Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 252
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 181(3): 728-744.e21, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32302591

ABSTRACT

Adoptive transfer of genetically modified immune cells holds great promise for cancer immunotherapy. CRISPR knockin targeting can improve cell therapies, but more high-throughput methods are needed to test which knockin gene constructs most potently enhance primary cell functions in vivo. We developed a widely adaptable technology to barcode and track targeted integrations of large non-viral DNA templates and applied it to perform pooled knockin screens in primary human T cells. Pooled knockin of dozens of unique barcoded templates into the T cell receptor (TCR)-locus revealed gene constructs that enhanced fitness in vitro and in vivo. We further developed pooled knockin sequencing (PoKI-seq), combining single-cell transcriptome analysis and pooled knockin screening to measure cell abundance and cell state ex vivo and in vivo. This platform nominated a novel transforming growth factor ß (TGF-ß) R2-41BB chimeric receptor that improved solid tumor clearance. Pooled knockin screening enables parallelized re-writing of endogenous genetic sequences to accelerate discovery of knockin programs for cell therapies.


Subject(s)
Gene Knock-In Techniques/methods , Genetic Engineering/methods , Immunotherapy/methods , Animals , Blood Cells , CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Humans , Mice , Mice, Inbred NOD , Mice, SCID , RNA, Guide, Kinetoplastida/genetics , Single-Cell Analysis/methods , T-Lymphocytes , Transcriptome/genetics
2.
Cell ; 183(6): 1479-1495.e20, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33171100

ABSTRACT

We present an integrated analysis of the clinical measurements, immune cells, and plasma multi-omics of 139 COVID-19 patients representing all levels of disease severity, from serial blood draws collected during the first week of infection following diagnosis. We identify a major shift between mild and moderate disease, at which point elevated inflammatory signaling is accompanied by the loss of specific classes of metabolites and metabolic processes. Within this stressed plasma environment at moderate disease, multiple unusual immune cell phenotypes emerge and amplify with increasing disease severity. We condensed over 120,000 immune features into a single axis to capture how different immune cell classes coordinate in response to SARS-CoV-2. This immune-response axis independently aligns with the major plasma composition changes, with clinical metrics of blood clotting, and with the sharp transition between mild and moderate disease. This study suggests that moderate disease may provide the most effective setting for therapeutic intervention.


Subject(s)
COVID-19 , Genomics , RNA-Seq , SARS-CoV-2 , Single-Cell Analysis , Adolescent , Adult , Aged , Aged, 80 and over , COVID-19/blood , COVID-19/immunology , Female , Humans , Male , Middle Aged , SARS-CoV-2/immunology , SARS-CoV-2/metabolism , Severity of Illness Index
3.
Nat Immunol ; 21(11): 1456-1466, 2020 11.
Article in English | MEDLINE | ID: mdl-32989329

ABSTRACT

Human regulatory T (Treg) cells are essential for immune homeostasis. The transcription factor FOXP3 maintains Treg cell identity, yet the complete set of key transcription factors that control Treg cell gene expression remains unknown. Here, we used pooled and arrayed Cas9 ribonucleoprotein screens to identify transcription factors that regulate critical proteins in primary human Treg cells under basal and proinflammatory conditions. We then generated 54,424 single-cell transcriptomes from Treg cells subjected to genetic perturbations and cytokine stimulation, which revealed distinct gene networks individually regulated by FOXP3 and PRDM1, in addition to a network coregulated by FOXO1 and IRF4. We also discovered that HIVEP2, to our knowledge not previously implicated in Treg cell function, coregulates another gene network with SATB1 and is important for Treg cell-mediated immunosuppression. By integrating CRISPR screens and single-cell RNA-sequencing profiling, we have uncovered transcriptional regulators and downstream gene networks in human Treg cells that could be targeted for immunotherapies.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transcriptome , Biomarkers , CRISPR-Cas Systems , Disease Susceptibility , Gene Knockout Techniques , Gene Targeting , Graft vs Host Disease/etiology , High-Throughput Nucleotide Sequencing , Humans
4.
Immunity ; 50(2): 362-377.e6, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30709738

ABSTRACT

Regulatory T (Treg) cells maintain immune tolerance through the master transcription factor forkhead box P3 (FOXP3), which is crucial for Treg cell function and homeostasis. We identified an IPEX (immune dysregulation polyendocrinopathy enteropathy X-linked) syndrome patient with a FOXP3 mutation in the domain swap interface of the protein. Recapitulation of this Foxp3 variant in mice led to the development of an autoimmune syndrome consistent with an unrestrained T helper type 2 (Th2) immune response. Genomic analysis of Treg cells by RNA-sequencing, Foxp3 chromatin immunoprecipitation followed by high-throughput DNA sequencing (ChIP-sequencing), and H3K27ac-HiChIP revealed a specific de-repression of the Th2 transcriptional program leading to the generation of Th2-like Treg cells that were unable to suppress extrinsic Th2 cells. Th2-like Treg cells showed increased intra-chromosomal interactions in the Th2 locus, leading to type 2 cytokine production. These findings identify a direct role for Foxp3 in suppressing Th2-like Treg cells and implicate additional pathways that could be targeted to restrain Th2 trans-differentiated Treg cells.


Subject(s)
Forkhead Transcription Factors/immunology , Mutation , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Child , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/immunology , Genetic Diseases, X-Linked/metabolism , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Polyendocrinopathies, Autoimmune/genetics , Polyendocrinopathies, Autoimmune/immunology , Polyendocrinopathies, Autoimmune/metabolism , T-Lymphocytes, Regulatory/metabolism , Th2 Cells/metabolism
5.
Nat Immunol ; 16(2): 188-96, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25559257

ABSTRACT

Foxp3(+) regulatory T cells (Treg cells) are required for immunological homeostasis. One notable distinction between conventional T cells (Tconv cells) and Treg cells is differences in the activity of phosphatidylinositol-3-OH kinase (PI(3)K); only Tconv cells downregulate PTEN, the main negative regulator of PI(3)K, upon activation. Here we found that control of PI(3)K in Treg cells was essential for lineage homeostasis and stability. Mice lacking Pten in Treg cells developed an autoimmune-lymphoproliferative disease characterized by excessive T helper type 1 (TH1) responses and B cell activation. Diminished control of PI(3)K activity in Treg cells led to reduced expression of the interleukin-2 (IL-2) receptor α subunit CD25, accumulation of Foxp3(+)CD25(-) cells and, ultimately, loss of expression of the transcription factor Foxp3 in these cells. Collectively, our data demonstrate that control of PI(3)K signaling by PTEN in Treg cells is critical for maintaining their homeostasis, function and stability.


Subject(s)
Homeostasis/immunology , Phosphatidylinositol 3-Kinases/metabolism , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Lineage , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Deletion , Mice , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction
6.
Proc Natl Acad Sci U S A ; 121(20): e2320268121, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38709934

ABSTRACT

Insulin is a central autoantigen in the pathogenesis of T1D, and thymic epithelial cell expression of insulin under the control of the Autoimmune Regulator (Aire) is thought to be a key component of maintaining tolerance to insulin. In spite of this general working model, direct detection of this thymic selection on insulin-specific T cells has been somewhat elusive. Here, we used a combination of highly sensitive T cell receptor transgenic models for detecting thymic selection and sorting and sequencing of Insulin-specific CD4+ T cells from Aire-deficient mice as a strategy to further define their selection. This analysis revealed a number of unique t cell receptor (TCR) clones in Aire-deficient hosts with high affinity for insulin/major histocompatibility complex (MHC) ligands. We then modeled the thymic selection of one of these clones in Aire-deficient versus wild-type hosts and found that this model clone could escape thymic negative selection in the absence of thymic Aire. Together, these results suggest that thymic expression of insulin plays a key role in trimming and removing high-affinity insulin-specific T cells from the repertoire to help promote tolerance.


