Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
2.
Nat Genet ; 55(6): 1034-1047, 2023 06.
Article in English | MEDLINE | ID: mdl-37277650

ABSTRACT

Down syndrome (DS), the genetic condition caused by trisomy 21, is characterized by variable cognitive impairment, immune dysregulation, dysmorphogenesis and increased prevalence of diverse co-occurring conditions. The mechanisms by which trisomy 21 causes these effects remain largely unknown. We demonstrate that triplication of the interferon receptor (IFNR) gene cluster on chromosome 21 is necessary for multiple phenotypes in a mouse model of DS. Whole-blood transcriptome analysis demonstrated that IFNR overexpression associates with chronic interferon hyperactivity and inflammation in people with DS. To define the contribution of this locus to DS phenotypes, we used genome editing to correct its copy number in a mouse model of DS, which normalized antiviral responses, prevented heart malformations, ameliorated developmental delays, improved cognition and attenuated craniofacial anomalies. Triplication of the Ifnr locus modulates hallmarks of DS in mice, suggesting that trisomy 21 elicits an interferonopathy potentially amenable to therapeutic intervention.


Subject(s)
Down Syndrome , Heart Defects, Congenital , Animals , Mice , Down Syndrome/genetics , Receptors, Interferon/genetics , Interferons , Phenotype , Disease Models, Animal
3.
Cell Rep ; 42(3): 112230, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36897777

ABSTRACT

Inactivation of the p53 tumor suppressor, either by mutations or through hyperactivation of repressors such as MDM2 and MDM4, is a hallmark of cancer. Although many inhibitors of the p53-MDM2/4 interaction have been developed, such as Nutlin, their therapeutic value is limited by highly heterogeneous cellular responses. We report here a multi-omics investigation of the cellular response to MDM2/4 inhibitors, leading to identification of FAM193A as a widespread regulator of p53 function. CRISPR screening identified FAM193A as necessary for the response to Nutlin. FAM193A expression correlates with Nutlin sensitivity across hundreds of cell lines. Furthermore, genetic codependency data highlight FAM193A as a component of the p53 pathway across diverse tumor types. Mechanistically, FAM193A interacts with MDM4, and FAM193A depletion stabilizes MDM4 and inhibits the p53 transcriptional program. Last, FAM193A expression is associated with better prognosis in multiple malignancies. Altogether, these results identify FAM193A as a positive regulator of p53.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Antineoplastic Agents/pharmacology , Apoptosis , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Neoplasms/pathology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
4.
Cell Death Differ ; 30(4): 952-965, 2023 04.
Article in English | MEDLINE | ID: mdl-36681780

ABSTRACT

The p53 transcription factor is a master regulator of cellular responses to stress that is commonly inactivated in diverse cancer types. Despite decades of research, the mechanisms by which p53 impedes tumorigenesis across vastly different cellular contexts requires further investigation. The bulk of research has been completed using in vitro studies of cancer cell lines or in vivo studies in mouse models, but much less is known about p53 action in diverse non-transformed human tissues. Here, we investigated how different cellular states modify the p53 transcriptional program in human cells through a combination of computational analyses of publicly available large-scale datasets and in vitro studies using an isogenic system consisting of induced pluripotent stem cells (iPSCs) and two derived lineages. Analysis of publicly available mRNA expression and genetic dependency data demonstrated wide variation in terms of expression and function of a core p53 transcriptional program across various tissues and lineages. To monitor the impact of cell differentiation on the p53 transcriptome within an isogenic cell culture system, we activated p53 by pharmacological inhibition of its negative regulator MDM2. Using cell phenotyping assays and genome wide transcriptome analyses, we demonstrated that cell differentiation confines and modifies the p53 transcriptional network in a lineage-specific fashion. Although hundreds of p53 target genes are transactivated in iPSCs, only a small fraction is transactivated in each of the differentiated lineages. Mechanistic studies using small molecule inhibitors and genetic knockdowns revealed the presence of two major regulatory mechanisms contributing to this massive heterogeneity across cellular states: gene silencing by epigenetic regulatory complexes and constitutive transactivation by lineage-specific transcription factors. Altogether, these results illuminate the impact of cell differentiation on the p53 program, thus advancing our understanding of how this tumor suppressor functions in different contexts.


Subject(s)
Neoplasms , Tumor Suppressor Protein p53 , Mice , Animals , Humans , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Transcriptional Activation/genetics , Transcription Factors/metabolism , Cell Differentiation/genetics , Neoplasms/genetics , Gene Silencing
5.
Nat Commun ; 13(1): 7400, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36456590

ABSTRACT

The p53 transcription factor is a master regulator of cellular stress responses inhibited by repressors such as MDM2 and the phosphatase PPM1D. Activation of p53 with pharmacological inhibitors of its repressors is being tested in clinical trials for cancer therapy, but efficacy has been limited by poor induction of tumor cell death. We demonstrate that dual inhibition of MDM2 and PPM1D induces apoptosis in multiple cancer cell types via amplification of the p53 transcriptional program through the eIF2α-ATF4 pathway. PPM1D inhibition induces phosphorylation of eIF2α, ATF4 accumulation, and ATF4-dependent enhancement of p53-dependent transactivation upon MDM2 inhibition. Dual inhibition of p53 repressors depletes heme and induces HRI-dependent eIF2α phosphorylation. Pharmacological induction of eIF2α phosphorylation synergizes with MDM2 inhibition to induce cell death and halt tumor growth in mice. These results demonstrate that PPM1D inhibits both the p53 network and the integrated stress response controlled by eIF2α-ATF4, with clear therapeutic implications.


Subject(s)
Cell Death , Neoplasms , Protein Phosphatase 2C , Transcriptional Activation , Tumor Suppressor Protein p53 , Animals , Mice , Apoptosis , Eukaryotic Initiation Factor-2/genetics , Phosphorylation , Transcription Factors , Tumor Suppressor Protein p53/genetics , Protein Phosphatase 2C/metabolism
6.
Cancers (Basel) ; 14(17)2022 Aug 31.
Article in English | MEDLINE | ID: mdl-36077780

ABSTRACT

The aryl hydrocarbon receptor (AhR) plays a wide range of physiological roles in cellular processes such as proliferation, migration or control of immune responses. Several studies have also indicated that AhR might contribute to the regulation of energy balance or cellular metabolism. We observed that the AhR is upregulated in tumor epithelial cells derived from colon cancer patients. Using wild-type and the corresponding AhR knockout (AhR KO) variants of human colon cancer cell lines HCT116 and HT-29, we analyzed possible role(s) of the AhR in cell proliferation and metabolism, with a focus on regulation of the synthesis of fatty acids (FAs). We observed a decreased proliferation rate in the AhR KO cells, which was accompanied with altered cell cycle progression, as well as a decreased ATP production. We also found reduced mRNA levels of key enzymes of the FA biosynthetic pathway in AhR KO colon cancer cells, in particular of stearoyl-CoA desaturase 1 (SCD1). The loss of AhR was also associated with reduced expression and/or activity of components of the PI3K/Akt pathway, which controls lipid metabolism, and other lipogenic transcriptional regulators, such as sterol regulatory element binding transcription factor 1 (SREBP1). Together, our data indicate that disruption of AhR activity in colon tumor cells may, likely in a cell-specific manner, limit their proliferation, which could be linked with a suppressive effect on their endogenous FA metabolism. More attention should be paid to potential mechanistic links between overexpressed AhR and colon tumor cell metabolism.

7.
Methods Mol Biol ; 2267: 19-56, 2021.
Article in English | MEDLINE | ID: mdl-33786783

ABSTRACT

The transcription factor p53 controls a gene expression program with pleiotropic effects on cell biology including cell cycle arrest and apoptosis. Identifying direct p53 target genes within this network and determining how they influence cell fate decisions downstream of p53 activation is a prerequisite for designing therapeutic approaches that target p53 to effectively kill cancer cells. Here we describe a comprehensive multi-omics approach for identifying genes that are direct transcriptional targets of p53. We provide detailed procedures for measuring global RNA polymerase activity, defining p53 binding sites across the genome, and quantifying changes in steady-state mRNA in response to p53 activation.


