Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 133
Filter
Add more filters

Publication year range
1.
Cell ; 173(1): 260-274.e25, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29551266

ABSTRACT

Protein degradation plays important roles in biological processes and is tightly regulated. Further, targeted proteolysis is an emerging research tool and therapeutic strategy. However, proteome-wide technologies to investigate the causes and consequences of protein degradation in biological systems are lacking. We developed "multiplexed proteome dynamics profiling" (mPDP), a mass-spectrometry-based approach combining dynamic-SILAC labeling with isobaric mass tagging for multiplexed analysis of protein degradation and synthesis. In three proof-of-concept studies, we uncover different responses induced by the bromodomain inhibitor JQ1 versus a JQ1 proteolysis targeting chimera; we elucidate distinct modes of action of estrogen receptor modulators; and we comprehensively classify HSP90 clients based on their requirement for HSP90 constitutively or during synthesis, demonstrating that constitutive HSP90 clients have lower thermal stability than non-clients, have higher affinity for the chaperone, vary between cell types, and change upon external stimuli. These findings highlight the potential of mPDP to identify dynamically controlled degradation mechanisms in cellular systems.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Proteome/analysis , Proteomics/methods , Azepines/chemistry , Azepines/metabolism , Azepines/pharmacology , Cell Line , Chromatography, High Pressure Liquid , Cluster Analysis , Estradiol/pharmacology , Humans , Isotope Labeling , Jurkat Cells , MCF-7 Cells , Neoplasm Proteins/metabolism , Proteins/antagonists & inhibitors , Proteins/metabolism , Proteolysis/drug effects , Receptors, Estrogen/metabolism , Tandem Mass Spectrometry , Triazoles/chemistry , Triazoles/metabolism , Triazoles/pharmacology
2.
Cell ; 158(5): 989-999, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25131989

ABSTRACT

Latent reservoirs of HIV-1-infected cells are refractory to antiretroviral therapies (ART) and remain the major barrier to curing HIV-1. Because latently infected cells are long-lived, immunologically invisible, and may undergo homeostatic proliferation, a "shock and kill" approach has been proposed to eradicate this reservoir by combining ART with inducers of viral transcription. However, all attempts to alter the HIV-1 reservoir in vivo have failed to date. Using humanized mice, we show that broadly neutralizing antibodies (bNAbs) can interfere with establishment of a silent reservoir by Fc-FcR-mediated mechanisms. In established infection, bNAbs or bNAbs plus single inducers are ineffective in preventing viral rebound. However, bNAbs plus a combination of inducers that act by independent mechanisms synergize to decrease the reservoir as measured by viral rebound. Thus, combinations of inducers and bNAbs constitute a therapeutic strategy that impacts the establishment and maintenance of the HIV-1 reservoir in humanized mice.


Subject(s)
Antibodies, Neutralizing/administration & dosage , HIV Infections/immunology , HIV-1/drug effects , Transcription, Genetic/drug effects , Virus Latency/drug effects , Animals , Anti-HIV Agents/therapeutic use , Antibodies, Neutralizing/immunology , CD4-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/administration & dosage , HIV Infections/virology , HIV-1/genetics , HIV-1/physiology , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Humans , Hydroxamic Acids/administration & dosage , Immunoglobulin Fc Fragments/immunology , Mice , Receptors, Fc/immunology , Vorinostat
3.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Article in English | MEDLINE | ID: mdl-33619107

ABSTRACT

Reactivation of human cytomegalovirus (HCMV) from latency is a major health consideration for recipients of stem-cell and solid organ transplantations. With over 200,000 transplants taking place globally per annum, virus reactivation can occur in more than 50% of cases leading to loss of grafts as well as serious morbidity and even mortality. Here, we present the most extensive screening to date of epigenetic inhibitors on HCMV latently infected cells and find that histone deacetylase inhibitors (HDACis) and bromodomain inhibitors are broadly effective at inducing virus immediate early gene expression. However, while HDACis, such as myeloid-selective CHR-4487, lead to production of infectious virions, inhibitors of bromodomain (BRD) and extraterminal proteins (I-BETs), including GSK726, restrict full reactivation. Mechanistically, we show that BET proteins (BRDs) are pivotally connected to regulation of HCMV latency and reactivation. Through BRD4 interaction, the transcriptional activator complex P-TEFb (CDK9/CycT1) is sequestered by repressive complexes during HCMV latency. Consequently, I-BETs allow release of P-TEFb and subsequent recruitment to promoters via the superelongation complex (SEC), inducing transcription of HCMV lytic genes encoding immunogenic antigens from otherwise latently infected cells. Surprisingly, this occurs without inducing many viral immunoevasins and, importantly, while also restricting viral DNA replication and full HCMV reactivation. Therefore, this pattern of HCMV transcriptional dysregulation allows effective cytotoxic immune targeting and killing of latently infected cells, thus reducing the latent virus genome load. This approach could be safely used to pre-emptively purge the virus latent reservoir prior to transplantation, thereby reducing HCMV reactivation-related morbidity and mortality.