Subject(s)
AIRE Protein , Insulin , Receptors, Antigen, T-Cell , Thymus Gland , Animals , Mice , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Clone Cells , Immune Tolerance , Insulin/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Thymus Gland/immunology , Thymus Gland/metabolism , Thymus Gland/cytology , Transcription Factors/metabolism , Transcription Factors/genetics
7.
Nature ; 582(7812): 416-420, 2020 06.
Article in English | MEDLINE | ID: mdl-32499641

ABSTRACT

Regulatory T (Treg) cells are required to control immune responses and maintain homeostasis, but are a significant barrier to antitumour immunity1. Conversely, Treg instability, characterized by loss of the master transcription factor Foxp3 and acquisition of proinflammatory properties2, can promote autoimmunity and/or facilitate more effective tumour immunity3,4. A comprehensive understanding of the pathways that regulate Foxp3 could lead to more effective Treg therapies for autoimmune disease and cancer. The availability of new functional genetic tools has enabled the possibility of systematic dissection of the gene regulatory programs that modulate Foxp3 expression. Here we developed a CRISPR-based pooled screening platform for phenotypes in primary mouse Treg cells and applied this technology to perform a targeted loss-of-function screen of around 500 nuclear factors to identify gene regulatory programs that promote or disrupt Foxp3 expression. We identified several modulators of Foxp3 expression, including ubiquitin-specific peptidase 22 (Usp22) and ring finger protein 20 (Rnf20). Usp22, a member of the deubiquitination module of the SAGA chromatin-modifying complex, was revealed to be a positive regulator that stabilized Foxp3 expression; whereas the screen suggested that Rnf20, an E3 ubiquitin ligase, can serve as a negative regulator of Foxp3. Treg-specific ablation of Usp22 in mice reduced Foxp3 protein levels and caused defects in their suppressive function that led to spontaneous autoimmunity but protected against tumour growth in multiple cancer models. Foxp3 destabilization in Usp22-deficient Treg cells could be rescued by ablation of Rnf20, revealing a reciprocal ubiquitin switch in Treg cells. These results reveal previously unknown modulators of Foxp3 and demonstrate a screening method that can be broadly applied to discover new targets for Treg immunotherapies for cancer and autoimmune disease.


Subject(s)
CRISPR-Cas Systems , Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Autoimmunity/immunology , Cells, Cultured , Forkhead Transcription Factors/biosynthesis , Gene Editing , Gene Expression Regulation , Humans , Immunotherapy , Male , Mice , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/prevention & control , Protein Stability , Reproducibility of Results , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Ubiquitin Thiolesterase/deficiency , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/metabolism
8.
Nat Immunol ; 14(8): 840-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23812098

ABSTRACT

Follicular helper T cells (TFH cells) are the prototypic helper T cell subset specialized to enable B cells to form germinal centers (GCs) and produce high-affinity antibodies. We found that expression of microRNAs (miRNAs) by T cells was essential for TFH cell differentiation. More specifically, we show that after immunization of mice with protein, the miRNA cluster miR-17∼92 was critical for robust differentiation and function of TFH cells in a cell-intrinsic manner that occurred regardless of changes in proliferation. In a viral infection model, miR-17∼92 restrained the expression of genes 'inappropriate' to the TFH cell subset, including the direct miR-17∼92 target Rora. Removal of one Rora allele partially 'rescued' the inappropriate gene signature in miR-17∼92-deficient TFH cells. Our results identify the miR-17∼92 cluster as a critical regulator of T cell-dependent antibody responses, TFH cell differentiation and the fidelity of the TFH cell gene-expression program.


Subject(s)
Cell Differentiation/immunology , Gene Expression Regulation/immunology , MicroRNAs/immunology , Nuclear Receptor Subfamily 1, Group F, Member 1/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adaptive Immunity/immunology , Animals , Arenaviridae Infections/immunology , Arenaviridae Infections/virology , Flow Cytometry , Immunohistochemistry , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Statistics, Nonparametric , T-Lymphocytes, Helper-Inducer/cytology
9.
Immunity ; 44(5): 973-88, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27192564

ABSTRACT

Ligation of the CD28 receptor on T cells provides a critical second signal alongside T cell receptor (TCR) ligation for naive T cell activation. Here, we discuss the expression, structure, and biochemistry of CD28 and its ligands. CD28 signals play a key role in many T cell processes, including cytoskeletal remodeling, production of cytokines, survival, and differentiation. CD28 ligation leads to unique epigenetic, transcriptional, and post-translational changes in T cells that cannot be recapitulated by TCR ligation alone. We discuss the function of CD28 and its ligands in both effector and regulatory T cells. CD28 is critical for regulatory T cell survival and the maintenance of immune homeostasis. We outline the roles that CD28 and its family members play in human disease and we review the clinical efficacy of drugs that block CD28 ligands. Despite the centrality of CD28 and its family members and ligands to immune function, many aspects of CD28 biology remain unclear. Translation of a basic understanding of CD28 function into immunomodulatory therapeutics has been uneven, with both successes and failures. Such real-world results might stem from multiple factors, including complex receptor-ligand interactions among CD28 family members, differences between the mouse and human CD28 families, and cell-type specific roles of CD28 family members.


Subject(s)
Autoimmune Diseases/immunology , CD28 Antigens/metabolism , CTLA-4 Antigen/antagonists & inhibitors , Immunotherapy/methods , T-Lymphocytes/immunology , Abatacept/therapeutic use , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/therapy , CD28 Antigens/genetics , CD28 Antigens/immunology , Homeostasis , Humans , Immunotherapy/trends , Lymphocyte Activation , Mice , Molecular Targeted Therapy , Receptor Cross-Talk , Receptors, Antigen, T-Cell/metabolism , Signal Transduction
10.
Blood ; 139(19): 2983-2997, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35226736

ABSTRACT

Despite advances in the field, chronic graft-versus-host-disease (cGVHD) remains a leading cause of morbidity and mortality following allogenic hematopoietic stem cell transplant. Because treatment options remain limited, we tested efficacy of anticancer, chromatin-modifying enzyme inhibitors in a clinically relevant murine model of cGVHD with bronchiolitis obliterans (BO). We observed that the novel enhancer of zeste homolog 2 (EZH2) inhibitor JQ5 and the BET-bromodomain inhibitor JQ1 each improved pulmonary function; impaired the germinal center (GC) reaction, a prerequisite in cGVHD/BO pathogenesis; and JQ5 reduced EZH2-mediated H3K27me3 in donor T cells. Using conditional EZH2 knockout donor cells, we demonstrated that EZH2 is obligatory for the initiation of cGVHD/BO. In a sclerodermatous cGVHD model, JQ5 reduced the severity of cutaneous lesions. To determine how the 2 drugs could lead to the same physiological improvements while targeting unique epigenetic processes, we analyzed the transcriptomes of splenic GCB cells (GCBs) from transplanted mice treated with either drug. Multiple inflammatory and signaling pathways enriched in cGVHD/BO GCBs were reduced by each drug. GCBs from JQ5- but not JQ1-treated mice were enriched for proproliferative pathways also seen in GCBs from bone marrow-only transplanted mice, likely reflecting their underlying biology in the unperturbed state. In conjunction with in vivo data, these insights led us to conclude that epigenetic targeting of the GC is a viable clinical approach for the treatment of cGVHD, and that the EZH2 inhibitor JQ5 and the BET-bromodomain inhibitor JQ1 demonstrated clinical potential for EZH2i and BETi in patients with cGVHD/BO.


Subject(s)
Bronchiolitis Obliterans , Enhancer of Zeste Homolog 2 Protein , Germinal Center , Graft vs Host Disease , Proteins , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Bronchiolitis Obliterans/genetics , Bronchiolitis Obliterans/metabolism , Bronchiolitis Obliterans/pathology , Chronic Disease , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Enzyme Inhibitors/pharmacology , Germinal Center/drug effects , Germinal Center/pathology , Graft vs Host Disease/drug therapy , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Humans , Mice , Proteins/metabolism , Transcriptome
11.
Trends Immunol ; 42(4): 293-311, 2021 04.
Article in English | MEDLINE | ID: mdl-33714688

ABSTRACT

Cancer immunotherapies can successfully activate immune responses towards certain tumors. However, this can also result in the development of treatment-induced immune-related adverse events (irAEs) in multiple tissues. Growing evidence suggests that cytokine production in response to these therapeutics potentiates the development of irAEs and may have predictive value as biomarkers for irAE occurrence. In addition, therapeutic agents that inhibit cytokine activity can limit the severity of irAEs, and their use is being tested in the clinical setting. This review provides an in-depth analysis of strategies to uncouple the cytokine response, that precipitates irAEs following cancer immunotherapies, from the benefit gained in promoting antitumor immunity.


Subject(s)
Cytokines , Immune Checkpoint Inhibitors , Neoplasms , Humans , Immune Checkpoint Inhibitors/adverse effects , Immunotherapy/adverse effects , Neoplasms/drug therapy
12.
Immunity ; 42(5): 779-81, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25992850

ABSTRACT

In this issue of Immunity, Spangler et al. and Mitra et al. demonstrate how structural changes in the IL-2 molecule alter interactions with the IL-2 receptor, leading to differential cellular targeting and biochemical responses and selective immune consequences.


Subject(s)
Antibodies/immunology , Interleukin-2/metabolism , Models, Molecular , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Animals , Humans
13.
Immunity ; 43(1): 161-74, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26092469

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) and regulatory T (Treg) cells are systemically induced by helminth infection but also sustain metabolic homeostasis in adipose tissue and contribute to tissue repair during injury. Here we show that interleukin-33 (IL-33) mediates activation of ILC2s and Treg cells in resting adipose tissue, but also after helminth infection or treatment with IL-2. Unexpectedly, ILC2-intrinsic IL-33 activation was required for Treg cell accumulation in vivo and was independent of ILC2 type 2 cytokines but partially dependent on direct co-stimulatory interactions via ICOSL-ICOS. IFN-γ inhibited ILC2 activation and Treg cell accumulation by IL-33 in infected tissue, as well as adipose tissue, where repression increased with aging and high-fat diet-induced obesity. IL-33 and ILC2s are central mediators of type 2 immune responses that promote tissue and metabolic homeostasis, and IFN-γ suppresses this pathway, likely to promote inflammatory responses and divert metabolic resources necessary to protect the host.


Subject(s)
Interferon-gamma/immunology , Interleukins/immunology , Lymphocyte Activation/immunology , Strongylida Infections/immunology , T-Lymphocytes, Regulatory/immunology , Aging/immunology , Animals , Diet, High-Fat , Enzyme Activation/immunology , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Inducible T-Cell Co-Stimulator Protein/immunology , Interferon-gamma/pharmacology , Interleukin-2/pharmacology , Interleukin-33 , Intra-Abdominal Fat/cytology , Intra-Abdominal Fat/immunology , Lectins, C-Type , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/microbiology , Lung/cytology , Lung/immunology , Lung/pathology , Mice , Mice, Transgenic , Nippostrongylus/immunology , Obesity/immunology , Receptors, Immunologic/biosynthesis , Strongylida Infections/parasitology
14.
Immunity ; 42(2): 227-238, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25680271

ABSTRACT

Regulatory T cells (Treg cells) are required for immune homeostasis. Chromatin remodeling is essential for establishing diverse cellular identities, but how the epigenetic program in Treg cells is maintained throughout the dynamic activation process remains unclear. Here we have shown that CD28 co-stimulation, an extracellular cue intrinsically required for Treg cell maintenance, induced the chromatin-modifying enzyme, Ezh2. Treg-specific ablation of Ezh2 resulted in spontaneous autoimmunity with reduced Foxp3(+) cells in non-lymphoid tissues and impaired resolution of experimental autoimmune encephalomyelitis. Utilizing a model designed to selectively deplete wild-type Treg cells in adult mice co-populated with Ezh2-deficient Treg cells, Ezh2-deficient cells were destabilized and failed to prevent autoimmunity. After activation, the transcriptome of Ezh2-deficient Treg cells was disrupted, with altered expression of Treg cell lineage genes in a pattern similar to Foxp3-deficient Treg cells. These studies reveal a critical role for Ezh2 in the maintenance of Treg cell identity during cellular activation.


Subject(s)
CD28 Antigens/immunology , Lymphocyte Activation/immunology , Polycomb Repressive Complex 2/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity/genetics , Autoimmunity/immunology , CD8-Positive T-Lymphocytes/immunology , Chromatin Assembly and Disassembly , Encephalomyelitis, Autoimmune, Experimental/immunology , Enhancer of Zeste Homolog 2 Protein , Female , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Heparin-binding EGF-like Growth Factor/genetics , Immune Tolerance/genetics , Immune Tolerance/immunology , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Transgenic , Polycomb Repressive Complex 2/genetics , Promoter Regions, Genetic/genetics , T-Lymphocytes, Regulatory/cytology
15.
Nature ; 559(7715): E13, 2018 07.
Article in English | MEDLINE | ID: mdl-29899441

ABSTRACT

In this Letter, analysis of steady-state regulatory T (Treg) cell percentages from Il2ra enhancer deletion (EDEL) and wild-type (WT) mice revealed no differences between them (Extended Data Fig. 9d). This analysis included two mice whose genotypes were incorrectly assigned. Even after correction of the genotypes, no significant differences in Treg cell percentages were seen when data across experimental cohorts were averaged (as was done in Extended Data Fig. 9d). However, if we normalize the corrected data to account for variation among experimental cohorts, a subtle decrease in EDEL Treg cell percentages is revealed and, using the corrected and normalized data, we have redrawn Extended Data Fig. 9d in Supplementary Fig. 1. The Supplementary Information to this Amendment contains the corrected and reanalysed Extended Data Fig. 9d. The sentence "This enhancer deletion (EDEL) strain also had no obvious T cell phenotypes at steady state (Extended Data Fig. 9)." should read: "This enhancer deletion (EDEL) strain had a small decrease in the percentage of Treg cells (Extended Data Fig. 9).". This error does not affect any of the main figures in the Letter or the data from mice with the human autoimmune-associated single nucleotide polymorphism (SNP) knocked in or with a 12-base-pair deletion at the site (12DEL). In addition, we stated in the Methods that we observed consistent immunophenotypes of EDEL mice across three founders, but in fact, we observed consistent phenotypes in mice from two founders. This does not change any of our conclusions and the original Letter has not been corrected.

16.
Nat Immunol ; 12(9): 898-907, 2011 Aug 14.
Article in English | MEDLINE | ID: mdl-21841785

ABSTRACT

Regulatory T cells (T(reg) cells) are essential for self-tolerance and immune homeostasis. Lack of effector T cell (T(eff) cell) function and gain of suppressive activity by T(reg) cells are dependent on the transcriptional program induced by Foxp3. Here we report that repression of SATB1, a genome organizer that regulates chromatin structure and gene expression, was crucial for the phenotype and function of T(reg) cells. Foxp3, acting as a transcriptional repressor, directly suppressed the SATB1 locus and indirectly suppressed it through the induction of microRNAs that bound the SATB1 3' untranslated region. Release of SATB1 from the control of Foxp3 in T(reg) cells caused loss of suppressive function, establishment of transcriptional T(eff) cell programs and induction of T(eff) cell cytokines. Our data support the proposal that inhibition of SATB1-mediated modulation of global chromatin remodeling is pivotal for maintaining T(reg) cell functionality.


Subject(s)
Chromatin Assembly and Disassembly/immunology , Forkhead Transcription Factors/immunology , Gene Expression Regulation , Matrix Attachment Region Binding Proteins/immunology , Self Tolerance , T-Lymphocytes, Regulatory/immunology , 3' Untranslated Regions/genetics , 3' Untranslated Regions/immunology , Animals , Cell Differentiation/drug effects , Chromatin Assembly and Disassembly/drug effects , Flow Cytometry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Genome, Human , Genome-Wide Association Study , Humans , Lentivirus , Lymphocyte Activation/drug effects , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/immunology , MicroRNAs/metabolism , MicroRNAs/pharmacology , RNA Interference , RNA, Small Interfering/immunology , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Self Tolerance/drug effects , Self Tolerance/genetics , Self Tolerance/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Transduction, Genetic
17.
Nature ; 549(7670): 111-115, 2017 09 07.
Article in English | MEDLINE | ID: mdl-28854172

ABSTRACT

The majority of genetic variants associated with common human diseases map to enhancers, non-coding elements that shape cell-type-specific transcriptional programs and responses to extracellular cues. Systematic mapping of functional enhancers and their biological contexts is required to understand the mechanisms by which variation in non-coding genetic sequences contributes to disease. Functional enhancers can be mapped by genomic sequence disruption, but this approach is limited to the subset of enhancers that are necessary in the particular cellular context being studied. We hypothesized that recruitment of a strong transcriptional activator to an enhancer would be sufficient to drive target gene expression, even if that enhancer was not currently active in the assayed cells. Here we describe a discovery platform that can identify stimulus-responsive enhancers for a target gene independent of stimulus exposure. We used tiled CRISPR activation (CRISPRa) to synthetically recruit a transcriptional activator to sites across large genomic regions (more than 100 kilobases) surrounding two key autoimmunity risk loci, CD69 and IL2RA. We identified several CRISPRa-responsive elements with chromatin features of stimulus-responsive enhancers, including an IL2RA enhancer that harbours an autoimmunity risk variant. Using engineered mouse models, we found that sequence perturbation of the disease-associated Il2ra enhancer did not entirely block Il2ra expression, but rather delayed the timing of gene activation in response to specific extracellular signals. Enhancer deletion skewed polarization of naive T cells towards a pro-inflammatory T helper (TH17) cell state and away from a regulatory T cell state. This integrated approach identifies functional enhancers and reveals how non-coding variation associated with human immune dysfunction alters context-specific gene programs.


Subject(s)
Autoimmunity/genetics , CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Enhancer Elements, Genetic/genetics , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/biosynthesis , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/immunology , Cell Differentiation , Cell Line , Chromatin/genetics , Female , Gene Expression Regulation/genetics , Humans , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Lectins, C-Type/biosynthesis , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Mice , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Th17 Cells/cytology , Th17 Cells/immunology
18.
N Engl J Med ; 381(7): 603-613, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31180194

ABSTRACT

BACKGROUND: Type 1 diabetes is a chronic autoimmune disease that leads to destruction of insulin-producing beta cells and dependence on exogenous insulin for survival. Some interventions have delayed the loss of insulin production in patients with type 1 diabetes, but interventions that might affect clinical progression before diagnosis are needed. METHODS: We conducted a phase 2, randomized, placebo-controlled, double-blind trial of teplizumab (an Fc receptor-nonbinding anti-CD3 monoclonal antibody) involving relatives of patients with type 1 diabetes who did not have diabetes but were at high risk for development of clinical disease. Patients were randomly assigned to a single 14-day course of teplizumab or placebo, and follow-up for progression to clinical type 1 diabetes was performed with the use of oral glucose-tolerance tests at 6-month intervals. RESULTS: A total of 76 participants (55 [72%] of whom were ≤18 years of age) underwent randomization - 44 to the teplizumab group and 32 to the placebo group. The median time to the diagnosis of type 1 diabetes was 48.4 months in the teplizumab group and 24.4 months in the placebo group; the disease was diagnosed in 19 (43%) of the participants who received teplizumab and in 23 (72%) of those who received placebo. The hazard ratio for the diagnosis of type 1 diabetes (teplizumab vs. placebo) was 0.41 (95% confidence interval, 0.22 to 0.78; P = 0.006 by adjusted Cox proportional-hazards model). The annualized rates of diagnosis of diabetes were 14.9% per year in the teplizumab group and 35.9% per year in the placebo group. There were expected adverse events of rash and transient lymphopenia. KLRG1+TIGIT+CD8+ T cells were more common in the teplizumab group than in the placebo group. Among the participants who were HLA-DR3-negative, HLA-DR4-positive, or anti-zinc transporter 8 antibody-negative, fewer participants in the teplizumab group than in the placebo group had diabetes diagnosed. CONCLUSIONS: Teplizumab delayed progression to clinical type 1 diabetes in high-risk participants. (Funded by the National Institutes of Health and others; ClinicalTrials.gov number, NCT01030861.).


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , CD3 Complex/antagonists & inhibitors , Diabetes Mellitus, Type 1/prevention & control , Adolescent , Adult , Antibodies, Monoclonal, Humanized/adverse effects , Child , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Disease Progression , Double-Blind Method , Exanthema/chemically induced , Female , Glucose Tolerance Test , HLA-DR3 Antigen , HLA-DR4 Antigen , Humans , Lymphocyte Count , Lymphopenia/chemically induced , Male , Middle Aged , Proportional Hazards Models , T-Lymphocytes/immunology , Young Adult
19.
Immunity ; 39(5): 949-62, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24238343

ABSTRACT

Stable Foxp3 expression is crucial for regulatory T (Treg) cell function. We observed that antigen-driven activation and inflammation in the CNS promoted Foxp3 instability selectively in the autoreactive Treg cells that expressed high amounts of Foxp3 before experimental autoimmune encephalitis induction. Treg cells with a demethylated Treg-cell-specific demethylated region in the Foxp3 locus downregulated Foxp3 transcription in the inflamed CNS during the induction phase of the response. Stable Foxp3 expression returned at the population level with the resolution of inflammation or was rescued by IL-2-anti-IL-2 complex treatment during the antigen priming phase. Thus, a subset of fully committed self-antigen-specific Treg cells lost Foxp3 expression during an inflammatory autoimmune response and might be involved in inadequate control of autoimmunity. These results have important implications for Treg cell therapies and give insights into the dynamics of the Treg cell network during autoreactive CD4(+) T cell effector responses in vivo.


Subject(s)
Autoantigens/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Forkhead Transcription Factors/physiology , Gene Expression Regulation/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Lineage , Central Nervous System/immunology , DNA Methylation , Down-Regulation/immunology , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Genes, Reporter , Lymph Nodes/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/immunology , Peptide Fragments/immunology , Receptors, Interleukin-2/physiology , Recombinant Fusion Proteins/immunology , Regulatory Sequences, Nucleic Acid , Specific Pathogen-Free Organisms
20.
J Immunol ; 204(4): 747-751, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31924653

ABSTRACT

IL-6 binds to the IL-6R α-chain (IL-6Rα) and signals via the signal transducer gp130. Recently, IL-6 was found to also bind to the cell surface glycoprotein CD5, which would then engage gp130 in the absence of IL-6Rα. However, the biological relevance of this alternative pathway is under debate. In this study, we developed a mouse model, in which murine IL-6 is overexpressed in a CD11c-Cre-dependent manner. Transgenic mice developed a lethal immune dysregulation syndrome with increased numbers of Ly-6G+ neutrophils and Ly-6Chi monocytes/macrophages. IL-6 overexpression promoted activation of CD4+ T cells while suppressing CD5+ B-1a cell development. However, additional ablation of IL-6Rα protected IL-6-overexpressing mice from IL-6-triggered inflammation and fully phenocopied IL-6Rα-deficient mice without IL-6 overexpression. Mechanistically, IL-6Rα deficiency completely prevented downstream activation of STAT3 in response to IL-6. Altogether, our data clarify that IL-6Rα is the only biologically relevant receptor for IL-6 in mice.


Subject(s)
Interleukin-6/immunology , Receptors, Interleukin-6/immunology , Signal Transduction/immunology , Animals , Mice , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL