Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Annu Rev Immunol ; 32: 489-511, 2014.
Article in English | MEDLINE | ID: mdl-24555473

ABSTRACT

A fundamental property of cells of the innate immune system is their ability to elicit a transcriptional response to a microbial stimulus or danger signal with a high degree of cell type and stimulus specificity. The selective response activates effector pathways to control the insult and plays a central role in regulating adaptive immunity through the differential regulation of cytokine genes. Selectivity is dictated by signaling pathways and their transcription factor targets. However, a growing body of evidence supports models in which different subsets of genes exhibit distinct chromatin features that play active roles in shaping the response. Chromatin also participates in innate memory mechanisms that can promote tolerance to a stimulus or prime cells for a more robust response. These findings have generated interest in the capacity to modulate chromatin regulators with small-molecule compounds for the treatment of diseases associated with innate or adaptive immunity.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , Immunity, Innate/physiology , Animals , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunologic Memory/genetics , Immunologic Memory/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/therapy , Organ Specificity/genetics , Organ Specificity/immunology , Transcription, Genetic
2.
Cell ; 173(5): 1150-1164.e14, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29706544

ABSTRACT

Tandem repeats (TRs) are generated by DNA replication errors and retain a high level of instability, which in principle would make them unsuitable for integration into gene regulatory networks. However, the appearance of DNA sequence motifs recognized by transcription factors may turn TRs into functional cis-regulatory elements, thus favoring their stabilization in genomes. Here, we show that, in human cells, the transcriptional repressor ZEB1, which promotes the maintenance of mesenchymal features largely by suppressing epithelial genes and microRNAs, occupies TRs harboring dozens of copies of its DNA-binding motif within genomic loci relevant for maintenance of epithelial identity. The deletion of one such TR caused quasi-mesenchymal cancer cells to reacquire epithelial features, partially recapitulating the effects of ZEB1 gene deletion. These data demonstrate that the high density of identical motifs in TRs can make them suitable platforms for recruitment of transcriptional repressors, thus promoting their exaptation into pre-existing cis-regulatory networks.


Subject(s)
Tandem Repeat Sequences/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism , Adult , Animals , Base Sequence , Cell Line, Tumor , Chromatin Immunoprecipitation , Female , Gene Expression , Humans , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Mouth Mucosa/metabolism , Polymorphism, Single Nucleotide , Protein Binding , Transcription Factors/metabolism , Zinc Finger E-box-Binding Homeobox 1/deficiency , Zinc Finger E-box-Binding Homeobox 1/genetics
3.
Nat Immunol ; 21(4): 388-399, 2020 04.
Article in English | MEDLINE | ID: mdl-32205878

ABSTRACT

Understanding the mechanisms that modulate helper T lymphocyte functions is crucial to decipher normal and pathogenic immune responses in humans. To identify molecular determinants influencing the pathogenicity of T cells, we separated ex vivo-isolated primary human memory T lymphocytes on the basis of their ability to produce high levels of inflammatory cytokines. We found that the inflammatory, cytokine-producing phenotype of memory T lymphocytes was defined by a specific core gene signature and was mechanistically regulated by the constitutive activation of the NF-κB pathway and by the expression of the transcriptional repressor BHLHE40. BHLHE40 attenuated the expression of anti-inflammatory factors, including miR-146a, a negative regulator of NF-κB activation and ZC3H12D, an RNase of the Regnase-1 family able to degrade inflammatory transcripts. Our data reveal a molecular network regulating the proinflammatory phenotype of human memory T lymphocytes, with the potential to contribute to disease.


Subject(s)
Gene Expression Regulation/immunology , Immunologic Memory/immunology , Inflammation/immunology , Cell Line , Cell Line, Tumor , Cytokines/immunology , HEK293 Cells , Humans , Jurkat Cells , Lymphocyte Activation/immunology , NF-kappa B/immunology , Phenotype , T-Lymphocytes/immunology
4.
Mol Cell ; 84(5): 967-980.e10, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38242130

ABSTRACT

Histone-modifying enzymes depend on the availability of cofactors, with acetyl-coenzyme A (CoA) being required for histone acetyltransferase (HAT) activity. The discovery that mitochondrial acyl-CoA-producing enzymes translocate to the nucleus suggests that high concentrations of locally synthesized metabolites may impact acylation of histones and other nuclear substrates, thereby controlling gene expression. Here, we show that 2-ketoacid dehydrogenases are stably associated with the Mediator complex, thus providing a local supply of acetyl-CoA and increasing the generation of hyper-acetylated histone tails. Nitric oxide (NO), which is produced in large amounts in lipopolysaccharide-stimulated macrophages, inhibited the activity of Mediator-associated 2-ketoacid dehydrogenases. Elevation of NO levels and the disruption of Mediator complex integrity both affected de novo histone acetylation within a shared set of genomic regions. Our findings indicate that the local supply of acetyl-CoA generated by 2-ketoacid dehydrogenases bound to Mediator is required to maximize acetylation of histone tails at sites of elevated HAT activity.


Subject(s)
Histones , Nitric Oxide , Histones/genetics , Histones/metabolism , Acetyl Coenzyme A/metabolism , Acetylation , Nitric Oxide/metabolism , Mediator Complex/metabolism , Oxidoreductases/metabolism
5.
Nat Immunol ; 20(7): 783-792, 2019 07.
Article in English | MEDLINE | ID: mdl-31213714

ABSTRACT

Adaptation is the ability of cells, tissues and organisms to rapidly and reversibly modify their properties to maximize fitness in a changing environment. The activity of immune-system components unfolds in the remarkably heterogeneous milieus to which they are exposed in different tissues, during homeostasis or during various acute or chronic pathological states. Therefore, adaptation is essential for immune cells to tune their responses to a large variety of contexts and conditions. The adaptation of immune cells reflects the integration of multiple inputs acting simultaneously or in a temporal sequence, which eventually leads to transcriptional reprogramming and to various functional consequences, some of which extend beyond the duration of the stimulus. A range of adaptive responses have been observed in both adaptive immune cells and innate immune cells; these are referred to with terms such as 'plasticity', 'priming', 'training', 'exhaustion' and 'tolerance', among others, all of which can be useful for defining a certain immunological process or outcome but whose underlying molecular frameworks are often incompletely understood. Here we review and analyze mechanisms of adaptation and memory in immunity with the aim of providing basic concepts that rationalize the properties and molecular bases of these essential processes.


Subject(s)
Adaptation, Physiological , Immunity , Immunologic Memory , Adaptive Immunity , Animals , Gene Expression Regulation , Histones/metabolism , Humans , Hypersensitivity/immunology , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Immune Tolerance , Immunity, Innate , Organ Specificity/immunology , Phenotype , Signal Transduction
6.
Cell ; 165(1): 18-19, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-27015303

ABSTRACT

Genomic analyses are commonly used to infer trends and broad rules underlying transcriptional control. The innovative approach by Tong et al. to interrogate genomic datasets allows extracting mechanistic information on the specific regulation of individual genes.


Subject(s)
Beauty , Genome , Gene Expression Regulation , Genomics , Humans
7.
Genes Dev ; 37(21-24): 1017-1040, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38092518

ABSTRACT

Transcription termination pathways mitigate the detrimental consequences of unscheduled promiscuous initiation occurring at hundreds of thousands of genomic cis-regulatory elements. The Restrictor complex, composed of the Pol II-interacting protein WDR82 and the RNA-binding protein ZC3H4, suppresses processive transcription at thousands of extragenic sites in mammalian genomes. Restrictor-driven termination does not involve nascent RNA cleavage, and its interplay with other termination machineries is unclear. Here we show that efficient termination at Restrictor-controlled extragenic transcription units involves the recruitment of the protein phosphatase 1 (PP1) regulatory subunit PNUTS, a negative regulator of the SPT5 elongation factor, and Symplekin, a protein associated with RNA cleavage complexes but also involved in cleavage-independent and phosphatase-dependent termination of noncoding RNAs in yeast. PNUTS and Symplekin act synergistically with, but independently from, Restrictor to dampen processive extragenic transcription. Moreover, the presence of limiting nuclear levels of Symplekin imposes a competition for its recruitment among multiple transcription termination machineries, resulting in mutual regulatory interactions. Hence, by synergizing with Restrictor, Symplekin and PNUTS enable efficient termination of processive, long-range extragenic transcription.


Subject(s)
RNA Polymerase II , Transcription, Genetic , Animals , RNA Polymerase II/metabolism , Regulatory Sequences, Nucleic Acid , RNA-Binding Proteins/metabolism , Protein Processing, Post-Translational , Mammals/genetics
8.
Nat Immunol ; 19(9): 932-941, 2018 09.
Article in English | MEDLINE | ID: mdl-30127433

ABSTRACT

Cohesin is important for 3D genome organization. Nevertheless, even the complete removal of cohesin has surprisingly little impact on steady-state gene transcription and enhancer activity. Here we show that cohesin is required for the core transcriptional response of primary macrophages to microbial signals, and for inducible enhancer activity that underpins inflammatory gene expression. Consistent with a role for inflammatory signals in promoting myeloid differentiation of hematopoietic stem and progenitor cells (HPSCs), cohesin mutations in HSPCs led to reduced inflammatory gene expression and increased resistance to differentiation-inducing inflammatory stimuli. These findings uncover an unexpected dependence of inducible gene expression on cohesin, link cohesin with myeloid differentiation, and may help explain the prevalence of cohesin mutations in human acute myeloid leukemia.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Differentiation/genetics , Cell Self Renewal/genetics , Chromosomal Proteins, Non-Histone/metabolism , Hematopoietic Stem Cells/physiology , Leukemia, Myeloid, Acute/genetics , Macrophages/physiology , Nuclear Proteins/genetics , Phosphoproteins/genetics , Animals , Cell Cycle Proteins/genetics , Cells, Cultured , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans , Inflammation/genetics , Lipopolysaccharides/immunology , Mice , Mice, Knockout , Mutation/genetics , Cohesins
9.
Genes Dev ; 36(7-8): 414-432, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35361678

ABSTRACT

Six methyltransferases divide labor in establishing genomic profiles of histone H3 lysine 9 methylation (H3K9me), an epigenomic modification controlling constitutive heterochromatin, gene repression, and silencing of retroelements. Among them, SETDB1 is recruited to active chromatin domains to silence the expression of endogenous retroviruses. In the context of experiments aimed at determining the impact of SETDB1 on stimulus-inducible gene expression in macrophages, we found that loss of H3K9me3 caused by SETDB1 depletion was associated with increased recruitment of CTCF to >1600 DNA binding motifs contained within SINE B2 repeats, a previously unidentified target of SETDB1-mediated repression. CTCF is an essential regulator of chromatin folding that restrains DNA looping by cohesin, thus creating boundaries among adjacent topological domains. Increased CTCF binding to SINE B2 repeats enhanced insulation at hundreds of sites and increased loop formation within topological domains containing lipopolysaccharide-inducible genes, which correlated with their impaired regulation in response to stimulation. These data indicate a role of H3K9me3 in restraining genomic distribution and activity of CTCF, with an impact on chromatin organization and gene regulation.


Subject(s)
Chromatin , Gene Silencing , Heterochromatin , Methylation , Retroelements
10.
Nat Immunol ; 18(5): 530-540, 2017 05.
Article in English | MEDLINE | ID: mdl-28288101

ABSTRACT

Stimulation of macrophages with interferon-γ (IFN-γ) and interleukin 4 (IL-4) triggers distinct and opposing activation programs. During mixed infections or cancer, macrophages are often exposed to both cytokines, but how these two programs influence each other remains unclear. We found that IFN-γ and IL-4 mutually inhibited the epigenomic and transcriptional changes induced by each cytokine alone. Computational and functional analyses revealed the genomic bases for gene-specific cross-repression. For instance, while binding motifs for the transcription factors STAT1 and IRF1 were associated with robust and IL-4-resistant responses to IFN-γ, their coexistence with binding sites for auxiliary transcription factors such as AP-1 generated vulnerability to IL-4-mediated inhibition. These data provide a core mechanistic framework for the integration of signals that control macrophage activation in complex environmental conditions.


Subject(s)
Cell Differentiation , Epigenesis, Genetic , Macrophages/physiology , Proto-Oncogene Proteins c-myc/metabolism , Transcriptional Activation , Animals , Cell Line , Gene Expression Regulation , Humans , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Interferon-gamma/metabolism , Interleukin-4/metabolism , Mice , Mice, Inbred Strains , Proto-Oncogene Proteins c-myc/genetics , RNA, Small Interfering/genetics , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction , Transcription Factor AP-1/metabolism
11.
Nat Immunol ; 17(1): 26-33, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26681459

ABSTRACT

In tissues, macrophages are exposed to metabolic, homeostatic and immunoregulatory signals of local or systemic origin that influence their basal functions and responses to danger signals. Signal-transduction pathways regulated by extracellular signals are coupled to distinct sets of broadly expressed stimulus-regulated transcription factors whose ability to elicit gene-expression changes is influenced by the accessibility of their binding sites in the macrophage genome. In turn, accessibility of macrophage-specific transcriptional regulatory elements (enhancers and promoters) is specified by transcription factors that determine the macrophage lineage or impose their tissue-specific properties. Here we review recent findings that advance the understanding of mechanisms underlying priming and signal-dependent activation of macrophages and discuss the effect of genetic variation on these processes.


Subject(s)
Epigenesis, Genetic/immunology , Macrophage Activation/immunology , Macrophages , Animals , Gene Expression Regulation/immunology , Humans , Macrophages/cytology , Macrophages/immunology , Signal Transduction/immunology
12.
Cell ; 152(1-2): 157-71, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23332752

ABSTRACT

According to current models, once the cell has reached terminal differentiation, the enhancer repertoire is completely established and maintained by cooperatively acting lineage-specific transcription factors (TFs). TFs activated by extracellular stimuli operate within this predetermined repertoire, landing close to where master regulators are constitutively bound. Here, we describe latent enhancers, defined as regions of the genome that in terminally differentiated cells are unbound by TFs and lack the histone marks characteristic of enhancers but acquire these features in response to stimulation. Macrophage stimulation caused sequential binding of stimulus-activated and lineage-determining TFs to these regions, enabling deposition of enhancer marks. Once unveiled, many of these enhancers did not return to a latent state when stimulation ceased; instead, they persisted and mediated a faster and stronger response upon restimulation. We suggest that stimulus-specific expansion of the cis-regulatory repertoire provides an epigenomic memory of the exposure to environmental agents.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation , Macrophages/metabolism , Animals , Cell Differentiation , Epigenomics , Histone Code , Lipopolysaccharides/metabolism , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism
13.
Cell ; 153(1): 101-11, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23540693

ABSTRACT

LINE-1 (L1) retrotransposons are mobile genetic elements comprising ~17% of the human genome. New L1 insertions can profoundly alter gene function and cause disease, though their significance in cancer remains unclear. Here, we applied enhanced retrotransposon capture sequencing (RC-seq) to 19 hepatocellular carcinoma (HCC) genomes and elucidated two archetypal L1-mediated mechanisms enabling tumorigenesis. In the first example, 4/19 (21.1%) donors presented germline retrotransposition events in the tumor suppressor mutated in colorectal cancers (MCC). MCC expression was ablated in each case, enabling oncogenic ß-catenin/Wnt signaling. In the second example, suppression of tumorigenicity 18 (ST18) was activated by a tumor-specific L1 insertion. Experimental assays confirmed that the L1 interrupted a negative feedback loop by blocking ST18 repression of its enhancer. ST18 was also frequently amplified in HCC nodules from Mdr2(-/-) mice, supporting its assignment as a candidate liver oncogene. These proof-of-principle results substantiate L1-mediated retrotransposition as an important etiological factor in HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , DNA Mutational Analysis , Genes, Tumor Suppressor , Liver Neoplasms/genetics , Long Interspersed Nucleotide Elements , Mutagenesis, Insertional , ATP Binding Cassette Transporter, Subfamily B/genetics , Adult , Aged , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Female , Humans , Male , Mice , Middle Aged , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , ATP-Binding Cassette Sub-Family B Member 4
14.
Cell ; 150(2): 279-90, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22817891

ABSTRACT

Macrophages respond to inflammatory stimuli by modulating the expression of hundreds of genes in a defined temporal cascade, with diverse transcriptional and posttranscriptional mechanisms contributing to the regulatory network. We examined proinflammatory gene regulation in activated macrophages by performing RNA-seq with fractionated chromatin-associated, nucleoplasmic, and cytoplasmic transcripts. This methodological approach allowed us to separate the synthesis of nascent transcripts from transcript processing and the accumulation of mature mRNAs. In addition to documenting the subcellular locations of coding and noncoding transcripts, the results provide a high-resolution view of the relationship between defined promoter and chromatin properties and the temporal regulation of diverse classes of coexpressed genes. The data also reveal a striking accumulation of full-length yet incompletely spliced transcripts in the chromatin fraction, suggesting that splicing often occurs after transcription has been completed, with transcripts retained on the chromatin until fully spliced.


Subject(s)
Chromatin/genetics , Gene Expression Profiling , Inflammation/genetics , Macrophages/metabolism , RNA Splicing , Animals , Gene Expression Regulation , Lipid A/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Receptor, Interferon alpha-beta/genetics , Receptors, Interferon/genetics , Sequence Analysis, RNA , Transcription, Genetic
15.
Genes Dev ; 33(17-18): 1159-1174, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31371436

ABSTRACT

Accessibility of the genomic regulatory information is largely controlled by the nucleosome-organizing activity of transcription factors (TFs). While stimulus-induced TFs bind to genomic regions that are maintained accessible by lineage-determining TFs, they also increase accessibility of thousands of cis-regulatory elements. Nucleosome remodeling events underlying such changes and their interplay with basal positioning are unknown. Here, we devised a novel quantitative framework discriminating different types of nucleosome remodeling events in micrococcal nuclease ChIP-seq (chromatin immunoprecipitation [ChIP] combined with high-throughput sequencing) data sets and used it to analyze nucleosome dynamics at stimulus-regulated cis-regulatory elements. At enhancers, remodeling preferentially affected poorly positioned nucleosomes while sparing well-positioned nucleosomes flanking the enhancer core, indicating that inducible TFs do not suffice to overrule basal nucleosomal organization maintained by lineage-determining TFs. Remodeling events appeared to be combinatorially driven by multiple TFs, with distinct TFs showing, however, different remodeling efficiencies. Overall, these data provide a systematic view of the impact of stimulation on nucleosome organization and genome accessibility in mammalian cells.


Subject(s)
Nucleosomes/metabolism , Regulatory Elements, Transcriptional/physiology , Transcription Factors/metabolism , Animals , Cells, Cultured , Chromatin Immunoprecipitation , High-Throughput Nucleotide Sequencing , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Micrococcal Nuclease/metabolism
16.
Immunity ; 46(5): 764-766, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28514680

ABSTRACT

Mouse blood monocytes include two main subsets usually discriminated by the expression of the Ly6C surface marker. The study by Mildner et al. (2017) in this issue of Immunity clarifies the transcriptional circuits controlling the generation of Ly6C- cells from their obligate precursors, the Ly6C+ monocytes.


Subject(s)
Antigens, Ly , Monocytes/immunology , Animals , Mice , Mice, Inbred C57BL
17.
EMBO J ; 40(13): e107206, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33844319

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC), one of the most highly lethal tumors, is characterized by complex histology, with a massive fibrotic stroma in which both pseudo-glandular structures and compact nests of abnormally differentiated tumor cells are embedded, in different proportions and with different mutual relationships in space. This complexity and the heterogeneity of the tumor component have hindered the development of a broadly accepted, clinically actionable classification of PDACs, either on a morphological or a molecular basis. Here, we discuss evidence suggesting that such heterogeneity can to a large extent, albeit not exclusively, be traced back to two main classes of PDAC cells that commonly coexist in the same tumor: cells that maintained their ability to differentiate toward endodermal, mucin-producing epithelia and epithelial cells unable to form glandular structures and instead characterized by various levels of squamous differentiation and the expression of mesenchymal lineage genes. The underlying gene regulatory networks and how they are controlled by distinct transcription factors, as well as the practical implications of these two different populations of tumor cells, are discussed.


Subject(s)
Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Transcription, Genetic/genetics , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Differentiation/genetics , Epithelial Cells/pathology , Epithelium/pathology , Gene Expression Regulation, Neoplastic/genetics , Gene Regulatory Networks/genetics , Humans , Transcription Factors/genetics
18.
Gastroenterology ; 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38971196

ABSTRACT

BACKGROUND AND AIMS: WNT signaling is central to spatial tissue arrangement, regulating stem cell activity, and represents the hallmark of gastrointestinal cancers. While its role in driving intestinal tumors is well characterized, WNT's role in gastric tumorigenesis remains elusive. METHODS: We have developed mouse models to control the specific expression of an oncogenic form of B-CATENIN in combination with MYC activation in Lgr5+ cells of the gastric antrum. We used multi-omics approaches applied in vivo and in organoid models to characterize their cooperation in driving gastric tumorigenesis. RESULTS: We report that constitutive B-CATENIN stabilization in the stomach has negligible oncogenic effects and requires MYC activation to induce gastric tumour formation. While physiologically low MYC levels in gastric glands limit B-CATENIN transcriptional activity, increased MYC expression unleashes the WNT oncogenic transcriptional program, promoting B-CATENIN enhancer invasion without a direct transcriptional cooperation. MYC activation induces a metabolic rewiring that suppresses lysosomal biogenesis through mTOR and ERK activation and MiT/TFE inhibition. This prevents EPCAM degradation by macropinocytosis, promoting B-CATENIN chromatin accumulation and activation of WNT oncogenic transcription. CONCLUSION: Our results uncovered a new signaling framework with important implications for the control of gastric epithelial architecture and WNT-dependent oncogenic transformation.

19.
Mol Syst Biol ; 20(6): 626-650, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38724853

ABSTRACT

More than 500 kinases are implicated in the control of most cellular process in mammals, and deregulation of their activity is linked to cancer and inflammatory disorders. 80 clinical kinase inhibitors (CKIs) have been approved for clinical use and hundreds are in various stages of development. However, CKIs inhibit other kinases in addition to the intended target(s), causing both enhanced clinical effects and undesired side effects that are only partially predictable based on in vitro selectivity profiling. Here, we report an integrative approach grounded on the use of chromatin modifications as unbiased, information-rich readouts of the functional effects of CKIs on macrophage activation. This approach exceeded the performance of transcriptome-based approaches and allowed us to identify similarities and differences among CKIs with identical intended targets, to recognize novel CKI specificities and to pinpoint CKIs that may be repurposed to control inflammation, thus supporting the utility of this strategy to improve selection and use of CKIs in clinical settings.


Subject(s)
Epigenome , Protein Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Humans , Animals , Mice , Macrophage Activation/drug effects , Macrophage Activation/genetics , Macrophages/drug effects , Macrophages/metabolism
20.
Nat Immunol ; 14(8): 777-84, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23867934

ABSTRACT

Standard definitions of immunological memory are all built on the idea that once infected, animals are protected more efficiently against a second infection. This common view overlooks an unavoidable consequence of the exposure of cells to pathogens, danger signals and environmental agents in general: stimuli change cell properties and activity in a transient yet sustained manner that extends beyond the exposure time and modulates the response of cells of both the innate and adaptive immune systems to secondary stimulation. We suggest that this transient phenomenon represents 'short-term memory' of environmental exposure and discuss the evidence that this is mediated by the persistence of long-lived regulatory molecules, notably a subset of newly deposited chromatin modifications and inducible noncoding RNAs.


Subject(s)
Chromatin/immunology , Environmental Exposure , Immunologic Memory/immunology , Memory, Short-Term/physiology , MicroRNAs/immunology , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Chromatin/genetics , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Immunologic Memory/genetics , MicroRNAs/genetics
SELECTION OF CITATIONS
SEARCH DETAIL