Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.594
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 22(3): 370-380, 2021 03.
Article in English | MEDLINE | ID: mdl-33574619

ABSTRACT

During chronic infection and cancer, a self-renewing CD8+ T cell subset maintains long-term immunity and is critical to the effectiveness of immunotherapy. These stem-like CD8+ T cells diverge from other CD8+ subsets early after chronic viral infection. However, pathways guarding stem-like CD8+ T cells against terminal exhaustion remain unclear. Here, we show that the gene encoding transcriptional repressor BACH2 is transcriptionally and epigenetically active in stem-like CD8+ T cells but not terminally exhausted cells early after infection. BACH2 overexpression enforced stem-like cell fate, whereas BACH2 deficiency impaired stem-like CD8+ T cell differentiation. Single-cell transcriptomic and epigenomic approaches revealed that BACH2 established the transcriptional and epigenetic programs of stem-like CD8+ T cells. In addition, BACH2 suppressed the molecular program driving terminal exhaustion through transcriptional repression and epigenetic silencing. Thus, our study reveals a new pathway that enforces commitment to stem-like CD8+ lineage and prevents an alternative terminally exhausted cell fate.


Subject(s)
Arenaviridae Infections/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Epigenesis, Genetic , Precursor Cells, T-Lymphoid/metabolism , Transcription, Genetic , Animals , Arenaviridae Infections/genetics , Arenaviridae Infections/immunology , Arenaviridae Infections/virology , Basic-Leucine Zipper Transcription Factors/deficiency , Basic-Leucine Zipper Transcription Factors/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cell Lineage , Cells, Cultured , Chronic Disease , Disease Models, Animal , Host-Pathogen Interactions , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/pathogenicity , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Precursor Cells, T-Lymphoid/immunology , Precursor Cells, T-Lymphoid/virology , Signal Transduction
2.
Immunity ; 57(5): 1005-1018.e7, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38697116

ABSTRACT

Cytokine expression during T cell differentiation is a highly regulated process that involves long-range promoter-enhancer and CTCF-CTCF contacts at cytokine loci. Here, we investigated the impact of dynamic chromatin loop formation within the topologically associating domain (TAD) in regulating the expression of interferon gamma (IFN-γ) and interleukin-22 (IL-22); these cytokine loci are closely located in the genome and are associated with complex enhancer landscapes, which are selectively active in type 1 and type 3 lymphocytes. In situ Hi-C analyses revealed inducible TADs that insulated Ifng and Il22 enhancers during Th1 cell differentiation. Targeted deletion of a 17 bp boundary motif of these TADs imbalanced Th1- and Th17-associated immunity, both in vitro and in vivo, upon Toxoplasma gondii infection. In contrast, this boundary element was dispensable for cytokine regulation in natural killer cells. Our findings suggest that precise cytokine regulation relies on lineage- and developmental stage-specific interactions of 3D chromatin architectures and enhancer landscapes.


Subject(s)
CCCTC-Binding Factor , Cell Differentiation , Interferon-gamma , Interleukin-22 , Interleukins , Th1 Cells , Animals , CCCTC-Binding Factor/metabolism , CCCTC-Binding Factor/genetics , Th1 Cells/immunology , Mice , Cell Differentiation/immunology , Interferon-gamma/metabolism , Binding Sites , Interleukins/metabolism , Interleukins/genetics , Enhancer Elements, Genetic/genetics , Mice, Inbred C57BL , Chromatin/metabolism , Toxoplasmosis/immunology , Toxoplasmosis/parasitology , Toxoplasmosis/genetics , Gene Expression Regulation , Toxoplasma/immunology , Cytokines/metabolism , Cell Lineage , Th17 Cells/immunology
3.
Nat Immunol ; 20(6): 711-723, 2019 06.
Article in English | MEDLINE | ID: mdl-31061530

ABSTRACT

Resting CD4+ T cells are highly resistant to the production of human immunodeficiency virus type 1 (HIV-1). However, the mechanism by which resting CD4+ T cells restrict such production in the late viral replication phase of infection has remained unclear. In this study, we found that the cell membrane metalloprotease TRAB domain-containing protein 2A (TRABD2A) inhibited this production in resting CD4+ T cells by degrading the virion structural precursor polyprotein Gag at the plasma membrane. Depletion or inhibition of metalloprotease activity by TRABD2A profoundly enhanced HIV-1 production in resting CD4+ T cells. TRABD2A expression was much higher in resting CD4+ T cells than in activated CD4+ T cells and was considerably reduced by T cell activation. Moreover, reexpressing TRABD2A reinforced the resistance of activated CD4+ T cells to the production of HIV-1 progeny. Collectively, these results elucidate the molecular mechanism employed by resting CD4+ T cells to potently restrict the assembly and production of HIV-1 progeny.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/virology , HIV-1/physiology , Metalloproteases/genetics , Virus Replication , gag Gene Products, Human Immunodeficiency Virus/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , Cations , Cell Line , Enzyme Activation , Gene Expression , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Membrane Proteins/metabolism , Metalloproteases/antagonists & inhibitors , Metalloproteases/metabolism , Multigene Family , Proteolysis , SAM Domain and HD Domain-Containing Protein 1/genetics , SAM Domain and HD Domain-Containing Protein 1/metabolism , Viral Load
4.
Nat Immunol ; 20(7): 890-901, 2019 07.
Article in English | MEDLINE | ID: mdl-31209400

ABSTRACT

Progenitor-like CD8+ T cells mediate long-term immunity to chronic infection and cancer and respond potently to immune checkpoint blockade. These cells share transcriptional regulators with memory precursor cells, including T cell-specific transcription factor 1 (TCF1), but it is unclear whether they adopt distinct programs to adapt to the immunosuppressive environment. By comparing the single-cell transcriptomes and epigenetic profiles of CD8+ T cells responding to acute and chronic viral infections, we found that progenitor-like CD8+ T cells became distinct from memory precursor cells before the peak of the T cell response. We discovered a coexpression gene module containing Tox that exhibited higher transcriptional activity associated with more abundant active histone marks in progenitor-like cells than memory precursor cells. Moreover, thymocyte selection-associated high mobility group box protein TOX (TOX) promoted the persistence of antiviral CD8+ T cells and was required for the programming of progenitor-like CD8+ T cells. Thus, long-term CD8+ T cell immunity to chronic viral infection requires unique transcriptional and epigenetic programs associated with the transcription factor TOX.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Gene Expression Regulation , Homeodomain Proteins/genetics , Infections/etiology , Single-Cell Analysis , Animals , Biomarkers , Chromatin Immunoprecipitation , Epigenesis, Genetic , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Homeodomain Proteins/metabolism , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immunologic Memory , Infections/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Mice , Time Factors , Transcriptome
5.
Cell ; 165(5): 1120-1133, 2016 May 19.
Article in English | MEDLINE | ID: mdl-27156451

ABSTRACT

Innate lymphoid cells (ILCs) play key roles in host defense, barrier integrity, and homeostasis and mirror adaptive CD4(+) T helper (Th) cell subtypes in both usage of effector molecules and transcription factors. To better understand the relationship between ILC subsets and their Th cell counterparts, we measured genome-wide chromatin accessibility. We find that chromatin in proximity to effector genes is selectively accessible in ILCs prior to high-level transcription upon activation. Accessibility of these regions is acquired in a stepwise manner during development and changes little after in vitro or in vivo activation. Conversely, dramatic chromatin remodeling occurs in naive CD4(+) T cells during Th cell differentiation using a type-2-infection model. This alteration results in a substantial convergence of Th2 cells toward ILC2 regulomes. Our data indicate extensive sharing of regulatory circuitry across the innate and adaptive compartments of the immune system, in spite of their divergent developing pathways.


Subject(s)
Gene Regulatory Networks , Lymphocytes/cytology , Lymphocytes/immunology , Animals , Cell Lineage , Female , Gene Expression Regulation , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Transcriptome
6.
Nat Immunol ; 19(12): 1403-1414, 2018 12.
Article in English | MEDLINE | ID: mdl-30397350

ABSTRACT

Repair of tissue damaged during inflammatory processes is key to the return of local homeostasis and restoration of epithelial integrity. Here we describe CD161+ regulatory T (Treg) cells as a distinct, highly suppressive population of Treg cells that mediate wound healing. These Treg cells were enriched in intestinal lamina propria, particularly in Crohn's disease. CD161+ Treg cells had an all-trans retinoic acid (ATRA)-regulated gene signature, and CD161 expression on Treg cells was induced by ATRA, which directly regulated the CD161 gene. CD161 was co-stimulatory, and ligation with the T cell antigen receptor induced cytokines that accelerated the wound healing of intestinal epithelial cells. We identified a transcription-factor network, including BACH2, RORγt, FOSL2, AP-1 and RUNX1, that controlled expression of the wound-healing program, and found a CD161+ Treg cell signature in Crohn's disease mucosa associated with reduced inflammation. These findings identify CD161+ Treg cells as a population involved in controlling the balance between inflammation and epithelial barrier healing in the gut.


Subject(s)
Intestinal Mucosa/immunology , NK Cell Lectin-Like Receptor Subfamily B/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Tretinoin/immunology , Wound Healing/immunology , Crohn Disease/immunology , Humans
7.
Immunity ; 54(3): 514-525.e6, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33657395

ABSTRACT

MicroRNAs are important regulators of immune responses. Here, we show miR-221 and miR-222 modulate the intestinal Th17 cell response. Expression of miR-221 and miR-222 was induced by proinflammatory cytokines and repressed by the cytokine TGF-ß. Molecular targets of miR-221 and miR-222 included Maf and Il23r, and loss of miR-221 and miR-222 expression shifted the transcriptomic spectrum of intestinal Th17 cells to a proinflammatory signature. Although the loss of miR-221 and miR-222 was tolerated for maintaining intestinal Th17 cell homeostasis in healthy mice, Th17 cells lacking miR-221 and miR-222 expanded more efficiently in response to IL-23. Both global and T cell-specific deletion of miR-221 and miR-222 rendered mice prone to mucosal barrier damage. Collectively, these findings demonstrate that miR-221 and miR-222 are an integral part of intestinal Th17 cell response that are induced after IL-23 stimulation to constrain the magnitude of proinflammatory response.


Subject(s)
Inflammation/immunology , Interleukin-23/metabolism , Intestinal Mucosa/immunology , MicroRNAs/genetics , Th17 Cells/immunology , Animals , Feedback, Physiological , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-maf/metabolism , Receptors, Interleukin/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
8.
Nature ; 629(8014): 1047-1054, 2024 May.
Article in English | MEDLINE | ID: mdl-38778108

ABSTRACT

Wireless modules that provide telecommunications and power-harvesting capabilities enabled by radio-frequency (RF) electronics are vital components of skin-interfaced stretchable electronics1-7. However, recent studies on stretchable RF components have demonstrated that substantial changes in electrical properties, such as a shift in the antenna resonance frequency, occur even under relatively low elastic strains8-15. Such changes lead directly to greatly reduced wireless signal strength or power-transfer efficiency in stretchable systems, particularly in physically dynamic environments such as the surface of the skin. Here we present strain-invariant stretchable RF electronics capable of completely maintaining the original RF properties under various elastic strains using a 'dielectro-elastic' material as the substrate. Dielectro-elastic materials have physically tunable dielectric properties that effectively avert frequency shifts arising in interfacing RF electronics. Compared with conventional stretchable substrate materials, our material has superior electrical, mechanical and thermal properties that are suitable for high-performance stretchable RF electronics. In this paper, we describe the materials, fabrication and design strategies that serve as the foundation for enabling the strain-invariant behaviour of key RF components based on experimental and computational studies. Finally, we present a set of skin-interfaced wireless healthcare monitors based on strain-invariant stretchable RF electronics with a wireless operational distance of up to 30 m under strain.


Subject(s)
Elasticity , Electronics , Equipment Design , Radio Waves , Skin , Stress, Mechanical , Wearable Electronic Devices , Wireless Technology , Humans , Electronics/instrumentation , Wireless Technology/instrumentation , Monitoring, Physiologic/instrumentation
9.
Nat Immunol ; 18(7): 813-823, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28530713

ABSTRACT

The transcriptional programs that guide lymphocyte differentiation depend on the precise expression and timing of transcription factors (TFs). The TF BACH2 is essential for T and B lymphocytes and is associated with an archetypal super-enhancer (SE). Single-nucleotide variants in the BACH2 locus are associated with several autoimmune diseases, but BACH2 mutations that cause Mendelian monogenic primary immunodeficiency have not previously been identified. Here we describe a syndrome of BACH2-related immunodeficiency and autoimmunity (BRIDA) that results from BACH2 haploinsufficiency. Affected subjects had lymphocyte-maturation defects that caused immunoglobulin deficiency and intestinal inflammation. The mutations disrupted protein stability by interfering with homodimerization or by causing aggregation. We observed analogous lymphocyte defects in Bach2-heterozygous mice. More generally, we observed that genes that cause monogenic haploinsufficient diseases were substantially enriched for TFs and SE architecture. These findings reveal a previously unrecognized feature of SE architecture in Mendelian diseases of immunity: heterozygous mutations in SE-regulated genes identified by whole-exome/genome sequencing may have greater significance than previously recognized.


Subject(s)
Autoimmune Diseases/genetics , Basic-Leucine Zipper Transcription Factors/genetics , Immunologic Deficiency Syndromes/genetics , Adrenal Cortex Hormones/therapeutic use , Adult , Autoimmune Diseases/complications , Colitis/complications , Colitis/genetics , Colitis/pathology , Female , Fever/complications , Fever/drug therapy , Fever/genetics , Haploinsufficiency , Heterozygote , Humans , Immunologic Deficiency Syndromes/complications , Lymphopenia/complications , Lymphopenia/genetics , Male , Middle Aged , Mutation , Pancytopenia/complications , Pancytopenia/drug therapy , Pancytopenia/genetics , Pedigree , Polymorphism, Single Nucleotide , Recurrence , Respiratory Tract Infections/complications , Respiratory Tract Infections/diagnostic imaging , Respiratory Tract Infections/genetics , Splenomegaly/complications , Splenomegaly/genetics , Syndrome , Tomography, X-Ray Computed , Young Adult
10.
Immunity ; 53(4): 745-758.e4, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33010223

ABSTRACT

Innate immune responses rely on rapid and precise gene regulation mediated by accessibility of regulatory regions to transcription factors (TFs). In natural killer (NK) cells and other innate lymphoid cells, competent enhancers are primed during lineage acquisition, and formation of de novo enhancers characterizes the acquisition of innate memory in activated NK cells and macrophages. Here, we investigated how primed and de novo enhancers coordinate to facilitate high-magnitude gene induction during acute activation. Epigenomic and transcriptomic analyses of regions near highly induced genes (HIGs) in NK cells both in vitro and in a model of Toxoplasma gondii infection revealed de novo chromatin accessibility and enhancer remodeling controlled by signal-regulated TFs STATs. Acute NK cell activation redeployed the lineage-determining TF T-bet to de novo enhancers, independent of DNA-sequence-specific motif recognition. Thus, acute stimulation reshapes enhancer function through the combinatorial usage and repurposing of both lineage-determining and signal-regulated TFs to ensure an effective response.


Subject(s)
Enhancer Elements, Genetic/genetics , Enhancer Elements, Genetic/immunology , Killer Cells, Natural/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Animals , Chromatin/genetics , Chromatin/immunology , Female , Gene Expression/genetics , Gene Expression/immunology , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Toxoplasma/immunology , Toxoplasmosis/genetics , Toxoplasmosis/immunology
12.
Immunity ; 50(1): 106-120.e10, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30650370

ABSTRACT

CD4+ T helper (Th) differentiation is regulated by diverse inputs, including the vitamin A metabolite retinoic acid (RA). RA acts through its receptor RARα to repress transcription of inflammatory cytokines, but is also essential for Th-mediated immunity, indicating complex effects of RA on Th specification and the outcome of the immune response. We examined the impact of RA on the genome-wide transcriptional response during Th differentiation to multiple subsets. RA effects were subset-selective and were most significant in Th9 cells. RA globally antagonized Th9-promoting transcription factors and inhibited Th9 differentiation. RA directly targeted the extended Il9 locus and broadly modified the Th9 epigenome through RARα. RA-RARα activity limited murine Th9-associated pulmonary inflammation, and human allergic inflammation was associated with reduced expression of RA target genes. Thus, repression of the Th9 program is a major function of RA-RARα signaling in Th differentiation, arguing for a role for RA in interleukin 9 (IL-9) related diseases.


Subject(s)
Hypersensitivity/immunology , Lung/physiology , Pneumonia/immunology , Retinoic Acid Receptor alpha/metabolism , T-Lymphocytes, Helper-Inducer/physiology , Animals , Epigenetic Repression , HEK293 Cells , Humans , Hypersensitivity/genetics , Interleukin-9/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/genetics , Retinoic Acid Receptor alpha/genetics , Signal Transduction , Transcription, Genetic , Tretinoin/metabolism
13.
Cell ; 152(3): 620-32, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23352430

ABSTRACT

DNA double-strand breaks (DSBs) in B lymphocytes arise stochastically during replication or as a result of targeted DNA damage by activation-induced cytidine deaminase (AID). Here we identify recurrent, early replicating, and AID-independent DNA lesions, termed early replication fragile sites (ERFSs), by genome-wide localization of DNA repair proteins in B cells subjected to replication stress. ERFSs colocalize with highly expressed gene clusters and are enriched for repetitive elements and CpG dinucleotides. Although distinct from late-replicating common fragile sites (CFS), the stability of ERFSs and CFSs is similarly dependent on the replication-stress response kinase ATR. ERFSs break spontaneously during replication, but their fragility is increased by hydroxyurea, ATR inhibition, or deregulated c-Myc expression. Moreover, greater than 50% of recurrent amplifications/deletions in human diffuse large B cell lymphoma map to ERFSs. In summary, we have identified a source of spontaneous DNA lesions that drives instability at preferred genomic sites.


Subject(s)
Chromosome Fragile Sites , DNA Replication , Eukaryota/genetics , Genomic Instability , Prokaryotic Cells/physiology , Animals , Biomechanical Phenomena , DNA Repair , Humans
14.
Cell ; 151(5): 981-93, 2012 Nov 21.
Article in English | MEDLINE | ID: mdl-23178119

ABSTRACT

Signaling pathways are intimately involved in cellular differentiation, allowing cells to respond to their environment by regulating gene expression. Although enhancers are recognized as key elements that regulate selective gene expression, the interplay between signaling pathways and actively used enhancer elements is not clear. Here, we use CD4(+) T cells as a model of differentiation, mapping the activity of cell-type-specific enhancer elements in T helper 1 (Th1) and Th2 cells. Our data establish that STAT proteins have a major impact on the activation of lineage-specific enhancers and the suppression of enhancers associated with alternative cell fates. Transcriptome analysis further supports a functional role for enhancers regulated by STATs. Importantly, expression of lineage-defining master regulators in STAT-deficient cells fails to fully recover the chromatin signature of STAT-dependent enhancers. Thus, these findings point to a critical role of STATs as environmental sensors in dynamically molding the specialized enhancer architecture of differentiating cells.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Cell Differentiation , Enhancer Elements, Genetic , STAT Transcription Factors/metabolism , Th1 Cells/cytology , Th2 Cells/cytology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , GATA3 Transcription Factor/metabolism , Genes, Essential , Mice , Mice, Inbred C57BL , STAT Transcription Factors/genetics , T-Box Domain Proteins/metabolism , p300-CBP Transcription Factors/metabolism
15.
Mol Cell ; 75(6): 1229-1242.e5, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31377117

ABSTRACT

Interferon gamma (IFN-γ), critical for host defense and tumor surveillance, requires tight control of its expression. Multiple cis-regulatory elements exist around Ifng along with a non-coding transcript, Ifng-as1 (also termed NeST). Here, we describe two genetic models generated to dissect the molecular functions of this locus and its RNA product. DNA deletion within the Ifng-as1 locus disrupted chromatin organization of the extended Ifng locus, impaired Ifng response, and compromised host defense. Insertion of a polyA signal ablated the Ifng-as1 full-length transcript and impaired host defense, while allowing proper chromatin structure. Transient knockdown of Ifng-as1 also reduced IFN-γ production. In humans, discordant expression of IFNG and IFNG-AS1 was evident in memory T cells, with high expression of this long non-coding RNA (lncRNA) and low expression of the cytokine. These results establish Ifng-as1 as an important regulator of Ifng expression, as a DNA element and transcribed RNA, involved in dynamic and cell state-specific responses to infection.


Subject(s)
Gene Expression Regulation/immunology , Immunologic Memory , Infections/immunology , Interferon-gamma/immunology , RNA, Untranslated/immunology , T-Lymphocytes/immunology , Animals , Chromatin/genetics , Chromatin/immunology , Female , Gene Knockdown Techniques , Infections/genetics , Infections/pathology , Interferon-gamma/genetics , Mice , RNA, Untranslated/genetics , T-Lymphocytes/pathology
16.
Proc Natl Acad Sci U S A ; 121(6): e2309096120, 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38285934

ABSTRACT

Invisibility, a fascinating ability of hiding objects within environments, has attracted broad interest for a long time. However, current invisibility technologies are still restricted to stationary environments and narrow band. Here, we experimentally demonstrate a Chimera metasurface for multiterrain invisibility by synthesizing the natural camouflage traits of various poikilotherms. The metasurface achieves chameleon-like broadband in situ tunable microwave reflection mimicry of realistic water surface, shoal, beach/desert, grassland, and frozen ground from 8 to 12 GHz freely via the circuit-topology-transited mode evolution, while remaining optically transparent as an invisible glass frog. Additionally, the mechanic-driven Chimera metasurface without active electrothermal effect, owning a bearded dragon-like thermal acclimation, can decrease the maximum thermal imaging difference to 3.1 °C in tested realistic terrains, which cannot be recognized by human eyes. Our work transitions camouflage technologies from the constrained scenario to ever-changing terrains and constitutes a big advance toward the new-generation reconfigurable electromagnetics with circuit-topology dynamics.

17.
Immunity ; 46(6): 983-991.e4, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28623086

ABSTRACT

Host defense requires the specification of CD4+ helper T (Th) cells into distinct fates, including Th1 cells that preferentially produce interferon-γ (IFN-γ). IFN-γ, a member of a large family of anti-pathogenic and anti-tumor IFNs, induces T-bet, a lineage-defining transcription factor for Th1 cells, which in turn supports IFN-γ production in a feed-forward manner. Herein, we show that a cell-intrinsic role of T-bet influences how T cells perceive their secreted product in the environment. In the absence of T-bet, IFN-γ aberrantly induced a type I IFN transcriptomic program. T-bet preferentially repressed genes and pathways ordinarily activated by type I IFNs to ensure that its transcriptional response did not evoke an aberrant amplification of type I IFN signaling circuitry, otherwise triggered by its own product. Thus, in addition to promoting Th1 effector commitment, T-bet acts as a repressor in differentiated Th1 cells to prevent abberant autocrine type I IFN and downstream signaling.


Subject(s)
Autocrine Communication , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , T-Box Domain Proteins/metabolism , Th1 Cells/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Humans , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , T-Box Domain Proteins/genetics , Th1 Cells/microbiology , Th1 Cells/virology , Transcriptome
18.
Mol Cell ; 71(6): 1040-1050.e8, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30146314

ABSTRACT

In mammals, gene silencing by the RNA-induced silencing complex (RISC) is a well-understood cytoplasmic posttranscriptional gene regulatory mechanism. Here, we show that embryonic stem cells (ESCs) contain high levels of nuclear AGO proteins and that in ESCs nuclear AGO protein activity allows for the onset of differentiation. In the nucleus, AGO proteins interact with core RISC components, including the TNRC6 proteins and the CCR4-NOT deadenylase complex. In contrast to cytoplasmic miRNA-mediated gene silencing that mainly operates on cis-acting elements in mRNA 3' untranslated (UTR) sequences, in the nucleus AGO binding in the coding sequence and potentially introns also contributed to post-transcriptional gene silencing. Thus, nuclear localization of AGO proteins in specific cell types leads to a previously unappreciated expansion of the miRNA-regulated transcriptome.


Subject(s)
Argonaute Proteins/physiology , Gene Silencing/physiology , MicroRNAs/physiology , Animals , Argonaute Proteins/genetics , Cell Differentiation/genetics , Cell Line , Cell Nucleus , Cytoplasm , Embryonic Stem Cells/metabolism , Humans , Mammals , Mice , MicroRNAs/genetics , RNA Interference , RNA Stability , RNA, Messenger , RNA, Small Interfering , RNA-Binding Proteins , RNA-Induced Silencing Complex/genetics , RNA-Induced Silencing Complex/metabolism , Transcription Factors
19.
Mol Cell ; 71(1): 129-141.e8, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29979962

ABSTRACT

The enhancer regions of the myogenic master regulator MyoD give rise to at least two enhancer RNAs. Core enhancer eRNA (CEeRNA) regulates transcription of the adjacent MyoD gene, whereas DRReRNA affects expression of Myogenin in trans. We found that DRReRNA is recruited at the Myogenin locus, where it colocalizes with Myogenin nascent transcripts. DRReRNA associates with the cohesin complex, and this association correlates with its transactivating properties. Despite being expressed in undifferentiated cells, cohesin is not loaded on Myogenin until the cells start expressing DRReRNA, which is then required for cohesin chromatin recruitment and maintenance. Functionally, depletion of either cohesin or DRReRNA reduces chromatin accessibility, prevents Myogenin activation, and hinders muscle cell differentiation. Thus, DRReRNA ensures spatially appropriate cohesin loading in trans to regulate gene expression.


Subject(s)
Cell Cycle Proteins/biosynthesis , Chromosomal Proteins, Non-Histone/biosynthesis , Enhancer Elements, Genetic , Muscle, Skeletal/metabolism , Myogenin/biosynthesis , RNA, Untranslated/metabolism , Transcription, Genetic , Animals , Cell Cycle Proteins/genetics , Cell Differentiation , Chromatin/genetics , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/genetics , HEK293 Cells , Humans , Mice , Muscle, Skeletal/cytology , MyoD Protein/biosynthesis , MyoD Protein/genetics , Myogenin/genetics , RNA, Untranslated/genetics , Cohesins
20.
J Neurosci ; 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39060175

ABSTRACT

Protein synthesis in response to neuronal activity, known as activity-dependent translation, is critical for synaptic plasticity and memory formation. However, the signaling cascades that couple neuronal activity to the translational events remains elusive. In this study, we identified the role of calmodulin (CaM), a conserved Ca2+-binding protein, in rRNA biogenesis in neurons. We found the CaM-regulated rRNA synthesis is Ca2+-dependent and necessary for nascent protein synthesis and axon growth in hippocampal neurons. Mechanistically, CaM interacts with nucleolar DDX21 in a Ca2+-dependent manner to regulate nascent rRNA transcription within nucleoli. We further found CaM alters the conformation of DDX21 to liberate the DDX21-sequestered RPA194, the catalytic subunit of RNA polymerase I, to facilitate transcription of rDNA. Using high-throughput screening, we identified the small molecules Batefenterol and Indacaterol that attenuate the CaM-DDX21 interaction and suppress nascent rRNA synthesis and axon growth in hippocampal neurons. These results unveiled the previously unrecognized role of CaM as a messenger to link the activity-induced Ca2+ influx to the nucleolar events essential for protein synthesis. We thus identified the ability of CaM to transmit information to the nucleoli of neurons in response to stimulation.Significance statement Protein synthesis in response to neuronal activity, known as activity-dependent translation, is critical for synaptic plasticity and long-term memory formation. In this study, we identify the novel role of calmodulin (CaM), a highly conserved Ca2+-binding protein, which is well-known by regulating myriad vital biological processes, in activity-dependent translation by regulating rRNA synthesis in neurons. We find that CaM can shuttle into the nucleolus upon depolarization and modulate the activity-induced de novo rRNA biogenesis, which is associated with ribosome assembly and protein synthesis in neurons. Mechanistically, CaM interacts with DDX21, an RNA helicase directly associated with Pol I subunit, to regulate the transcription of rDNA. Our study demonstrates CaM as a messenger linking neuronal activity to ribosome-dependent protein biosynthesis.

SELECTION OF CITATIONS
SEARCH DETAIL