Subject(s)
Chromatin Immunoprecipitation Sequencing/methods , Genomics/methods , RNA-Seq/methods , Transcriptome , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line , Humans , Transcriptional Activation
8.
Nat Commun ; 12(1): 1375, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33654095

ABSTRACT

Cellular adaptation to hypoxia is a hallmark of cancer, but the relative contribution of hypoxia-inducible factors (HIFs) versus other oxygen sensors to tumorigenesis is unclear. We employ a multi-omics pipeline including measurements of nascent RNA to characterize transcriptional changes upon acute hypoxia. We identify an immediate early transcriptional response that is strongly dependent on HIF1A and the kinase activity of its cofactor CDK8, includes indirect repression of MYC targets, and is highly conserved across cancer types. HIF1A drives this acute response via conserved high-occupancy enhancers. Genetic screen data indicates that, in normoxia, HIF1A displays strong cell-autonomous tumor suppressive effects through a gene module mediating mTOR inhibition. Conversely, in advanced malignancies, expression of a module of HIF1A targets involved in collagen remodeling is associated with poor prognosis across diverse cancer types. In this work, we provide a valuable resource for investigating context-dependent roles of HIF1A and its targets in cancer biology.


Subject(s)
Gene Regulatory Networks , Genes, Tumor Suppressor , Genomics , Hypoxia/genetics , Oncogenes , Cell Line, Tumor , Cell Survival , Cyclin-Dependent Kinase 8/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Genome, Human , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasms/genetics , Neoplasms/pathology , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , TOR Serine-Threonine Kinases/metabolism , Transcription, Genetic , Transcriptional Activation/genetics , Up-Regulation/genetics
9.
Chemosphere ; 263: 128126, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33297115

ABSTRACT

Deciphering the role of the aryl hydrocarbon receptor (AhR) in lung cancer cells may help us to better understand the role of toxic AhR ligands in lung carcinogenesis, including cancer progression. We employed human lung carcinoma A549 cells to investigate their fate after continuous two-week exposure to model AhR agonists, genotoxic benzo[a]pyrene (BaP; 1 µM) and non-genotoxic 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; 10 nM). While TCDD increased proliferative rate of A549 cells, exposure to BaP decreased cell proliferation and induced epithelial-to-mesenchymal transition (EMT)-like phenotype, which was associated with enhanced cell migration, invasion, and altered cell morphology. Although TCDD also suppressed expression of E-cadherin and activated some genes linked to EMT, it did not induce the EMT-like phenotype. The results of transcriptomic analysis, and the opposite effects of BaP and TCDD on cell proliferation, indicated that a delay in cell cycle progression, together with a slight increase of senescence (when coupled with AhR activation), favors the induction of EMT-like phenotype. The shift towards EMT-like phenotype observed after simultaneous treatment with TCDD and mitomycin C (an inhibitor of cell proliferation) confirmed the hypothesis. Since BaP decreased cell proliferative rate via induction of p21 expression, we generated the A549 cell model with reduced p21 expression and exposed it to BaP for two weeks. The p21 knockdown suppressed the BaP-mediated EMT-like phenotype in A549 cells, thus confirming that a delayed cell cycle progression, together with p21-dependent induction of senescence-related chemokine CCL2, may contribute to induction of EMT-like cell phenotype in lung cells exposed to genotoxic AhR ligands.


Subject(s)
Carcinoma , Lung Neoplasms , Benzo(a)pyrene/toxicity , Epithelial Cells , Humans , Lung , Lung Neoplasms/genetics , Phenotype , Receptors, Aryl Hydrocarbon/genetics
10.
Cell Rep ; 33(7): 108407, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33207208

ABSTRACT

Individuals with Down syndrome (DS; trisomy 21) display hyperactivation of interferon (IFN) signaling and chronic inflammation, which could potentially be explained by the extra copy of four IFN receptor (IFNR) genes encoded on chromosome 21. However, the clinical effects of IFN hyperactivity in DS remain undefined. Here, we report that a commonly used mouse model of DS overexpresses IFNR genes and shows hypersensitivity to IFN ligands in diverse immune cell types. When treated repeatedly with a TLR3 agonist to induce chronic inflammation, these animals overexpress key IFN-stimulated genes, induce cytokine production, exhibit liver pathology, and undergo rapid weight loss. Importantly, the lethal immune hypersensitivity and cytokine production and the ensuing pathology are ameliorated by JAK1 inhibition. These results indicate that individuals with DS may experience harmful hyperinflammation upon IFN-inducing immune stimuli, as observed during severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, pointing to JAK1 inhibition as a strategy to restore immune homeostasis in DS.


Subject(s)
Azetidines/therapeutic use , Down Syndrome/immunology , Hypersensitivity/drug therapy , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Sulfonamides/therapeutic use , Animals , Down Syndrome/complications , Female , Hypersensitivity/etiology , Hypersensitivity/immunology , Immunity, Innate , Interferon-alpha/metabolism , Liver/immunology , Male , Mice , Mice, Inbred C57BL , Purines , Pyrazoles , Toll-Like Receptors/metabolism
11.
Int J Mol Sci ; 21(8)2020 Apr 17.
Article in English | MEDLINE | ID: mdl-32316498

ABSTRACT

The efforts for therapeutic targeting of the aryl hydrocarbon receptor (AhR) have emerged in recent years. We investigated the effects of available antimigraine triptan drugs, having an indole core in their structure, on AhR signaling in human hepatic and intestinal cells. Activation of AhR in reporter gene assays was observed for Avitriptan and to a lesser extent for Donitriptan, while other triptans were very weak or no activators of AhR. Using competitive binding assay and by homology docking, we identified Avitriptan as a low-affinity ligand of AhR. Avitriptan triggered nuclear translocation of AhR and increased binding of AhR in CYP1A1 promotor DNA, as revealed by immune-fluorescence microscopy and chromatin immune-precipitation assay, respectively. Strong induction of CYP1A1 mRNA was achieved by Avitriptan in wild type but not in AhR-knockout, immortalized human hepatocytes, implying that induction of CYP1A1 is AhR-dependent. Increased levels of CYP1A1 mRNA by Avitriptan were observed in human colon carcinoma cells LS180 but not in primary cultures of human hepatocytes. Collectively, we show that Avitriptan is a weak ligand and activator of human AhR, which induces the expression of CYP1A1 in a cell-type specific manner. Our data warrant the potential off-label therapeutic application of Avitriptan as an AhR-agonist drug.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cytochrome P-450 CYP1A1/genetics , Hepatocytes/metabolism , Intestinal Mucosa/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Sulfonamides/pharmacology , Tryptamines/pharmacology , Basic Helix-Loop-Helix Transcription Factors/agonists , Basic Helix-Loop-Helix Transcription Factors/chemistry , Cells, Cultured , Drug Repositioning , Enzyme Activation/drug effects , Humans , Ligands , Models, Molecular , Molecular Docking Simulation , Organ Specificity , Promoter Regions, Genetic/drug effects , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/chemistry , Up-Regulation
12.
Cell Rep ; 30(13): 4355-4369.e6, 2020 03 31.
Article in English | MEDLINE | ID: mdl-32234473

ABSTRACT

Activation of p53 by the small molecule Nutlin can result in a combination of cell cycle arrest and apoptosis. The relative strength of these events is difficult to predict by classical gene expression analysis, leaving uncertainty as to the therapeutic benefits. In this study, we report a translational control mechanism shaping p53-dependent apoptosis. Using polysome profiling, we establish Nutlin-induced apoptosis to associate with the enhanced translation of mRNAs carrying multiple copies of an identified 3' UTR CG-rich motif mediating p53-dependent death (CGPD-motif). We identify PCBP2 and DHX30 as CGPD-motif interactors. We find that in cells undergoing persistent cell cycle arrest in response to Nutlin, CGPD-motif mRNAs are repressed by the PCBP2-dependent binding of DHX30 to the motif. Upon DHX30 depletion in these cells, the translation of CGPD-motif mRNAs increases, and the response to Nutlin shifts toward apoptosis. Instead, DHX30 inducible overexpression in SJSA1 cells leads to decreased translation of CGPD-motif mRNAs.


Subject(s)
Apoptosis/drug effects , Imidazoles/pharmacology , Piperazines/pharmacology , Protein Biosynthesis/drug effects , RNA Helicases/metabolism , RNA-Binding Proteins/metabolism , 3' Untranslated Regions/genetics , Base Sequence , Cell Cycle Checkpoints/drug effects , Cell Line , Gene Expression Regulation/drug effects , Gene Silencing/drug effects , Humans , Neoplasm Proteins/metabolism , Nucleotide Motifs/genetics , Phenotype , Polyribosomes/drug effects , Polyribosomes/metabolism , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism
13.
Mol Cell ; 76(3): 485-499.e8, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31495563

ABSTRACT

Transcriptional responses to external stimuli remain poorly understood. Using global nuclear run-on followed by sequencing (GRO-seq) and precision nuclear run-on sequencing (PRO-seq), we show that CDK8 kinase activity promotes RNA polymerase II pause release in response to interferon-γ (IFN-γ), a universal cytokine involved in immunity and tumor surveillance. The Mediator kinase module contains CDK8 or CDK19, which are presumed to be functionally redundant. We implemented cortistatin A, chemical genetics, transcriptomics, and other methods to decouple their function while assessing enzymatic versus structural roles. Unexpectedly, CDK8 and CDK19 regulated different gene sets via distinct mechanisms. CDK8-dependent regulation required its kinase activity, whereas CDK19 governed IFN-γ responses through its scaffolding function (i.e., it was kinase independent). Accordingly, CDK8, not CDK19, phosphorylates the STAT1 transcription factor (TF) during IFN-γ stimulation, and CDK8 kinase inhibition blocked activation of JAK-STAT pathway TFs. Cytokines such as IFN-γ rapidly mobilize TFs to "reprogram" cellular transcription; our results implicate CDK8 and CDK19 as essential for this transcriptional reprogramming.


Subject(s)
Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinases/metabolism , Fibroblasts/drug effects , Interferon-gamma/pharmacology , Transcription, Genetic/drug effects , Animals , Cyclin-Dependent Kinase 8/genetics , Cyclin-Dependent Kinases/genetics , Fibroblasts/enzymology , Fibroblasts/virology , HCT116 Cells , Host-Pathogen Interactions , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , RNA Polymerase II/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction , Vesiculovirus/pathogenicity
14.
Cell Rep ; 24(12): 3224-3236, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30232004

ABSTRACT

The transcriptional repressor ΔNp63α is a potent oncogene widely overexpressed in squamous cell carcinomas (SCCs) of diverse tissue origins, where it promotes malignant cell proliferation and survival. We report here the results of a genome-wide CRISPR screen to identify pathways controlling ΔNp63α-dependent cell proliferation, which revealed that the small GTPase RHOA blocks cell division upon ΔNp63α knockdown. After ΔNp63α depletion, RHOA activity is increased, and cells undergo RHOA-dependent proliferation arrest along with transcriptome changes indicative of increased TGF-ß signaling. Mechanistically, ΔNp63α represses transcription of TGFB2, which induces a cell cycle arrest that is partially dependent on RHOA. Ectopic TGFB2 activates RHOA and impairs SCC proliferation, and TGFB2 neutralization restores cell proliferation during ΔNp63α depletion. Genomic data from tumors demonstrate inactivation of RHOA and the TGFBR2 receptor and ΔNp63α overexpression in more than 80% of lung SCCs. These results reveal a signaling pathway controlling SCC proliferation that is potentially amenable to pharmacological intervention.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cell Proliferation , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Transcription Factors/genetics , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta2/metabolism , Tumor Suppressor Proteins/genetics , rhoA GTP-Binding Protein/metabolism
15.
Toxicol Lett ; 292: 162-174, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29704546

ABSTRACT

Exposure to persistent ligands of aryl hydrocarbon receptor (AhR) has been found to cause lung cancer in experimental animals, and lung adenocarcinomas are often associated with enhanced AhR expression and aberrant AhR activation. In order to better understand the action of toxic AhR ligands in lung epithelial cells, we performed global gene expression profiling and analyze TCDD-induced changes in A549 transcriptome, both sensitive and non-sensitive to CH223191 co-treatment. Comparison of our data with results from previously reported microarray and ChIP-seq experiments enabled us to identify candidate genes, which expression status reflects exposure of lung cancer cells to TCDD, and to predict processes, pathways (e.g. ER stress, Wnt/ß-cat, IFNÉ£, EGFR/Erbb1), putative TFs (e.g. STAT, AP1, E2F1, TCF4), which may be implicated in adaptive response of lung cells to TCDD-induced AhR activation. Importantly, TCDD-like expression fingerprint of selected genes was observed also in A549 cells exposed acutely to both toxic (benzo[a]pyrene, benzo[k]fluoranthene) and endogenous AhR ligands (2-(1H-Indol-3-ylcarbonyl)-4-thiazolecarboxylic acid methyl ester and 6-formylindolo[3,2-b]carbazole). Overall, our results suggest novel cellular candidates, which could help to improve monitoring of AhR-dependent transcriptional activity during acute exposure of lung cells to distinct types of environmental pollutants.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/agonists , Environmental Pollutants/toxicity , Gene Expression Regulation, Neoplastic/drug effects , Lung Neoplasms/genetics , Receptors, Aryl Hydrocarbon/agonists , Transcriptome/drug effects , A549 Cells , Azo Compounds/toxicity , Basic Helix-Loop-Helix Transcription Factors/metabolism , Benzo(a)pyrene/toxicity , Carbazoles/toxicity , Fluorenes/toxicity , Gene Expression Profiling/methods , Gene Regulatory Networks/drug effects , Humans , Indoles/toxicity , Ligands , Lung Neoplasms/metabolism , Oligonucleotide Array Sequence Analysis , Polychlorinated Dibenzodioxins/toxicity , Pyrazoles/toxicity , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction/drug effects , Thiazoles/toxicity , Time Factors , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects
16.
Dev Cell ; 44(5): 555-565.e3, 2018 03 12.
Article in English | MEDLINE | ID: mdl-29533771

ABSTRACT

Macroautophagy (autophagy) is intimately linked with cell death and allows cells to evade apoptosis. This has prompted clinical trials to combine autophagy inhibitors with other drugs with the aim of increasing the likelihood of cancer cells dying. However, the molecular basis for such effects is unknown. Here, we describe a transcriptional mechanism that connects autophagy to apoptosis. The autophagy-regulating transcription factor, FOXO3a, is itself turned over by basal autophagy creating a potential feedback loop. Increased FOXO3a upon autophagy inhibition stimulates transcription of the pro-apoptotic BBC3/PUMA gene to cause apoptosis sensitization. This mechanism explains how autophagy inhibition can sensitize tumor cells to chemotherapy drugs and allows an autophagy inhibitor to change the action of an MDM2-targeted drug from growth inhibition to apoptosis, reducing tumor burden in vivo. Thus, a link between two processes mediated via a single transcription factor binding site in the genome can be leveraged to improve anti-cancer therapies.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Autophagy/drug effects , Breast Neoplasms/pathology , Colonic Neoplasms/pathology , Forkhead Box Protein O3/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Proto-Oncogene Proteins/metabolism , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Female , Forkhead Box Protein O3/genetics , Humans , Proto-Oncogene Proteins/genetics , Tumor Cells, Cultured
17.
Cell Death Differ ; 25(1): 133-143, 2018 01.
Article in English | MEDLINE | ID: mdl-29125602

ABSTRACT

p53 is a transcription factor that suppresses tumor growth through regulation of dozens of target genes with diverse biological functions. The activity of this master transcription factor is inactivated in nearly all tumors, either by mutations in the TP53 locus or by oncogenic events that decrease the activity of the wild-type protein, such as overexpression of the p53 repressor MDM2. However, despite decades of intensive research, our collective understanding of the p53 signaling cascade remains incomplete. In this review, we focus on recent advances in our understanding of mechanisms of p53-dependent transcriptional control as they relate to five key areas: (1) the functionally distinct N-terminal transactivation domains, (2) the diverse regulatory roles of its C-terminal domain, (3) evidence that p53 is solely a direct transcriptional activator, not a direct repressor, (4) the ability of p53 to recognize many of its enhancers across diverse chromatin environments, and (5) mechanisms that modify the p53-dependent transcriptional program in a context-dependent manner.


Subject(s)
Transcriptional Activation , Tumor Suppressor Protein p53/metabolism , Animals , Humans , Mice , Protein Domains , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/chemistry
18.
Cell Rep ; 21(6): 1495-1506, 2017 Nov 07.
Article in English | MEDLINE | ID: mdl-29117556

ABSTRACT

Aerobic glycolysis, also known as the Warburg effect, is a hallmark of cancerous tissues. Despite its importance in cancer development, our understanding of mechanisms driving this form of metabolic reprogramming is incomplete. We report here an analysis of colorectal cancer cells engineered to carry a single point mutation in the active site of the Mediator-associated kinase CDK8, creating hypomorphic alleles sensitive to bulky ATP analogs. Transcriptome analysis revealed that CDK8 kinase activity is required for the expression of many components of the glycolytic cascade. CDK8 inhibition impairs glucose transporter expression, glucose uptake, glycolytic capacity and reserve, as well as cell proliferation and anchorage-independent growth, both in normoxia and hypoxia. Importantly, CDK8 impairment sensitizes cells to pharmacological glycolysis inhibition, a result reproduced with Senexin A, a dual inhibitor of CDK8/CDK19. Altogether, these results contribute to our understanding of CDK8 as an oncogene, and they justify investigations to target CDK8 in highly glycolytic tumors.


Subject(s)
Cyclin-Dependent Kinase 8/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 8/antagonists & inhibitors , Cyclin-Dependent Kinase 8/genetics , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Deoxyglucose/metabolism , Deoxyglucose/pharmacology , Down-Regulation/drug effects , Gene Editing , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , Glycolysis/drug effects , HCT116 Cells , Hexokinase/genetics , Hexokinase/metabolism , Humans , Mutagenesis, Site-Directed , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Transcriptome/drug effects , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
19.
Genome Res ; 27(10): 1645-1657, 2017 10.
Article in English | MEDLINE | ID: mdl-28904012

ABSTRACT

The tumor suppressor TP53 is the most frequently mutated gene product in human cancer. Close to half of all solid tumors carry inactivating mutations in the TP53 gene, while in the remaining cases, TP53 activity is abrogated by other oncogenic events, such as hyperactivation of its endogenous repressors MDM2 or MDM4. Despite identification of hundreds of genes regulated by this transcription factor, it remains unclear which direct target genes and downstream pathways are essential for the tumor suppressive function of TP53. We set out to address this problem by generating multiple genomic data sets for three different cancer cell lines, allowing the identification of distinct sets of TP53-regulated genes, from early transcriptional targets through to late targets controlled at the translational level. We found that although TP53 elicits vastly divergent signaling cascades across cell lines, it directly activates a core transcriptional program of ∼100 genes with diverse biological functions, regardless of cell type or cellular response to TP53 activation. This core program is associated with high-occupancy TP53 enhancers, high levels of paused RNA polymerases, and accessible chromatin. Interestingly, two different shRNA screens failed to identify a single TP53 target gene required for the anti-proliferative effects of TP53 during pharmacological activation in vitro. Furthermore, bioinformatics analysis of thousands of cancer genomes revealed that none of these core target genes are frequently inactivated in tumors expressing wild-type TP53. These results support the hypothesis that TP53 activates a genetically robust transcriptional program with highly distributed tumor suppressive functions acting in diverse cellular contexts.


Subject(s)
Enhancer Elements, Genetic , Neoplasms/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Cell Cycle Proteins , Humans , MCF-7 Cells , Neoplasms/genetics , Neoplasms/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/genetics
20.
Oncotarget ; 7(15): 20425-39, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26967250

ABSTRACT

The dual specificity phosphatase Cdc25A is a key regulator of the cell cycle that promotes cell cycle progression by dephosphorylating and activating cyclin-dependent kinases. In response to genotoxicants, Cdc25A undergoes posttranslational modifications which contribute to its proteasome-mediated degradation and consequent cell cycle checkpoint arrest. The most thoroughly studied Cdc25A modification is phosphorylation. We now provide the first evidence that Cdc25A can be acetylated and that it directly interacts with the ARD1 acetyltransferase which acetylates Cdc25A both biochemically and in cultured cells. When acetylated, Cdc25A has an extended half-life. We have also identified the class IV histone deacetylase, HDAC11, as a Cdc25A deacetylase. We further show that DNA damage, such as exposure to methyl methanesulfonate (MMS), etoposide or arsenic, increases Cdc25A acetylation. Importantly, this acetylation modulates Cdc25A phosphatase activity and its function as a cell cycle regulator, and may reflect a cellular response to DNA damage. Since Cdc25A, ARD1, and HDAC11 are frequently dysregulated in multiple types of cancer, our findings may provide insight into a novel mechanism in carcinogenesis.


Subject(s)
Histone Deacetylases/metabolism , N-Terminal Acetyltransferase A/metabolism , N-Terminal Acetyltransferase E/metabolism , Neoplasms/pathology , cdc25 Phosphatases/metabolism , Acetylation , Apoptosis , Cell Cycle , Cell Proliferation , Cells, Cultured , DNA Damage , HEK293 Cells , Humans , Neoplasms/metabolism , Phosphorylation , Protein Processing, Post-Translational , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...