Subject(s)
Cell Cycle Proteins/genetics , Cytomegalovirus/immunology , DNA, Viral/genetics , Epigenesis, Genetic , Histone Deacetylases/genetics , Positive Transcriptional Elongation Factor B/genetics , Transcription Factors/genetics , Azepines/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Benzodiazepines/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/immunology , Cyclin T/genetics , Cyclin T/immunology , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/immunology , Cytomegalovirus/drug effects , Cytomegalovirus/genetics , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , DNA Replication/drug effects , DNA, Viral/antagonists & inhibitors , DNA, Viral/immunology , Genes, Immediate-Early , Genes, Reporter , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/immunology , Host-Pathogen Interactions , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Models, Biological , Positive Transcriptional Elongation Factor B/immunology , Primary Cell Culture , Promoter Regions, Genetic , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology , THP-1 Cells , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Transcription Factors/antagonists & inhibitors , Transcription Factors/immunology , Transcription, Genetic , Virus Activation/drug effects , Virus Latency/drug effects
4.
Curr Issues Mol Biol ; 45(5): 4228-4245, 2023 May 11.
Article in English | MEDLINE | ID: mdl-37232738

ABSTRACT

SP140 is an epigenetic reader protein expressed predominantly in immune cells. GWAS studies have shown an association between SP140 single nucleotide polymorphisms (SNPs) and diverse autoimmune and inflammatory diseases, suggesting a possible pathogenic role for SP140 in immune-mediated diseases. We previously demonstrated that treatment of human macrophages with the novel selective inhibitor of the SP140 protein (GSK761) reduced the expression of endotoxin-induced cytokines, implicating a role of SP140 in the function of inflammatory macrophages. In this study, we investigated the effects of GSK761 on in vitro human dendritic cell (DC) differentiation and maturation, assessing the expression of cytokines and co-stimulatory molecules and their capacity to stimulate T-cell activation and induce phenotypic changes. In DCs, lipopolysaccharide (LPS) stimulation induced an increase in SP140 expression and its recruitment to transcription start sites (TSS) of pro-inflammatory cytokine genes. Moreover, LPS-induced cytokines such as TNF, IL-6, and IL-1ß were reduced in GSK761- or SP140 siRNA- treated DCs. Although GSK761 did not significantly affect the expression of surface markers that define the differentiation of CD14+ monocytes into immature DCs (iDCs), subsequent maturation of iDCs to mature DCs was significantly inhibited. GSK761 strongly reduced expression of the maturation marker CD83, the co-stimulatory molecules CD80 and CD86, and the lipid-antigen presentation molecule CD1b. Finally, when the ability of DCs to stimulate recall T-cell responses by vaccine-specific T cells was assessed, T cells stimulated by GSK761-treated DCs showed reduced TBX21 and RORA expression and increased FOXP3 expression, indicating a preferential generation of regulatory T cells. Overall, this study suggests that SP140 inhibition enhances the tolerogenic properties of DCs, supporting the rationale of targeting SP140 in autoimmune and inflammatory diseases where DC-mediated inflammatory responses contribute to disease pathogenesis.

5.
Br J Cancer ; 129(1): 163-174, 2023 07.
Article in English | MEDLINE | ID: mdl-37120667

ABSTRACT

BACKGROUND: Epigenomic dysregulation has been linked to solid tumour malignancies, including ovarian cancers. Profiling of re-programmed enhancer locations associated with disease has the potential to improve stratification and thus therapeutic choices. Ovarian cancers are subdivided into histological subtypes that have significant molecular and clinical differences, with high-grade serous carcinoma representing the most common and aggressive subtype. METHODS: We interrogated the enhancer landscape(s) of normal ovary and subtype-specific ovarian cancer states using publicly available data. With an initial focus on H3K27ac histone mark, we developed a computational pipeline to predict drug compound activity based on epigenomic stratification. Lastly, we substantiated our predictions in vitro using patient-derived clinical samples and cell lines. RESULTS: Using our in silico approach, we highlighted recurrent and privative enhancer landscapes and identified the differential enrichment of a total of 164 transcription factors involved in 201 protein complexes across the subtypes. We pinpointed SNS-032 and EHMT2 inhibitors BIX-01294 and UNC0646 as therapeutic candidates in high-grade serous carcinoma, as well as probed the efficacy of specific inhibitors in vitro. CONCLUSION: Here, we report the first attempt to exploit ovarian cancer epigenomic landscapes for drug discovery. This computational pipeline holds enormous potential for translating epigenomic profiling into therapeutic leads.


Subject(s)
Carcinoma , Cystadenocarcinoma, Serous , Ovarian Neoplasms , Humans , Female , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/metabolism , Histocompatibility Antigens/therapeutic use , Histone-Lysine N-Methyltransferase
6.
Nat Immunol ; 12(1): 29-36, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21131967

ABSTRACT

Signaling via the methylation of lysine residues in proteins has been linked to diverse biological and disease processes, yet the catalytic activity and substrate specificity of many human protein lysine methyltransferases (PKMTs) are unknown. We screened over 40 candidate PKMTs and identified SETD6 as a methyltransferase that monomethylated chromatin-associated transcription factor NF-κB subunit RelA at Lys310 (RelAK310me1). SETD6-mediated methylation rendered RelA inert and attenuated RelA-driven transcriptional programs, including inflammatory responses in primary immune cells. RelAK310me1 was recognized by the ankryin repeat of the histone methyltransferase GLP, which under basal conditions promoted a repressed chromatin state at RelA target genes through GLP-mediated methylation of histone H3 Lys9 (H3K9). NF-κB-activation-linked phosphorylation of RelA at Ser311 by protein kinase C-ζ (PKC-ζ) blocked the binding of GLP to RelAK310me1 and relieved repression of the target gene. Our findings establish a previously uncharacterized mechanism by which chromatin signaling regulates inflammation programs.


Subject(s)
Arthritis, Rheumatoid/immunology , NF-kappa B/metabolism , Protein Methyltransferases/metabolism , Transcription Factor RelA/metabolism , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Chromatin Assembly and Disassembly/genetics , DNA Methylation , HEK293 Cells , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Inflammation , Lysine/metabolism , NF-kappa B/genetics , NF-kappa B/immunology , Protein Binding/genetics , Protein Methyltransferases/genetics , Protein Methyltransferases/immunology , RNA, Small Interfering/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology
7.
Trends Immunol ; 41(1): 29-45, 2020 01.
Article in English | MEDLINE | ID: mdl-31813765

ABSTRACT

Memory T cells possess functional differences from naïve T cells that powerfully contribute to the efficiency of secondary immune responses. These abilities are imprinted during the primary response, linked to the acquisition of novel patterns of gene expression. Underlying this are alterations at the chromatin level (epigenetic modifications) that regulate constitutive and inducible gene transcription. T cell epigenetic memory can persist long-term, contributing to long-lasting immunity after infection or vaccination. However, acquired epigenetic states can also hinder effective tumor immunity or contribute to autoimmunity. The growing understanding of epigenetic gene regulation as it relates to both the stability and malleability of T cell memory may offer the potential to selectively modify T cell memory in disease by targeting epigenetic mechanisms.


Subject(s)
Epigenesis, Genetic , Immunologic Memory , T-Lymphocytes , Chromatin/immunology , Epigenesis, Genetic/immunology , Gene Expression Regulation , Humans , Immunologic Memory/genetics , T-Lymphocytes/immunology
8.
J Immunol ; 207(2): 555-568, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34233910

ABSTRACT

As key cells of the immune system, macrophages coordinate the activation and regulation of the immune response. Macrophages present a complex phenotype that can vary from homeostatic, proinflammatory, and profibrotic to anti-inflammatory phenotypes. The factors that drive the differentiation from monocyte to macrophage largely define the resultant phenotype, as has been shown by the differences found in M-CSF- and GM-CSF-derived macrophages. We explored alternative inflammatory mediators that could be used for in vitro differentiation of human monocytes into macrophages. IFN-γ is a potent inflammatory mediator produced by lymphocytes in disease and infections. We used IFN-γ to differentiate human monocytes into macrophages and characterized the cells at a functional and proteomic level. IFN-γ alone was sufficient to generate macrophages (IFN-γ Mϕ) that were phagocytic and responsive to polarization. We demonstrate that IFN-γ Mϕ are potent activators of T lymphocytes that produce IL-17 and IFN-γ. We identified potential markers (GBP-1, IP-10, IL-12p70, and IL-23) of IFN-γ Mϕ and demonstrate that these markers are enriched in the skin of patients with inflamed psoriasis. Collectively, we show that IFN-γ can drive human monocyte to macrophage differentiation, leading to bona fide macrophages with inflammatory characteristics.


Subject(s)
Cell Differentiation/physiology , Inflammation/metabolism , Interferon-gamma/metabolism , Macrophages/metabolism , Monocytes/metabolism , Psoriasis/metabolism , Biomarkers/metabolism , Cells, Cultured , Humans , Macrophage Colony-Stimulating Factor/metabolism , Phenotype , Proteomics/methods , Skin/metabolism
9.
Proc Natl Acad Sci U S A ; 117(11): 6056-6066, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32123118

ABSTRACT

T helper (Th) cells are CD4+ effector T cells that play a critical role in immunity by shaping the inflammatory cytokine environment in a variety of physiological and pathological situations. Using a combined chemico-genetic approach, we identify histone H3K27 demethylases KDM6A and KDM6B as central regulators of human Th subsets. The prototypic KDM6 inhibitor GSK-J4 increases genome-wide levels of the repressive H3K27me3 chromatin mark and leads to suppression of the key transcription factor RORγt during Th17 differentiation. In mature Th17 cells, GSK-J4 induces an altered transcriptional program with a profound metabolic reprogramming and concomitant suppression of IL-17 cytokine levels and reduced proliferation. Single-cell analysis reveals a specific shift from highly inflammatory cell subsets toward a resting state upon demethylase inhibition. The root cause of the observed antiinflammatory phenotype in stimulated Th17 cells is reduced expression of key metabolic transcription factors, such as PPRC1. Overall, this leads to reduced mitochondrial biogenesis, resulting in a metabolic switch with concomitant antiinflammatory effects. These data are consistent with an effect of GSK-J4 on Th17 T cell differentiation pathways directly related to proliferation and include regulation of effector cytokine profiles. This suggests that inhibiting KDM6 demethylases may be an effective, even in the short term, therapeutic target for autoimmune diseases, including ankylosing spondylitis.


Subject(s)
Benzazepines/pharmacology , Histone Demethylases/metabolism , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Pyrimidines/pharmacology , Th17 Cells/metabolism , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Benzazepines/therapeutic use , Cell Differentiation/drug effects , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/immunology , Histone Code/drug effects , Histone Demethylases/antagonists & inhibitors , Humans , Interleukin-17/metabolism , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Primary Cell Culture , Pyrimidines/therapeutic use , RNA-Seq , Spondylitis, Ankylosing/drug therapy , Spondylitis, Ankylosing/immunology , Th17 Cells/drug effects , Th17 Cells/immunology , Transcription Factors/metabolism
10.
BMC Biol ; 20(1): 182, 2022 08 19.
Article in English | MEDLINE | ID: mdl-35986286

ABSTRACT

BACKGROUND: SP140 is a bromodomain-containing protein expressed predominantly in immune cells. Genetic polymorphisms and epigenetic modifications in the SP140 locus have been linked to Crohn's disease (CD), suggesting a role in inflammation. RESULTS: We report the development of the first small molecule SP140 inhibitor (GSK761) and utilize this to elucidate SP140 function in macrophages. We show that SP140 is highly expressed in CD mucosal macrophages and in in vitro-generated inflammatory macrophages. SP140 inhibition through GSK761 reduced monocyte-to-inflammatory macrophage differentiation and lipopolysaccharide (LPS)-induced inflammatory activation, while inducing the generation of CD206+ regulatory macrophages that were shown to associate with a therapeutic response to anti-TNF in CD patients. SP140 preferentially occupies transcriptional start sites in inflammatory macrophages, with enrichment at gene loci encoding pro-inflammatory cytokines/chemokines and inflammatory pathways. GSK761 specifically reduces SP140 chromatin binding and thereby expression of SP140-regulated genes. GSK761 inhibits the expression of cytokines, including TNF, by CD14+ macrophages isolated from CD intestinal mucosa. CONCLUSIONS: This study identifies SP140 as a druggable epigenetic therapeutic target for CD.


Subject(s)
Crohn Disease , Tumor Necrosis Factor Inhibitors , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Crohn Disease/genetics , Crohn Disease/metabolism , Cytokines/genetics , Cytokines/metabolism , Epigenesis, Genetic , Humans , Macrophages , Transcription Factors/genetics
11.
Int J Cancer ; 151(2): 275-286, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35239184

ABSTRACT

Radiotherapy can induce various adverse effects including fibrosis in cancer patients. Radiation-induced aberrant expression of profibrotic genes has been associated with dysregulated epigenetic mechanisms. Pan-BET (bromodomain and extraterminal domain) inhibitors, such as JQ1 and I-BET151, have been reported to attenuate the profibrotic response after irradiation. Despite their profound preclinical efficacy, the clinical utility of pan-inhibitors is limited due to observed cytotoxicicities. Recently, inhibitors were developed that selectively target the first (BD1) and second (BD2) bromodomain of the BET proteins (iBET-BD1 [GSK778] and iBET-BD2 [GSK046]). Here, their potential to attenuate radiation-induced fibroblast activation with low-toxicity was investigated. Our results indicated that cell proliferation and cell cycle progression in fibroblasts from BJ cells and six donors were reduced when treated with I-BET151 and iBET-BD1, but not with iBET-BD2. After irradiation, induction of DGKA and profibrotic markers, especially COL1A1 and ACTA2, was attenuated with all BET inhibitors. H3K27ac enrichment was similar at the DGKA enhancer region after I-BET151 treatment and irradiation, but was reduced at the COL1A1 transcription start site and the ACTA2 enhancer site. iBET-BD2 did not change H3K27ac levels in these regions. BRD4 occupancy at these regions was not altered by any of the compounds. Cell migration activity was measured as a characteristic independent of extracellular matrix production and was unchanged in fibroblasts after irradiation and BET inhibitor-treatment. In conclusion, iBET-BD2 efficiently suppressed radiation-induced expression of DGKA and profibrotic markers without showing cytotoxicity. Thus BD2-selective targeting is a promising new therapeutic avenue for further investigations to prevent or attenuate radiotherapy-induced fibrosis.


Subject(s)
Antineoplastic Agents , Nuclear Proteins , Antineoplastic Agents/pharmacology , Cell Cycle Proteins/metabolism , Fibroblasts/metabolism , Fibrosis , Humans , Nuclear Proteins/metabolism , Protein Domains , Transcription Factors/metabolism
12.
Br J Clin Pharmacol ; 88(12): 5238-5256, 2022 12.
Article in English | MEDLINE | ID: mdl-35655123

ABSTRACT

AIMS: To improve the tolerability and therapeutic application of histone deacetylase inhibitors (HDACi), by application of an esterase-sensitive motif (ESM), to target pharmacological activity directly to mononuclear myeloid cells expressing the processing enzyme carboxylesterase-1 (CES1). METHODS: This first-in-human study comprised single and multiple ascending dose cohorts to determine safety and tolerability. Pharmacodynamic parameters included acetylation, cytokine inhibition and intracellular concentrations of processed acid metabolite in isolated monocytes. Mechanistic work was conducted in vitro and in a CES1/Es1elo mouse strain. RESULTS: ESM-HDAC391 showed transient systemic exposure (plasma half-life of 21-30 min) but selective retention of processed acid for at least 12 hours, resulting in robust targeted mechanistic engagement (increased acetylation in monocytes plus inhibition of ex vivo stimulated cytokine production). ESM-HDAC391 was well tolerated and clinical toxicities common to non-targeted HDACi were not observed. ESM-HDAC391 treatment was accompanied by the novel finding of a dose-dependent monocyte depletion that was transient and reversible and which plateaued at 0.06 × 109 monocytes/L after repeat dosing with 20 or 40 mg. Characterisation of monocyte depletion in transgenic mice (CES1/Es1elo ) suggested that colony stimulating factor 1 receptor loss on circulating cells contributed to ESM-HDAC-mediated depletion. Further mechanistic investigations using human monocytes in vitro demonstrated HDACi-mediated change in myeloid fate through modulation of colony stimulating factor 1 receptor and downstream effects on cell differentiation. CONCLUSION: These findings demonstrate selective targeting of monocytes in humans using the ESM approach and identify monocytopaenia as a novel outcome of ESM-HDACi treatment, with implications for potential benefit of these molecules in myeloid-driven diseases.


Subject(s)
Esterases , Histone Deacetylase Inhibitors , Humans , Animals , Mice , Histone Deacetylase Inhibitors/pharmacology , Macrophage Colony-Stimulating Factor , Cytokines
13.
Nature ; 525(7570): 538-42, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26367796

ABSTRACT

Bromodomain and extra terminal protein (BET) inhibitors are first-in-class targeted therapies that deliver a new therapeutic opportunity by directly targeting bromodomain proteins that bind acetylated chromatin marks. Early clinical trials have shown promise, especially in acute myeloid leukaemia, and therefore the evaluation of resistance mechanisms is crucial to optimize the clinical efficacy of these drugs. Here we use primary mouse haematopoietic stem and progenitor cells immortalized with the fusion protein MLL-AF9 to generate several single-cell clones that demonstrate resistance, in vitro and in vivo, to the prototypical BET inhibitor, I-BET. Resistance to I-BET confers cross-resistance to chemically distinct BET inhibitors such as JQ1, as well as resistance to genetic knockdown of BET proteins. Resistance is not mediated through increased drug efflux or metabolism, but is shown to emerge from leukaemia stem cells both ex vivo and in vivo. Chromatin-bound BRD4 is globally reduced in resistant cells, whereas the expression of key target genes such as Myc remains unaltered, highlighting the existence of alternative mechanisms to regulate transcription. We demonstrate that resistance to BET inhibitors, in human and mouse leukaemia cells, is in part a consequence of increased Wnt/ß-catenin signalling, and negative regulation of this pathway results in restoration of sensitivity to I-BET in vitro and in vivo. Together, these findings provide new insights into the biology of acute myeloid leukaemia, highlight potential therapeutic limitations of BET inhibitors, and identify strategies that may enhance the clinical utility of these unique targeted therapies.


Subject(s)
Benzodiazepines/pharmacology , Drug Resistance, Neoplasm/drug effects , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Nuclear Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Animals , Azepines/pharmacology , Cell Cycle Proteins , Cell Line, Tumor , Cells, Cultured , Chromatin/metabolism , Clone Cells/drug effects , Clone Cells/metabolism , Clone Cells/pathology , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/drug effects , Genes, myc/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Molecular Targeted Therapy , Neoplastic Stem Cells/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Triazoles/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
14.
Mol Cell ; 51(3): 310-25, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23932714

ABSTRACT

Recent studies suggest a hierarchical model in which lineage-determining factors act in a collaborative manner to select and prime cell-specific enhancers, thereby enabling signal-dependent transcription factors to bind and function in a cell-type-specific manner. Consistent with this model, TLR4 signaling primarily regulates macrophage gene expression through a pre-existing enhancer landscape. However, TLR4 signaling also induces priming of ∼3,000 enhancer-like regions de novo, enabling visualization of intermediates in enhancer selection and activation. Unexpectedly, we find that enhancer transcription precedes local mono- and dimethylation of histone H3 lysine 4 (H3K4me1/2). H3K4 methylation at de novo enhancers is primarily dependent on the histone methyltransferases Mll1, Mll2/4, and Mll3 and is significantly reduced by inhibition of RNA polymerase II elongation. Collectively, these findings suggest an essential role of enhancer transcription in H3K4me1/2 deposition at de novo enhancers that is independent of potential functions of the resulting eRNA transcripts.


Subject(s)
Enhancer Elements, Genetic , Macrophage Activation/genetics , Toll-Like Receptor 4/metabolism , Animals , Base Sequence , CCAAT-Enhancer-Binding Proteins/metabolism , Cells, Cultured , DNA Methylation , Gene Expression , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Myeloid-Lymphoid Leukemia Protein/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins/metabolism , RNA Polymerase II/antagonists & inhibitors , Sequence Analysis, DNA , Signal Transduction , Trans-Activators/metabolism , Transcription Factor RelA/metabolism , Transcription, Genetic
15.
Genes Dev ; 27(16): 1731-8, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23964091

ABSTRACT

Recent advances in the enzymology of transcription and chromatin regulation have led to the discovery of proteins that play a prominent role in cell differentiation and the maintenance of specialized cell functions. Knowledge about post-synthetic DNA and histone modifications as well as information about the rules that guide the formation of multimolecular chromatin-bound complexes have helped to delineate gene-regulating pathways and describe how these pathways are altered in various pathological conditions. The present review focuses on the emerging area of therapeutic interference with chromatin function for the purpose of cancer treatment and immunomodulation.


Subject(s)
Antineoplastic Agents/therapeutic use , Chromatin/drug effects , Immunity , Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Humans , Immunity/drug effects , Immunity/genetics , Immunomodulation/drug effects , Polycomb Repressive Complex 2/metabolism , Transcription Factors/metabolism
16.
Eur J Immunol ; 49(11): 2044-2050, 2019 11.
Article in English | MEDLINE | ID: mdl-31206650

ABSTRACT

Changes in the epigenetic landscape of immune cells are a crucial component of gene activation during the induction of inflammatory responses, therefore it has been hypothesized that epigenetic modulation could be employed to restore homeostasis in inflammatory scenarios. Fungal pathogens cause a large burden of morbidity and even mortality due to the hyperinflammatory processes that induce mucosal, allergic or systemic infections. Bromodomain and extraterminal domain (BET) proteins are considered as one as the most tantalizing pharmacological targets for the modulation of inflammatory responses at the epigenetic level. Nothing is known of the role of BET inhibitors on the inflammation induced by fungal pathogens. In the present study, we assessed the in vitro efficacy of the small molecular histone mimic BET inhibitor I-BET151 to modulate innate immune responses during fungal-immune interaction with the clinically relevant fungal pathogens Candida albicans and Aspergillus fumigatus. Our results prove that BET inhibitors (I-BETs) represent an important modulator of inflammation induced by fungal pathogens: both direct production of proinflammatory cytokines and the induction of trained immunity were inhibited by I-BET151. These modulatory effects are likely to have important potential implications in clinically relevant situations.


Subject(s)
Gene Expression Regulation/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Host-Pathogen Interactions/drug effects , Immunologic Factors/pharmacology , Monocytes/drug effects , Neutrophils/drug effects , Aspergillus fumigatus/immunology , Aspergillus fumigatus/pathogenicity , Candida albicans/immunology , Candida albicans/pathogenicity , Endocytosis/drug effects , Endocytosis/genetics , Endocytosis/immunology , Gene Expression Regulation/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Interleukin-6/immunology , Interleukins/genetics , Interleukins/immunology , Monocytes/immunology , Monocytes/microbiology , Neutrophils/immunology , Neutrophils/microbiology , Primary Cell Culture , Signal Transduction , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Interleukin-22
17.
Nature ; 514(7523): 513-7, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25132549

ABSTRACT

T-cell acute lymphoblastic leukaemia (T-ALL) is a haematological malignancy with a dismal overall prognosis, including a relapse rate of up to 25%, mainly because of the lack of non-cytotoxic targeted therapy options. Drugs that target the function of key epigenetic factors have been approved in the context of haematopoietic disorders, and mutations that affect chromatin modulators in a variety of leukaemias have recently been identified; however, 'epigenetic' drugs are not currently used for T-ALL treatment. Recently, we described that the polycomb repressive complex 2 (PRC2) has a tumour-suppressor role in T-ALL. Here we delineated the role of the histone 3 lysine 27 (H3K27) demethylases JMJD3 and UTX in T-ALL. We show that JMJD3 is essential for the initiation and maintenance of T-ALL, as it controls important oncogenic gene targets by modulating H3K27 methylation. By contrast, we found that UTX functions as a tumour suppressor and is frequently genetically inactivated in T-ALL. Moreover, we demonstrated that the small molecule inhibitor GSKJ4 (ref. 5) affects T-ALL growth, by targeting JMJD3 activity. These findings show that two proteins with a similar enzymatic function can have opposing roles in the context of the same disease, paving the way for treating haematopoietic malignancies with a new category of epigenetic inhibitors.


Subject(s)
Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Animals , Benzazepines/pharmacology , Epigenesis, Genetic/drug effects , Histone Demethylases/genetics , Histones/chemistry , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Lysine/metabolism , Methylation/drug effects , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Pyrimidines/pharmacology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
18.
J Biol Chem ; 293(7): 2422-2437, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29301935

ABSTRACT

Natural killer (NK) cells are innate lymphocytes, important in immune surveillance and elimination of stressed, transformed, or virus-infected cells. They critically shape the inflammatory cytokine environment to orchestrate interactions of cells of the innate and adaptive immune systems. Some studies have reported that NK cell activation and cytokine secretion are controlled epigenetically but have yielded only limited insight into the mechanisms. Using chemical screening with small-molecule inhibitors of chromatin methylation and acetylation, further validated by knockdown approaches, we here identified Jumonji-type histone H3K27 demethylases as key regulators of cytokine production in human NK cell subsets. The prototypic JMJD3/UTX (Jumonji domain-containing protein 3) H3K27 demethylase inhibitor GSK-J4 increased global levels of the repressive H3K27me3 mark around transcription start sites of effector cytokine genes. Moreover, GSK-J4 reduced IFN-γ, TNFα, granulocyte-macrophage colony-stimulating factor (GM-CSF), and interleukin-10 levels in cytokine-stimulated NK cells while sparing their cytotoxic killing activity against cancer cells. The anti-inflammatory effect of GSK-J4 in NK cell subsets, isolated from peripheral blood or tissue from individuals with rheumatoid arthritis (RA), coupled with an inhibitory effect on formation of bone-resorbing osteoclasts, suggested that histone demethylase inhibition has broad utility for modulating immune and inflammatory responses. Overall, our results indicate that H3K27me3 is a dynamic and important epigenetic modification during NK cell activation and that JMJD3/UTX-driven H3K27 demethylation is critical for NK cell function.


Subject(s)
Arthritis, Rheumatoid/enzymology , Histones/immunology , Jumonji Domain-Containing Histone Demethylases/immunology , Killer Cells, Natural/enzymology , Amino Acid Motifs , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Histones/chemistry , Histones/genetics , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Killer Cells, Natural/immunology , Phenotype , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
19.
Nat Chem Biol ; 12(7): 539-45, 2016 07.
Article in English | MEDLINE | ID: mdl-27214403

ABSTRACT

Members of the KDM5 (also known as JARID1) family are 2-oxoglutarate- and Fe(2+)-dependent oxygenases that act as histone H3K4 demethylases, thereby regulating cell proliferation and stem cell self-renewal and differentiation. Here we report crystal structures of the catalytic core of the human KDM5B enzyme in complex with three inhibitor chemotypes. These scaffolds exploit several aspects of the KDM5 active site, and their selectivity profiles reflect their hybrid features with respect to the KDM4 and KDM6 families. Whereas GSK-J1, a previously identified KDM6 inhibitor, showed about sevenfold less inhibitory activity toward KDM5B than toward KDM6 proteins, KDM5-C49 displayed 25-100-fold selectivity between KDM5B and KDM6B. The cell-permeable derivative KDM5-C70 had an antiproliferative effect in myeloma cells, leading to genome-wide elevation of H3K4me3 levels. The selective inhibitor GSK467 exploited unique binding modes, but it lacked cellular potency in the myeloma system. Taken together, these structural leads deliver multiple starting points for further rational and selective inhibitor design.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Jumonji Domain-Containing Histone Demethylases/chemistry , Jumonji Domain-Containing Histone Demethylases/metabolism , Multiple Myeloma/drug therapy , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Histone Demethylases/metabolism , Humans , Models, Molecular , Multiple Myeloma/pathology , Protein Conformation , Structure-Activity Relationship
20.
PLoS Biol ; 13(12): e1002316, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26646171

ABSTRACT

Trypanosoma brucei, the causative agent of African sleeping sickness, is transmitted to its mammalian host by the tsetse. In the fly, the parasite's surface is covered with invariant procyclin, while in the mammal it resides extracellularly in its bloodstream form (BF) and is densely covered with highly immunogenic Variant Surface Glycoprotein (VSG). In the BF, the parasite varies this highly immunogenic surface VSG using a repertoire of ~2500 distinct VSG genes. Recent reports in mammalian systems point to a role for histone acetyl-lysine recognizing bromodomain proteins in the maintenance of stem cell fate, leading us to hypothesize that bromodomain proteins may maintain the BF cell fate in trypanosomes. Using small-molecule inhibitors and genetic mutants for individual bromodomain proteins, we performed RNA-seq experiments that revealed changes in the transcriptome similar to those seen in cells differentiating from the BF to the insect stage. This was recapitulated at the protein level by the appearance of insect-stage proteins on the cell surface. Furthermore, bromodomain inhibition disrupts two major BF-specific immune evasion mechanisms that trypanosomes harness to evade mammalian host antibody responses. First, monoallelic expression of the antigenically varied VSG is disrupted. Second, rapid internalization of antibodies bound to VSG on the surface of the trypanosome is blocked. Thus, our studies reveal a role for trypanosome bromodomain proteins in maintaining bloodstream stage identity and immune evasion. Importantly, bromodomain inhibition leads to a decrease in virulence in a mouse model of infection, establishing these proteins as potential therapeutic drug targets for trypanosomiasis. Our 1.25Å resolution crystal structure of a trypanosome bromodomain in complex with I-BET151 reveals a novel binding mode of the inhibitor, which serves as a promising starting point for rational drug design.


Subject(s)
Models, Molecular , Protozoan Proteins/metabolism , Transcription Factors/metabolism , Trypanosoma brucei brucei/physiology , Amino Acid Substitution , Animals , Binding Sites , Cell Line , Gene Knockdown Techniques , Gene Knockout Techniques , Immune Evasion , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Conformation , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Survival Analysis , Transcription Factors/antagonists & inhibitors , Transcription Factors/chemistry , Transcription Factors/genetics , Trypanocidal Agents/pharmacology , Trypanocidal Agents/therapeutic use , Trypanosoma brucei brucei/drug effects , Trypanosoma brucei brucei/immunology , Trypanosoma brucei brucei/pathogenicity , Trypanosomiasis, African/drug therapy , Trypanosomiasis, African/parasitology , Trypanosomiasis, African/physiopathology , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL