Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.132
Filter
Add more filters

Publication year range
1.
Cell ; 185(5): 794-814.e30, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35182466

ABSTRACT

Congenital heart disease (CHD) is present in 1% of live births, yet identification of causal mutations remains challenging. We hypothesized that genetic determinants for CHDs may lie in the protein interactomes of transcription factors whose mutations cause CHDs. Defining the interactomes of two transcription factors haplo-insufficient in CHD, GATA4 and TBX5, within human cardiac progenitors, and integrating the results with nearly 9,000 exomes from proband-parent trios revealed an enrichment of de novo missense variants associated with CHD within the interactomes. Scoring variants of interactome members based on residue, gene, and proband features identified likely CHD-causing genes, including the epigenetic reader GLYR1. GLYR1 and GATA4 widely co-occupied and co-activated cardiac developmental genes, and the identified GLYR1 missense variant disrupted interaction with GATA4, impairing in vitro and in vivo function in mice. This integrative proteomic and genetic approach provides a framework for prioritizing and interrogating genetic variants in heart disease.


Subject(s)
GATA4 Transcription Factor/metabolism , Heart Defects, Congenital , Nuclear Proteins/metabolism , Oxidoreductases/metabolism , Transcription Factors , Animals , Heart Defects, Congenital/genetics , Mice , Mutation , Proteomics , T-Box Domain Proteins/genetics , Transcription Factors/genetics
2.
Nat Immunol ; 25(7): 1172-1182, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38871999

ABSTRACT

Natural killer (NK) cells traffic through the blood and mount cytolytic and interferon-γ (IFNγ)-focused responses to intracellular pathogens and tumors. Type 1 innate lymphoid cells (ILC1s) also produce type 1 cytokines but reside in tissues and are not cytotoxic. Whether these differences reflect discrete lineages or distinct states of a common cell type is not understood. Using single-cell RNA sequencing and flow cytometry, we focused on populations of TCF7+ cells that contained precursors for NK cells and ILC1s and identified a subset of bone marrow lineage-negative NK receptor-negative cells that expressed the transcription factor Eomes, termed EomeshiNKneg cells. Transfer of EomeshiNKneg cells into Rag2-/-Il2rg-/- recipients generated functional NK cells capable of preventing metastatic disease. By contrast, transfer of PLZF+ ILC precursors generated a mixture of ILC1s, ILC2s and ILC3s that lacked cytotoxic potential. These findings identified EomeshiNKneg cells as the bone marrow precursor to classical NK cells and demonstrated that the NK and ILC1 lineages diverged early during development.


Subject(s)
Killer Cells, Natural , T-Box Domain Proteins , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Animals , T-Box Domain Proteins/metabolism , T-Box Domain Proteins/genetics , Mice , Mice, Knockout , Cell Lineage/immunology , Mice, Inbred C57BL , Immunity, Innate , Cell Differentiation/immunology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Single-Cell Analysis
3.
Nat Immunol ; 25(8): 1460-1473, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38956380

ABSTRACT

Group 1 innate lymphoid cells (ILC1s) are cytotoxic and interferon gamma-producing lymphocytes lacking antigen-specific receptors, which include ILC1s and natural killer (NK) cells. In mice, ILC1s differ from NK cells, as they develop independently of the NK-specifying transcription factor EOMES, while requiring the repressor ZFP683 (ZNF683 in humans) for tissue residency. Here we identify highly variable ILC1 subtypes across tissues through investigation of human ILC1 diversity by single-cell RNA sequencing and flow cytometry. The intestinal epithelium contained abundant mature EOMES- ILC1s expressing PRDM1 rather than ZNF683, alongside a few immature TCF7+PRDM1- ILC1s. Other tissues harbored NK cells expressing ZNF683 and EOMES transcripts; however, EOMES protein content was variable. These ZNF683+ NK cells are tissue-imprinted NK cells phenotypically resembling ILC1s. The tissue ILC1-NK spectrum also encompassed conventional NK cells and NK cells distinguished by PTGDS expression. These findings establish a foundation for evaluating phenotypic and functional changes within the NK-ILC1 spectrum in diseases.


