

ABSTRACT

Since the advent of the chromosome conformation capture technology, our understanding of the human genome 3D organization has grown rapidly and we now know that human interphase chromosomes are folded into multiple layers of hierarchical structures and each layer can play a critical role in transcriptional regulation. Alterations in any one of these finely-tuned layers can lead to unwanted cascade of molecular events and ultimately drive the manifestation of diseases and phenotypes. Here we discuss, starting from chromosome level organization going down to single nucleotide changes, recent studies linking diseases or phenotypes to changes in the 3D genome architecture.


Subject(s)
Chromosomes/genetics , Disease/genetics , Nucleotides/genetics , Chromosomes/chemistry , Humans , Nucleotides/chemistry , Phenotype
20.
J Immunol ; 203(2): 329-337, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31175163

ABSTRACT

Despite recent advances in asthma management with anti-IL-5 therapies, many patients have eosinophilic asthma that remains poorly controlled. IL-3 shares a common ß subunit receptor with both IL-5 and GM-CSF but, through α-subunit-specific properties, uniquely influences eosinophil biology and may serve as a potential therapeutic target. We aimed to globally characterize the transcriptomic profiles of GM-CSF, IL-3, and IL-5 stimulation on human circulating eosinophils and identify differences in gene expression using advanced statistical modeling. Human eosinophils were isolated from the peripheral blood of healthy volunteers and stimulated with either GM-CSF, IL-3, or IL-5 for 48 h. RNA was then extracted and bulk sequencing performed. DESeq analysis identified differentially expressed genes and weighted gene coexpression network analysis independently defined modules of genes that are highly coexpressed. GM-CSF, IL-3, and IL-5 commonly upregulated 252 genes and downregulated 553 genes, producing a proinflammatory and survival phenotype that was predominantly mediated through TWEAK signaling. IL-3 stimulation yielded the most numbers of differentially expressed genes that were also highly coexpressed (n = 119). These genes were enriched in pathways involving JAK/STAT signaling. GM-CSF and IL-5 stimulation demonstrated redundancy in eosinophil gene expression. In conclusion, IL-3 produces a distinct eosinophil gene expression program among the ß-chain receptor cytokines. IL-3-upregulated genes may provide a foundation for research into therapeutics for patients with eosinophilic asthma who do not respond to anti-IL-5 therapies.


Subject(s)
Cytokines/immunology , Eosinophils/immunology , Gene Expression/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Interleukin-3/immunology , Interleukin-5/immunology , Asthma/immunology , Down-Regulation/immunology , Humans , Signal Transduction/immunology , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL