Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Genome Res ; 32(7): 1328-1342, 2022 07.
Article in English | MEDLINE | ID: mdl-34162697

ABSTRACT

Broad domains of H3K4 methylation have been associated with consistent expression of tissue-specific, cell identity, and tumor suppressor genes. Here, we identified broad domain-associated genes in healthy human thymic T cell populations and a collection of T cell acute lymphoblastic leukemia (T-ALL) primary samples and cell lines. We found that broad domains are highly dynamic throughout T cell differentiation, and their varying breadth allows the distinction between normal and neoplastic cells. Although broad domains preferentially associate with cell identity and tumor suppressor genes in normal thymocytes, they flag key oncogenes in T-ALL samples. Moreover, the expression of broad domain-associated genes, both coding and noncoding, is frequently deregulated in T-ALL. Using two distinct leukemic models, we showed that the ectopic expression of T-ALL oncogenic transcription factor preferentially impacts the expression of broad domain-associated genes in preleukemic cells. Finally, an H3K4me3 demethylase inhibitor differentially targets T-ALL cell lines depending on the extent and number of broad domains. Our results show that the regulation of broad H3K4me3 domains is associated with leukemogenesis, and suggest that the presence of these structures might be used for epigenetic prioritization of cancer-relevant genes, including long noncoding RNAs.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Epigenesis, Genetic , Histones/metabolism , Humans , Oncogenes , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics
2.
Nucleic Acids Res ; 49(5): 2488-2508, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33533919

ABSTRACT

The ubiquitous family of dimeric transcription factors AP-1 is made up of Fos and Jun family proteins. It has long been thought to operate principally at gene promoters and how it controls transcription is still ill-understood. The Fos family protein Fra-1 is overexpressed in triple negative breast cancers (TNBCs) where it contributes to tumor aggressiveness. To address its transcriptional actions in TNBCs, we combined transcriptomics, ChIP-seqs, machine learning and NG Capture-C. Additionally, we studied its Fos family kin Fra-2 also expressed in TNBCs, albeit much less. Consistently with their pleiotropic effects, Fra-1 and Fra-2 up- and downregulate individually, together or redundantly many genes associated with a wide range of biological processes. Target gene regulation is principally due to binding of Fra-1 and Fra-2 at regulatory elements located distantly from cognate promoters where Fra-1 modulates the recruitment of the transcriptional co-regulator p300/CBP and where differences in AP-1 variant motif recognition can underlie preferential Fra-1- or Fra-2 bindings. Our work also shows no major role for Fra-1 in chromatin architecture control at target gene loci, but suggests collaboration between Fra-1-bound and -unbound enhancers within chromatin hubs sometimes including promoters for other Fra-1-regulated genes. Our work impacts our view of AP-1.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-fos/metabolism , Triple Negative Breast Neoplasms/genetics , Binding Sites , Cell Line, Tumor , Chromatin/chemistry , Chromatin/metabolism , Epigenesis, Genetic , Fos-Related Antigen-2/metabolism , Humans , Nucleotide Motifs , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fos/physiology , Transcription Factor AP-1/metabolism , Triple Negative Breast Neoplasms/metabolism , p300-CBP Transcription Factors/metabolism
3.
Cell Rep ; 32(7): 108048, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32814051

ABSTRACT

During thymic development and upon peripheral activation, T cells undergo extensive phenotypic and functional changes coordinated by lineage-specific developmental programs. To characterize the regulatory landscape controlling T cell identity, we perform a wide epigenomic and transcriptional analysis of mouse thymocytes and naive CD4 differentiated T helper cells. Our investigations reveal a dynamic putative enhancer landscape, and we could validate many of the enhancers using the high-throughput CapStarr sequencing (CapStarr-seq) approach. We find that genes using multiple promoters display increased enhancer usage, suggesting that apparent "enhancer redundancy" might relate to isoform selection. Furthermore, we can show that two Runx3 promoters display long-range interactions with specific enhancers. Finally, our analyses suggest a novel function for the PRC2 complex in the control of alternative promoter usage. Altogether, our study has allowed for the mapping of an exhaustive set of active enhancers and provides new insights into their function and that of PRC2 in controlling promoter choice during T cell differentiation.


Subject(s)
Polycomb-Group Proteins/genetics , T-Lymphocytes/metabolism , Animals , Cell Differentiation , Male , Mice
4.
Mol Cancer Res ; 17(10): 1999-2014, 2019 10.
Article in English | MEDLINE | ID: mdl-31300541

ABSTRACT

The architectural chromatin protein HMGA1 and the transcription factor Fra-1 are both overexpressed in aggressive triple-negative breast cancers (TNBC), where they both favor epithelial-to-mesenchymal transition, invasion, and metastasis. We therefore explored the possibility that Fra-1 might be involved in enhanced transcription of the HMGA1 gene in TNBCs by exploiting cancer transcriptome datasets and resorting to functional studies combining RNA interference, mRNA and transcriptional run-on assays, chromatin immunoprecipitation, and chromosome conformation capture approaches in TNBC model cell lines. Our bioinformatic analysis indicated that Fra-1 and HMGA1 expressions positively correlate in primary samples of patients with TNBC. Our functional studies showed that Fra-1 regulates HMGA1 mRNA expression at the transcriptional level via binding to enhancer elements located in the last two introns of the gene. Although Fra-1 binding is required for p300/CBP recruitment at the enhancer domain, this recruitment did not appear essential for Fra-1-stimulated HMGA1 gene expression. Strikingly, Fra-1 binding is required for efficient recruitment of RNA Polymerase II at the HMGA1 promoter. This is permitted owing to chromatin interactions bringing about the intragenic Fra-1-binding enhancers and the gene promoter region. Fra-1 is, however, not instrumental for chromatin loop formation at the HMGA1 locus but rather exerts its transcriptional activity by exploiting chromatin interactions preexisting to its binding. IMPLICATIONS: We demonstrate that Fra-1 bound to an intragenic enhancer region is required for RNA Pol II recruitement at the HMGA1 promoter. Thereby, we provide novel insights into the mechanisms whereby Fra-1 exerts its prooncogenic transcriptional actions in the TNBC pathologic context.


Subject(s)
HMGA1a Protein/genetics , Oncogenes/genetics , Transcription Factor AP-1/genetics , Transcription, Genetic/genetics , Triple Negative Breast Neoplasms/genetics , Cell Line, Tumor , Female , Humans
5.
Nucleic Acids Res ; 47(2): 700-715, 2019 01 25.
Article in English | MEDLINE | ID: mdl-30476274

ABSTRACT

Mammalian-wide interspersed repeats (MIRs) are retrotransposed elements of mammalian genomes. Here, we report the specific binding of zinc finger protein ZNF768 to the sequence motif GCTGTGTG (N20) CCTCTCTG in the core region of MIRs. ZNF768 binding is preferentially associated with euchromatin and promoter regions of genes. Binding was observed for genes expressed in a cell type-specific manner in human B cell line Raji and osteosarcoma U2OS cells. Mass spectrometric analysis revealed binding of ZNF768 to Elongator components Elp1, Elp2 and Elp3 and other nuclear factors. The N-terminus of ZNF768 contains a heptad repeat array structurally related to the C-terminal domain (CTD) of RNA polymerase II. This array evolved in placental animals but not marsupials and monotreme species, displays species-specific length variations, and possibly fulfills CTD related functions in gene regulation. We propose that the evolution of MIRs and ZNF768 has extended the repertoire of gene regulatory mechanisms in mammals and that ZNF768 binding is associated with cell type-specific gene expression.


Subject(s)
Retroelements , Transcription Factors/metabolism , Transcription, Genetic , Binding Sites , Cell Line, Tumor , Cell Survival , DNA/chemistry , DNA/metabolism , Euchromatin/metabolism , Gene Expression Regulation , Humans , Nucleotide Motifs , Repetitive Sequences, Nucleic Acid , Transcription Factors/chemistry
6.
Mol Cell ; 69(1): 48-61.e6, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29304333

ABSTRACT

The carboxy-terminal domain (CTD) of RNA polymerase (Pol) II is composed of a repetition of YSPTSPS heptads and functions as a loading platform for protein complexes that regulate transcription, splicing, and maturation of RNAs. Here, we studied mammalian CTD mutants to analyze the function of tyrosine1 residues in the transcription cycle. Mutation of 3/4 of the tyrosine residues (YFFF mutant) resulted in a massive read-through transcription phenotype in the antisense direction of promoters as well as in the 3' direction several hundred kilobases downstream of genes. The YFFF mutant shows reduced Pol II at promoter-proximal pause sites, a loss of interaction with the Mediator and Integrator complexes, and impaired recruitment of these complexes to chromatin. Consistent with these observations, Pol II loading at enhancers and maturation of snRNAs are altered in the YFFF context genome-wide. We conclude that tyrosine1 residues of the CTD control termination of transcription by Pol II.


Subject(s)
RNA Polymerase II/genetics , RNA, Messenger/biosynthesis , Transcription Termination, Genetic/physiology , Transcription, Genetic/physiology , Tyrosine/genetics , Cell Line, Tumor , Chromatin/metabolism , Humans , Mutation/genetics , Promoter Regions, Genetic/genetics , RNA Polymerase II/metabolism , RNA, Small Nuclear/genetics
7.
Nat Commun ; 7: 11841, 2016 06 17.
Article in English | MEDLINE | ID: mdl-27312418

ABSTRACT

NFATc1 plays a critical role in double-negative thymocyte survival and differentiation. However, the signals that regulate Nfatc1 expression are incompletely characterized. Here we show a developmental stage-specific differential expression pattern of Nfatc1 driven by the distal (P1) or proximal (P2) promoters in thymocytes. Whereas, preTCR-negative thymocytes exhibit only P2 promoter-derived Nfatc1ß expression, preTCR-positive thymocytes express both Nfatc1ß and P1 promoter-derived Nfatc1α transcripts. Inducing NFATc1α activity from P1 promoter in preTCR-negative thymocytes, in addition to the NFATc1ß from P2 promoter impairs thymocyte development resulting in severe T-cell lymphopenia. In addition, we show that NFATc1 activity suppresses the B-lineage potential of immature thymocytes, and consolidates their differentiation to T cells. Further, in the pTCR-positive DN3 cells, a threshold level of NFATc1 activity is vital in facilitating T-cell differentiation and to prevent Notch3-induced T-acute lymphoblastic leukaemia. Altogether, our results show NFATc1 activity is crucial in determining the T-cell fate of thymocytes.


Subject(s)
Lymphopenia/immunology , NFATC Transcription Factors/immunology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Receptor, Notch3/immunology , T-Lymphocytes/immunology , Thymocytes/immunology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Differentiation , Cell Lineage/genetics , Cell Lineage/immunology , Cell Survival , Gene Expression Regulation, Developmental/immunology , Lymphopenia/genetics , Lymphopenia/pathology , Mice , Mice, Knockout , NFATC Transcription Factors/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Promoter Regions, Genetic , Protein Isoforms/genetics , Protein Isoforms/immunology , Receptor, Notch3/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Signal Transduction , T-Lymphocytes/cytology , Thymocytes/cytology , Thymus Gland/cytology , Thymus Gland/immunology
8.
Bioinformatics ; 32(16): 2528-30, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27153642

ABSTRACT

UNLABELLED: We describe an R package designed for processing aligned reads from chromatin-oriented high-throughput sequencing experiments. Pasha (preprocessing of aligned sequences from HTS analyses) allows easy manipulation of aligned reads from short-read sequencing technologies (ChIP-seq, FAIRE-seq, MNase-Seq, …) and offers innovative approaches such as ChIP-seq reads elongation, nucleosome midpoint piling strategy for positioning analyses, or the ability to subset paired-end reads by groups of insert size that can contain biologically relevant information. AVAILABILITY AND IMPLEMENTATION: Pasha is a multi-platform R package, available on CRAN repositories under GPL-3 license (https://cran.r-project.org/web/packages/Pasha/). CONTACTS: rfenouil@gmail.com or jean-christophe.andrau@igmm.cnrs.fr SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Chromatin , High-Throughput Nucleotide Sequencing , Software , Nucleosomes
9.
Transcription ; 6(5): 91-101, 2015.
Article in English | MEDLINE | ID: mdl-26566685

ABSTRACT

Dynamic modification of heptad-repeats with the consensus sequence Tyr1-Ser2-Pro3-Thr4-Ser5-Pro6-Ser7 of RNA polymerase II (RNAPII) C-terminal domain (CTD) regulates transcription-coupled processes. Mass spectrometry analysis revealed that K7-residues in non-consensus repeats of human RNAPII are modified by acetylation, or mono-, di-, and tri-methylation. K7ac, K7me2, and K7me3 were found exclusively associated with phosphorylated CTD peptides, while K7me1 occurred also in non-phosphorylated CTD. The monoclonal antibody 1F5 recognizes K7me1/2 residues in CTD and reacts with RNAPIIA. Treatment of cellular extracts with phosphatase or of cells with the kinase inhibitor flavopiridol unmasked the K7me1/2 epitope in RNAPII0, consistent with the association of K7me1/2 marks with phosphorylated CTD peptides. Genome-wide profiling revealed high levels of K7me1/2 marks at the transcriptional start site of genes for sense and antisense transcribing RNAPII. The new K7 modifications further expand the mammalian CTD code to allow regulation of differential gene expression.


Subject(s)
Lysine/metabolism , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , Acetylation , Antibodies, Monoclonal/metabolism , Cell Line , Gene Expression Regulation , Humans , Mass Spectrometry , Methylation , Models, Molecular , Protein Structure, Tertiary , RNA Polymerase II/genetics , Transcription Initiation, Genetic
10.
Nat Commun ; 6: 6905, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25872643

ABSTRACT

Cell-type specific regulation of gene expression requires the activation of promoters by distal genomic elements defined as enhancers. The identification and the characterization of enhancers are challenging in mammals due to their genome complexity. Here we develop CapStarr-Seq, a novel high-throughput strategy to quantitatively assess enhancer activity in mammals. This approach couples capture of regions of interest to previously developed Starr-seq technique. Extensive assessment of CapStarr-seq demonstrates accurate quantification of enhancer activity. Furthermore, we find that enhancer strength is associated with binding complexity of tissue-specific transcription factors and super-enhancers, while additive enhancer activity isolates key genes involved in cell identity and function. The CapStarr-Seq thus provides a fast and cost-effective approach to assess the activity of potential enhancers for a given cell type and will be helpful in decrypting transcription regulation mechanisms.


Subject(s)
Enhancer Elements, Genetic/genetics , Gene Expression Regulation/genetics , Gene Expression/genetics , High-Throughput Nucleotide Sequencing/methods , Transcription Factors/genetics , Animals , Chromatin Immunoprecipitation , Male , Mice , NIH 3T3 Cells , Promoter Regions, Genetic/genetics , Sequence Analysis, DNA/methods
11.
Nat Commun ; 6: 6094, 2015 Jan 23.
Article in English | MEDLINE | ID: mdl-25615415

ABSTRACT

T-cell acute lymphoblastic leukaemias (T-ALL) are aggressive malignant proliferations characterized by high relapse rates and great genetic heterogeneity. TAL1 is amongst the most frequently deregulated oncogenes. Yet, over half of the TAL1(+) cases lack TAL1 lesions, suggesting unrecognized (epi)genetic deregulation mechanisms. Here we show that TAL1 is normally silenced in the T-cell lineage, and that the polycomb H3K27me3-repressive mark is focally diminished in TAL1(+) T-ALLs. Sequencing reveals that >20% of monoallelic TAL1(+) patients without previously known alterations display microinsertions or RAG1/2-mediated episomal reintegration in a single site 5' to TAL1. Using 'allelic-ChIP' and CrispR assays, we demonstrate that such insertions induce a selective switch from H3K27me3 to H3K27ac at the inserted but not the germline allele. We also show that, despite a considerable mechanistic diversity, the mode of oncogenic TAL1 activation, rather than expression levels, impact on clinical outcome. Altogether, these studies establish site-specific epigenetic desilencing as a mechanism of oncogenic activation.


Subject(s)
Alleles , Gene Expression Regulation, Leukemic , Polycomb-Group Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Acetylation , Adult , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chromatin Immunoprecipitation , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Genetic Loci , Histones/metabolism , Homeodomain Proteins/metabolism , Humans , Jurkat Cells , Methylation , Molecular Sequence Data , Mutagenesis, Insertional , Nuclear Proteins/metabolism , Plasmids/genetics , Polycomb-Group Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Survival Analysis , T-Cell Acute Lymphocytic Leukemia Protein 1 , Treatment Outcome
12.
Elife ; 3: e02105, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24842994

ABSTRACT

In mammals, the carboxy-terminal domain (CTD) of RNA polymerase (Pol) II consists of 52 conserved heptapeptide repeats containing the consensus sequence Tyr1-Ser2-Pro3-Thr4-Ser5-Pro6-Ser7. Post-translational modifications of the CTD coordinate the transcription cycle and various steps of mRNA maturation. Here we describe Tyr1 phosphorylation (Tyr1P) as a hallmark of promoter (5' associated) Pol II in mammalian cells, in contrast to what was described in yeast. Tyr1P is predominantly found in antisense orientation at promoters but is also specifically enriched at active enhancers. Mutation of Tyr1 to phenylalanine (Y1F) prevents the formation of the hyper-phosphorylated Pol IIO form, induces degradation of Pol II to the truncated Pol IIB form, and results in a lethal phenotype. Our results suggest that Tyr1P has evolved specialized and essential functions in higher eukaryotes associated with antisense promoter and enhancer transcription, and Pol II stability.DOI: http://dx.doi.org/10.7554/eLife.02105.001.


Subject(s)
Antisense Elements (Genetics) , Enhancer Elements, Genetic , Promoter Regions, Genetic , RNA Polymerase II/metabolism , Tyrosine/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , Humans , Mutation , Phosphorylation , RNA Polymerase II/chemistry , RNA Polymerase II/genetics
13.
BMC Genomics ; 14: 914, 2013 Dec 23.
Article in English | MEDLINE | ID: mdl-24365181

ABSTRACT

BACKGROUND: Divergent transcription is a wide-spread phenomenon in mammals. For instance, short bidirectional transcripts are a hallmark of active promoters, while longer transcripts can be detected antisense from active genes in conditions where the RNA degradation machinery is inhibited. Moreover, many described long non-coding RNAs (lncRNAs) are transcribed antisense from coding gene promoters. However, the general significance of divergent lncRNA/mRNA gene pair transcription is still poorly understood. Here, we used strand-specific RNA-seq with high sequencing depth to thoroughly identify antisense transcripts from coding gene promoters in primary mouse tissues. RESULTS: We found that a substantial fraction of coding-gene promoters sustain divergent transcription of long non-coding RNA (lncRNA)/mRNA gene pairs. Strikingly, upstream antisense transcription is significantly associated with genes related to transcriptional regulation and development. Their promoters share several characteristics with those of transcriptional developmental genes, including very large CpG islands, high degree of conservation and epigenetic regulation in ES cells. In-depth analysis revealed a unique GC skew profile at these promoter regions, while the associated coding genes were found to have large first exons, two genomic features that might enforce bidirectional transcription. Finally, genes associated with antisense transcription harbor specific H3K79me2 epigenetic marking and RNA polymerase II enrichment profiles linked to an intensified rate of early transcriptional elongation. CONCLUSIONS: We concluded that promoters of a class of transcription regulators are characterized by a specialized transcriptional control mechanism, which is directly coupled to relaxed bidirectional transcription.


Subject(s)
Antisense Elements (Genetics) , Promoter Regions, Genetic , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , Transcription, Genetic , Animals , Base Composition , Chromatin/genetics , CpG Islands , Epigenesis, Genetic , Exons , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Sequence Analysis, RNA , Thymocytes
14.
Methods ; 63(1): 3-17, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23597756

ABSTRACT

The explosion of high throughput sequencing technologies marked a turn in our way of understanding the complexity and diversity of the transcriptome, including noncoding transcription dependent on RNA polymerase II. Many new ncRNA populations were described in recent years, including for example TSS RNAs, lincRNAs, eRNAs, PROMPTS and several others. Besides the advances in the average depth coverage of RNA-seq experiments, various additional protocols are now available that can be used to address qualitative and quantitative aspects of the noncoding transcriptome complexity and function. In this review, we will focus on methods allowing isolation and characterization of complex RNA populations using sequencing based approaches, including conventional strategies already used for coding genome and more specific developments allowing, for example, the study of nascent strand transcription, protein-bound or structured RNAs.


Subject(s)
Genetic Variation , High-Throughput Nucleotide Sequencing , RNA, Long Noncoding/genetics , RNA, Untranslated/genetics , Animals , Base Sequence , Gene Expression Profiling , Genome , Sequence Analysis, RNA , Transcriptome/genetics
15.
J Virol ; 87(10): 5523-39, 2013 May.
Article in English | MEDLINE | ID: mdl-23468497

ABSTRACT

Hepatitis C virus (HCV) nonstructural protein 5A (NS5A) is involved in regulating viral replication through its direct interaction with the HCV RNA-dependent RNA polymerase. NS5A also alters infected cell metabolism through complex interactions with numerous host cell proteins. NS5A has furthermore been suggested to act as a transcriptional activator, although the impact on viral replication is unclear. To study this, HCV NS5A variants were amplified from hepatic tissue from an HCV-infected patient, and their abilities to activate gene transcription were analyzed in a single-hybrid yeast (Saccharomyces cerevisiae) model. Different variants isolated from the same patient displayed different transactivational activities. When these variants were inserted into the HCV subgenomic replicon system, they demonstrated various levels of RNA replication, which correlated with their transactivational activities. We showed that the C-terminal fragment of NS5A was localized to the nucleus and that a functional NS5A nuclear localization signal and cellular caspase activity were required for this process. Furthermore, nuclear localization of NS5A was necessary for viral replication. Finally, we demonstrate that nuclear NS5A binds to host cell promoters of several genes previously identified as important for efficient HCV RNA replication, inducing their transcription. Taken together, these results demonstrate a new mechanism by which HCV modulates its cellular environment, thereby enhancing viral replication.


Subject(s)
Hepacivirus/physiology , Host-Pathogen Interactions , Transcriptional Activation , Viral Nonstructural Proteins/metabolism , Virus Replication , Aged, 80 and over , Cell Nucleus/chemistry , Hepacivirus/genetics , Hepacivirus/isolation & purification , Hepacivirus/pathogenicity , Hepatitis C, Chronic/virology , Humans , Male , Nuclear Localization Signals , Promoter Regions, Genetic , Protein Binding , Saccharomyces cerevisiae/genetics , Viral Nonstructural Proteins/genetics
16.
Cell Host Microbe ; 12(4): 571-84, 2012 Oct 18.
Article in English | MEDLINE | ID: mdl-23084923

ABSTRACT

Type I interferons (IFNs) are central to antiviral defense, but how they orchestrate immune cell function is incompletely understood. We determined that IFNs produced during murine cytomegalovirus (MCMV) infection differentially affect dendritic cells (DCs) and natural killer (NK) cells. IFNs induce cell-intrinsic responses in DCs, activating antiproliferative, antiviral, and lymphocyte-activating gene networks, consistent with high activity of the transcription factor STAT1 in these cells. By comparison, NK cells exhibit lower STAT1 expression and reduced IFN responsiveness. Rather, IFNs indirectly affect NK cells by inducing IL-15, which activates the transcription factor E2F and stimulates genes promoting cell expansion. IFN cell-intrinsic responses are necessary in DCs, but not NK cells, for MCMV resistance. Thus, sensitivity to IFN-induced cytokines and differences in IFN receptor signaling program immune cells to mount distinct responses that promote viral control.


Subject(s)
Dendritic Cells/immunology , Interferon Type I/immunology , Killer Cells, Natural/immunology , Muromegalovirus/immunology , Animals , Cells, Cultured , Flow Cytometry , Gene Expression Profiling , Herpesviridae Infections/immunology , Herpesviridae Infections/veterinary , Mice , Mice, Inbred C57BL , Models, Biological , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...