Subject(s)
Immunity, Innate , Killer Cells, Natural , Lymphocytes , Positive Regulatory Domain I-Binding Factor 1 , T-Box Domain Proteins , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , T-Box Domain Proteins/metabolism , T-Box Domain Proteins/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Positive Regulatory Domain I-Binding Factor 1/genetics , Lymphocytes/immunology , Lymphocytes/metabolism , Single-Cell Analysis , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Animals , Mice , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Repressor Proteins/metabolism , Repressor Proteins/genetics
4.
Cell ; 183(7): 1826-1847.e31, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33296702

ABSTRACT

Inborn errors of human interferon gamma (IFN-γ) immunity underlie mycobacterial disease. We report a patient with mycobacterial disease due to inherited deficiency of the transcription factor T-bet. The patient has extremely low counts of circulating Mycobacterium-reactive natural killer (NK), invariant NKT (iNKT), mucosal-associated invariant T (MAIT), and Vδ2+ γδ T lymphocytes, and of Mycobacterium-non reactive classic TH1 lymphocytes, with the residual populations of these cells also producing abnormally small amounts of IFN-γ. Other lymphocyte subsets develop normally but produce low levels of IFN-γ, with the exception of CD8+ αß T and non-classic CD4+ αß TH1∗ lymphocytes, which produce IFN-γ normally in response to mycobacterial antigens. Human T-bet deficiency thus underlies mycobacterial disease by preventing the development of innate (NK) and innate-like adaptive lymphocytes (iNKT, MAIT, and Vδ2+ γδ T cells) and IFN-γ production by them, with mycobacterium-specific, IFN-γ-producing, purely adaptive CD8+ αß T, and CD4+ αß TH1∗ cells unable to compensate for this deficit.


Subject(s)
Adaptive Immunity , Immunity, Innate , Interferon-gamma/immunology , Mycobacterium/immunology , T-Box Domain Proteins/metabolism , Amino Acid Sequence , Base Sequence , Cell Lineage , Child, Preschool , Chromatin/metabolism , CpG Islands/genetics , DNA Methylation/genetics , Dendritic Cells/metabolism , Epigenesis, Genetic , Female , Homozygote , Humans , INDEL Mutation/genetics , Infant , Interferon-gamma/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Loss of Function Mutation/genetics , Male , Mycobacterium Infections/genetics , Mycobacterium Infections/immunology , Mycobacterium Infections/microbiology , Pedigree , T-Box Domain Proteins/chemistry , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/genetics , T-Lymphocytes, Helper-Inducer/immunology , Transcriptome/genetics
5.
Nat Immunol ; 22(10): 1245-1255, 2021 10.
Article in English | MEDLINE | ID: mdl-34556884

ABSTRACT

Innate lymphoid cells (ILCs) are guardians of mucosal immunity, yet the transcriptional networks that support their function remain poorly understood. We used inducible combinatorial deletion of key transcription factors (TFs) required for ILC development (RORγt, RORα and T-bet) to determine their necessity in maintaining ILC3 identity and function. Both RORγt and RORα were required to preserve optimum effector functions; however, RORα was sufficient to support robust interleukin-22 production among the lymphoid tissue inducer (LTi)-like ILC3 subset, but not natural cytotoxicity receptor (NCR)+ ILC3s. Lymphoid tissue inducer-like ILC3s persisted with only selective loss of phenotype and effector functions even after the loss of both TFs. In contrast, continued RORγt expression was essential to restrain transcriptional networks associated with type 1 immunity within NCR+ ILC3s, which coexpress T-bet. Full differentiation to an ILC1-like population required the additional loss of RORα. Together, these data demonstrate how TF networks integrate within mature ILCs after development to sustain effector functions, imprint phenotype and restrict alternative differentiation programs.


Subject(s)
Immunity, Innate/immunology , Lymphocytes/immunology , Animals , Cell Differentiation/immunology , Cell Lineage/immunology , Cells, Cultured , Female , Gene Expression Regulation/immunology , Immunity, Mucosal/immunology , Lymphoid Tissue/immunology , Male , Mice , Mice, Inbred C57BL , Natural Cytotoxicity Triggering Receptor 1/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , T-Box Domain Proteins/immunology , Transcription Factors/immunology
6.
Nat Immunol ; 22(10): 1231-1244, 2021 10.
Article in English | MEDLINE | ID: mdl-34556887

ABSTRACT

The generation of lymphoid tissues during embryogenesis relies on group 3 innate lymphoid cells (ILC3) displaying lymphoid tissue inducer (LTi) activity and expressing the master transcription factor RORγt. Accordingly, RORγt-deficient mice lack ILC3 and lymphoid structures, including lymph nodes (LN). Whereas T-bet affects differentiation and functions of ILC3 postnatally, the role of T-bet in regulating fetal ILC3 and LN formation remains completely unknown. Using multiple mouse models and single-cell analyses of fetal ILCs and ILC progenitors (ILCP), here we identify a key role for T-bet during embryogenesis and show that its deficiency rescues LN formation in RORγt-deficient mice. Mechanistically, T-bet deletion skews the differentiation fate of fetal ILCs and promotes the accumulation of PLZFhi ILCP expressing central LTi molecules in a RORα-dependent fashion. Our data unveil an unexpected role for T-bet and RORα during embryonic ILC function and highlight that RORγt is crucial in counteracting the suppressive effects of T-bet.


Subject(s)
Cell Differentiation/immunology , Immunity, Innate/immunology , Lymph Nodes/immunology , Lymphocytes/immunology , Nuclear Receptor Subfamily 1, Group F, Member 1/immunology , T-Box Domain Proteins/immunology , Animals , Cell Lineage/immunology , Female , Lymphoid Tissue/immunology , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , T-Lymphocytes, Helper-Inducer/immunology
7.
Nat Immunol ; 22(8): 996-1007, 2021 08.
Article in English | MEDLINE | ID: mdl-34282329

ABSTRACT

During chronic viral infection, CD8+ T cells develop into three major phenotypically and functionally distinct subsets: Ly108+TCF-1+ progenitors, Ly108-CX3CR1- terminally exhausted cells and the recently identified CX3CR1+ cytotoxic effector cells. Nevertheless, how CX3CR1+ effector cell differentiation is transcriptionally and epigenetically regulated remains elusive. Here, we identify distinct gene regulatory networks and epigenetic landscapes underpinning the formation of these subsets. Notably, our data demonstrate that CX3CR1+ effector cells bear a striking similarity to short-lived effector cells during acute infection. Genetic deletion of Tbx21 significantly diminished formation of the CX3CR1+ subset. Importantly, we further identify a previously unappreciated role for the transcription factor BATF in maintaining a permissive chromatin structure that allows the transition from TCF-1+ progenitors to CX3CR1+ effector cells. BATF directly bound to regulatory regions near Tbx21 and Klf2, modulating their enhancer accessibility to facilitate the transition. These mechanistic insights can potentially be harnessed to overcome T cell exhaustion during chronic infection and cancer.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , T-Box Domain Proteins/genetics , T-Lymphocyte Subsets/cytology , Animals , Antigens, Ly/metabolism , CX3C Chemokine Receptor 1/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Female , Hepatocyte Nuclear Factor 1-alpha/metabolism , Kruppel-Like Transcription Factors/genetics , Lymphocytic choriomeningitis virus/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocyte Subsets/immunology
8.
Nat Immunol ; 22(6): 735-745, 2021 06.
Article in English | MEDLINE | ID: mdl-34017124

ABSTRACT

Regulatory T (Treg) cells are a barrier for tumor immunity and a target for immunotherapy. Using single-cell transcriptomics, we found that CD4+ T cells infiltrating primary and metastatic colorectal cancer and non-small-cell lung cancer are highly enriched for two subsets of comparable size and suppressor function comprising forkhead box protein P3+ Treg and eomesodermin homolog (EOMES)+ type 1 regulatory T (Tr1)-like cells also expressing granzyme K and chitinase-3-like protein 2. EOMES+ Tr1-like cells, but not Treg cells, were clonally related to effector T cells and were clonally expanded in primary and metastatic tumors, which is consistent with their proliferation and differentiation in situ. Using chitinase-3-like protein 2 as a subset signature, we found that the EOMES+ Tr1-like subset correlates with disease progression but is also associated with response to programmed cell death protein 1-targeted immunotherapy. Collectively, these findings highlight the heterogeneity of Treg cells that accumulate in primary tumors and metastases and identify a new prospective target for cancer immunotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Clonal Hematopoiesis/immunology , Colorectal Neoplasms/immunology , Lung Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/secondary , Carcinoma, Non-Small-Cell Lung/therapy , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Proliferation/genetics , Chemotherapy, Adjuvant/methods , Chitinases/metabolism , Colectomy , Colon/pathology , Colon/surgery , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Datasets as Topic , Disease Progression , Drug Resistance, Neoplasm/immunology , Female , Flow Cytometry , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic/immunology , Granzymes/metabolism , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Kaplan-Meier Estimate , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Male , Middle Aged , Primary Cell Culture , Programmed Cell Death 1 Receptor/antagonists & inhibitors , RNA-Seq , Single-Cell Analysis , T-Box Domain Proteins/metabolism , T-Lymphocytes, Regulatory/metabolism
9.
Cell ; 172(1-2): 289-304.e18, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29307494

ABSTRACT

Non-coding regions comprise most of the human genome and harbor a significant fraction of risk alleles for neuropsychiatric diseases, yet their functions remain poorly defined. We created a high-resolution map of non-coding elements involved in human cortical neurogenesis by contrasting chromatin accessibility and gene expression in the germinal zone and cortical plate of the developing cerebral cortex. We link distal regulatory elements (DREs) to their cognate gene(s) together with chromatin interaction data and show that target genes of human-gained enhancers (HGEs) regulate cortical neurogenesis and are enriched in outer radial glia, a cell type linked to human cortical evolution. We experimentally validate the regulatory effects of predicted enhancers for FGFR2 and EOMES. We observe that common genetic variants associated with educational attainment, risk for neuropsychiatric disease, and intracranial volume are enriched within regulatory elements involved in cortical neurogenesis, demonstrating the importance of this early developmental process for adult human cognitive function.


Subject(s)
Cerebral Cortex/metabolism , Chromatin Assembly and Disassembly , Gene Expression Regulation, Developmental , Neurogenesis , Neurons/metabolism , Cell Line , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Chromatin/genetics , Chromatin/metabolism , Enhancer Elements, Genetic , Female , Humans , Male , Neurons/cytology , Polymorphism, Genetic , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
10.
Nat Immunol ; 21(5): 567-577, 2020 05.
Article in English | MEDLINE | ID: mdl-32284593

ABSTRACT

Unprimed mice harbor a substantial population of 'memory-phenotype' CD8+ T cells (CD8-MP cells) that exhibit hallmarks of activation and innate-like functional properties. Due to the lack of faithful markers to distinguish CD8-MP cells from bona fide CD8+ memory T cells, the developmental origins and antigen specificities of CD8-MP cells remain incompletely defined. Using deep T cell antigen receptor (TCR) sequencing, we found that the TCRs expressed by CD8-MP cells are highly recurrent and distinct from the TCRs expressed by naive-phenotype CD8+ T cells. CD8-MP clones exhibited reactivity to widely expressed self-ligands. T cell precursors expressing CD8-MP TCRs showed upregulation of the transcription factor Eomes during maturation in the thymus, prior to induction of the full memory phenotype, which is suggestive of a unique program triggered by recognition of self-ligands. Moreover, CD8-MP cells infiltrate oncogene-driven prostate tumors and express high densities of PD-1, which suggests potential roles in antitumor immunity and the response to immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Prostatic Neoplasms/immunology , Receptors, Antigen, T-Cell/genetics , T-Box Domain Proteins/metabolism , Thymus Gland/physiology , Animals , Autoantigens/immunology , Cell Differentiation , Clonal Selection, Antigen-Mediated , Clone Cells , Immunologic Memory , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Programmed Cell Death 1 Receptor/metabolism , T-Box Domain Proteins/genetics , Up-Regulation
11.
Nat Immunol ; 21(7): 766-776, 2020 07.
Article in English | MEDLINE | ID: mdl-32424367

ABSTRACT

Tissue-resident memory T (TRM) cells, functionally distinct from circulating memory T cells, have a critical role in protective immunity in tissues, are more efficacious when elicited after vaccination and yield more effective antitumor immunity, yet the signals that direct development of TRM cells are incompletely understood. Here we show that type 1 regulatory T (Treg) cells, which express the transcription factor T-bet, promote the generation of CD8+ TRM cells. The absence of T-bet-expressing type 1 Treg cells reduces the presence of TRM cells in multiple tissues and increases pathogen burden upon infectious challenge. Using infection models, we show that type 1 Treg cells are specifically recruited to local inflammatory sites via the chemokine receptor CXCR3. Close proximity with effector CD8+ T cells and Treg cell expression of integrin-ß8 endows the bioavailability of transforming growth factor-ß in the microenvironment, thereby promoting the generation of CD8+ TRM cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Communication/immunology , Cell Differentiation/immunology , Immunologic Memory , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/transplantation , Coccidiosis/immunology , Coccidiosis/parasitology , Disease Models, Animal , Eimeria/immunology , Female , Humans , Integrin beta Chains/metabolism , Male , Mice , Mice, Transgenic , Receptors, CXCR3/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/transplantation , Transforming Growth Factor beta/metabolism
12.
Immunity ; 56(10): 2177-2179, 2023 10 10.
Article in English | MEDLINE | ID: mdl-37820578

ABSTRACT

Cardiac lymphatics cooperate with the reparative immune response in myocardial healing after infarction. In this issue of Immunity, Wang and colleagues discover a mechanism underlying this cooperation, dependent on the transcription factor Tbx1 and responsible for the creation of an immunosuppressive niche that mitigates autoimmunity.


Subject(s)
Heart , T-Box Domain Proteins , T-Box Domain Proteins/genetics , Heart/physiology , Myocardium , Transcription Factors
13.
Nat Immunol ; 20(1): 10-17, 2019 01.
Article in English | MEDLINE | ID: mdl-30538328

ABSTRACT

Interleukin 15 (IL-15) is one of the most important cytokines that regulate the biology of natural killer (NK) cells1. Here we identified a signaling pathway-involving the serine-threonine kinase AKT and the transcription factor XBP1s, which regulates unfolded protein response genes2,3-that was activated in response to IL-15 in human NK cells. IL-15 induced the phosphorylation of AKT, which led to the deubiquitination, increased stability and nuclear accumulation of XBP1s protein. XBP1s bound to and recruited the transcription factor T-BET to the gene encoding granzyme B, leading to increased transcription. XBP1s positively regulated the cytolytic activity of NK cells against leukemia cells and was also required for IL-15-mediated NK cell survival through an anti-apoptotic mechanism. Thus, the newly identified IL-15-AKT-XBP1s signaling pathway contributes to enhanced effector functions and survival of human NK cells.


Subject(s)
Interleukin-15/metabolism , Killer Cells, Natural/immunology , Proto-Oncogene Proteins c-akt/metabolism , T-Box Domain Proteins/metabolism , X-Box Binding Protein 1/metabolism , Cell Survival , Cells, Cultured , Cytotoxicity, Immunologic , Gene Expression Regulation , Granzymes/genetics , Granzymes/metabolism , Humans , Phosphorylation , Protein Binding , Protein Stability , Signal Transduction , Ubiquitination , Unfolded Protein Response
14.
Nat Immunol ; 20(8): 980-991, 2019 08.
Article in English | MEDLINE | ID: mdl-31209406

ABSTRACT

Innate lymphoid cells (ILCs) are tissue-resident lymphocytes categorized on the basis of their core regulatory programs and the expression of signature cytokines. Human ILC3s that produce the cytokine interleukin-22 convert into ILC1-like cells that produce interferon-γ in vitro, but whether this conversion occurs in vivo remains unclear. In the present study we found that ILC3s and ILC1s in human tonsils represented the ends of a spectrum that included additional discrete subsets. RNA velocity analysis identified an intermediate ILC3-ILC1 cluster, which had strong directionality toward ILC1s. In humanized mice, the acquisition of ILC1 features by ILC3s showed tissue dependency. Chromatin studies indicated that the transcription factors Aiolos and T-bet cooperated to repress regulatory elements active in ILC3s. A transitional ILC3-ILC1 population was also detected in the human intestine. We conclude that ILC3s undergo conversion into ILC1-like cells in human tissues in vivo, and that tissue factors and Aiolos were required for this process.


Subject(s)
Immunity, Innate/immunology , Interferon-gamma/metabolism , Interleukins/metabolism , Intestinal Mucosa/immunology , Lymphocytes/immunology , Palatine Tonsil/immunology , Animals , Cell Differentiation/immunology , Cells, Cultured , Child , Child, Preschool , Humans , Ikaros Transcription Factor/metabolism , Intestinal Mucosa/cytology , Lymphocytes/classification , Lymphocytes/cytology , Mice , T-Box Domain Proteins/metabolism , Interleukin-22
15.
Immunity ; 55(4): 639-655.e7, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35381213

ABSTRACT

Adaptive CD4+ T helper cells and their innate counterparts, innate lymphoid cells, utilize an identical set of transcription factors (TFs) for their differentiation and functions. However, similarities and differences in the induction of these TFs in related lymphocytes are still elusive. Here, we show that T helper-1 (Th1) cells and natural killer (NK) cells displayed distinct epigenomes at the Tbx21 locus, which encodes T-bet, a critical TF for regulating type 1 immune responses. The initial induction of T-bet in NK precursors was dependent on the NK-specific DNase I hypersensitive site Tbx21-CNS-3, and the expression of the interleukin-18 (IL-18) receptor; IL-18 induced T-bet expression through the transcription factor RUNX3, which bound to Tbx21-CNS-3. By contrast, signal transducer and activator of transcription (STAT)-binding motifs within Tbx21-CNS-12 were critical for IL-12-induced T-bet expression during Th1 cell differentiation both in vitro and in vivo. Thus, type 1 innate and adaptive lymphocytes utilize distinct enhancer elements for their development and differentiation.


Subject(s)
Immunity, Innate , Interleukin-18 , Killer Cells, Natural , Th1 Cells , Cell Differentiation , Interleukin-18/metabolism , Killer Cells, Natural/immunology , T-Box Domain Proteins/metabolism , Th1 Cells/immunology , Transcription Factors/metabolism
16.
Immunity ; 55(2): 290-307.e5, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35090581

ABSTRACT

Tbet+CD11c+ B cells arise during type 1 pathogen challenge, aging, and autoimmunity in mice and humans. Here, we examined the developmental requirements of this B cell subset. In acute infection, T follicular helper (Tfh) cells, but not Th1 cells, drove Tbet+CD11c+ B cell generation through proximal delivery of help. Tbet+CD11c+ B cells developed prior to germinal center (GC) formation, exhibiting phenotypic and transcriptional profiles distinct from GC B cells. Fate tracking revealed that most Tbet+CD11c+ B cells developed independently of GC entry and cell-intrinsic Bcl6 expression. Tbet+CD11c+ and GC B cells exhibited minimal repertoire overlap, indicating distinct developmental pathways. As the infection resolved, Tbet+CD11c+ B cells localized to the marginal zone where splenic retention depended on integrins LFA-1 and VLA-4, forming a competitive memory subset that contributed to antibody production and secondary GC seeding upon rechallenge. Therefore, Tbet+CD11c+ B cells comprise a GC-independent memory subset capable of rapid and robust recall responses.


Subject(s)
B-Lymphocytes/immunology , CD11 Antigens/metabolism , Lymphocyte Subsets/immunology , T Follicular Helper Cells/immunology , T-Box Domain Proteins/metabolism , Virus Diseases/immunology , Animals , Antibodies, Viral/metabolism , B-Lymphocytes/metabolism , Cell Differentiation/immunology , Germinal Center/immunology , Alphainfluenzavirus/immunology , Integrins/metabolism , Lymphocyte Subsets/metabolism , Lymphocytic choriomeningitis virus/immunology , Memory B Cells/immunology , Memory B Cells/metabolism , Mice , Spleen/immunology
17.
Cell ; 166(3): 637-650, 2016 Jul 28.
Article in English | MEDLINE | ID: mdl-27471966

ABSTRACT

Most vertebrate oocytes contain a Balbiani body, a large, non-membrane-bound compartment packed with RNA, mitochondria, and other organelles. Little is known about this compartment, though it specifies germline identity in many non-mammalian vertebrates. We show Xvelo, a disordered protein with an N-terminal prion-like domain, is an abundant constituent of Xenopus Balbiani bodies. Disruption of the prion-like domain of Xvelo, or substitution with a prion-like domain from an unrelated protein, interferes with its incorporation into Balbiani bodies in vivo. Recombinant Xvelo forms amyloid-like networks in vitro. Amyloid-like assemblies of Xvelo recruit both RNA and mitochondria in binding assays. We propose that Xenopus Balbiani bodies form by amyloid-like assembly of Xvelo, accompanied by co-recruitment of mitochondria and RNA. Prion-like domains are found in germ plasm organizing proteins in other species, suggesting that Balbiani body formation by amyloid-like assembly could be a conserved mechanism that helps oocytes function as long-lived germ cells.


Subject(s)
Amyloid/metabolism , Organelle Biogenesis , T-Box Domain Proteins/metabolism , Xenopus Proteins/metabolism , Animals , Benzothiazoles , Female , Fluorescent Dyes , Mitochondria/metabolism , Oocytes/cytology , Organelles/metabolism , Prions/chemistry , Protein Domains , Protein Transport , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sf9 Cells , T-Box Domain Proteins/chemistry , T-Box Domain Proteins/genetics , Thiazoles , Xenopus Proteins/chemistry , Xenopus Proteins/genetics , Xenopus laevis , Zebrafish
18.
Cell ; 164(5): 999-1014, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26875865

ABSTRACT

Transcription factors (TFs) are thought to function with partners to achieve specificity and precise quantitative outputs. In the developing heart, heterotypic TF interactions, such as between the T-box TF TBX5 and the homeodomain TF NKX2-5, have been proposed as a mechanism for human congenital heart defects. We report extensive and complex interdependent genomic occupancy of TBX5, NKX2-5, and the zinc finger TF GATA4 coordinately controlling cardiac gene expression, differentiation, and morphogenesis. Interdependent binding serves not only to co-regulate gene expression but also to prevent TFs from distributing to ectopic loci and activate lineage-inappropriate genes. We define preferential motif arrangements for TBX5 and NKX2-5 cooperative binding sites, supported at the atomic level by their co-crystal structure bound to DNA, revealing a direct interaction between the two factors and induced DNA bending. Complex interdependent binding mechanisms reveal tightly regulated TF genomic distribution and define a combinatorial logic for heterotypic TF regulation of differentiation.


Subject(s)
GATA4 Transcription Factor/metabolism , Homeodomain Proteins/metabolism , Myocardium/cytology , Organogenesis , T-Box Domain Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation , Crystallography, X-Ray , Embryo, Mammalian/metabolism , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Mice , Mice, Transgenic , Models, Molecular , Myocardium/metabolism , Promoter Regions, Genetic , Protein Interaction Domains and Motifs , T-Box Domain Proteins/genetics , Transcription Factors/genetics
19.
Cell ; 167(7): 1734-1749.e22, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27984724

ABSTRACT

Mutation of highly conserved residues in transcription factors may affect protein-protein or protein-DNA interactions, leading to gene network dysregulation and human disease. Human mutations in GATA4, a cardiogenic transcription factor, cause cardiac septal defects and cardiomyopathy. Here, iPS-derived cardiomyocytes from subjects with a heterozygous GATA4-G296S missense mutation showed impaired contractility, calcium handling, and metabolic activity. In human cardiomyocytes, GATA4 broadly co-occupied cardiac enhancers with TBX5, another transcription factor that causes septal defects when mutated. The GATA4-G296S mutation disrupted TBX5 recruitment, particularly to cardiac super-enhancers, concomitant with dysregulation of genes related to the phenotypic abnormalities, including cardiac septation. Conversely, the GATA4-G296S mutation led to failure of GATA4 and TBX5-mediated repression at non-cardiac genes and enhanced open chromatin states at endothelial/endocardial promoters. These results reveal how disease-causing missense mutations can disrupt transcriptional cooperativity, leading to aberrant chromatin states and cellular dysfunction, including those related to morphogenetic defects.


Subject(s)
GATA4 Transcription Factor/genetics , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Chromatin , Enhancer Elements, Genetic , Female , Heart/growth & development , Humans , Induced Pluripotent Stem Cells , Male , Mutation, Missense , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , T-Box Domain Proteins/genetics
20.
Immunity ; 54(2): 276-290.e5, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33434494

ABSTRACT

The oropharyngeal mucosa serves as a perpetual pathogen entry point and a critical site for viral replication and spread. Here, we demonstrate that type 1 innate lymphoid cells (ILC1s) were the major immune force providing early protection during acute oral mucosal viral infection. Using intravital microscopy, we show that ILC1s populated and patrolled the uninfected labial mucosa. ILC1s produced interferon-γ (IFN-γ) in the absence of infection, leading to the upregulation of key antiviral genes, which were downregulated in uninfected animals upon genetic ablation of ILC1s or antibody-based neutralization of IFN-γ. Thus, tonic IFN-γ production generates increased oral mucosal viral resistance even before infection. Our results demonstrate barrier-tissue protection through tissue surveillance in the absence of rearranged-antigen receptors and the induction of an antiviral state during homeostasis. This aspect of ILC1 biology raises the possibility that these cells do not share true functional redundancy with other tissue-resident lymphocytes.


Subject(s)
Interferon-gamma/metabolism , Lymphocytes/immunology , Oropharynx/immunology , Respiratory Mucosa/immunology , Vaccinia virus/physiology , Vaccinia/immunology , Animals , Cells, Cultured , Disease Resistance , Humans , Immunity, Innate , Interferon-gamma/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Box Domain Proteins/genetics , Th1 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL