Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Nat Rev Mol Cell Biol ; 25(1): 46-64, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37710009

ABSTRACT

The forkhead box protein O (FOXO, consisting of FOXO1, FOXO3, FOXO4 and FOXO6) transcription factors are the mammalian orthologues of Caenorhabditis elegans DAF-16, which gained notoriety for its capability to double lifespan in the absence of daf-2 (the gene encoding the worm insulin receptor homologue). Since then, research has provided many mechanistic details on FOXO regulation and FOXO activity. Furthermore, conditional knockout experiments have provided a wealth of data as to how FOXOs control development and homeostasis at the organ and organism levels. The lifespan-extending capabilities of DAF-16/FOXO are highly correlated with their ability to induce stress response pathways. Exogenous and endogenous stress, such as cellular redox stress, are considered the main drivers of the functional decline that characterizes ageing. Functional decline often manifests as disease, and decrease in FOXO activity indeed negatively impacts on major age-related diseases such as cancer and diabetes. In this context, the main function of FOXOs is considered to preserve cellular and organismal homeostasis, through regulation of stress response pathways. Paradoxically, the same FOXO-mediated responses can also aid the survival of dysfunctional cells once these eventually emerge. This general property to control stress responses may underlie the complex and less-evident roles of FOXOs in human lifespan as opposed to model organisms such as C. elegans.


Subject(s)
Caenorhabditis elegans , Signal Transduction , Animals , Humans , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Signal Transduction/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Aging/genetics , Longevity/genetics , Mammals/metabolism
2.
Mol Cell ; 83(22): 4141-4157.e11, 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37977121

ABSTRACT

Biomolecular condensates have emerged as a major organizational principle in the cell. However, the formation, maintenance, and dissolution of condensates are still poorly understood. Transcriptional machinery partitions into biomolecular condensates at key cell identity genes to activate these. Here, we report a specific perturbation of WNT-activated ß-catenin condensates that disrupts oncogenic signaling. We use a live-cell condensate imaging method in human cancer cells to discover FOXO and TCF-derived peptides that specifically inhibit ß-catenin condensate formation on DNA, perturb nuclear ß-catenin condensates in cells, and inhibit ß-catenin-driven transcriptional activation and colorectal cancer cell growth. We show that these peptides compete with homotypic intermolecular interactions that normally drive condensate formation. Using this framework, we derive short peptides that specifically perturb condensates and transcriptional activation of YAP and TAZ in the Hippo pathway. We propose a "monomer saturation" model in which short interacting peptides can be used to specifically inhibit condensate-associated transcription in disease.


Subject(s)
Neoplasms , beta Catenin , Humans , beta Catenin/genetics , beta Catenin/metabolism , Signal Transduction , Hippo Signaling Pathway , Peptides/genetics
3.
Nat Rev Mol Cell Biol ; 14(2): 83-97, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23325358

ABSTRACT

Forkhead box O (FOXO) transcription factors are involved in the regulation of the cell cycle, apoptosis and metabolism. In model organisms, FOXO activity also affects stem cell maintenance and lifespan as well as age-related diseases, such as cancer and diabetes. Multiple upstream pathways regulate FOXO activity through post-translational modifications and nuclear-cytoplasmic shuttling of both FOXO and its regulators. The diversity of this upstream regulation and the downstream effects of FOXOs suggest that they function as homeostasis regulators to maintain tissue homeostasis over time and coordinate a response to environmental changes, including growth factor deprivation, metabolic stress (starvation) and oxidative stress.


Subject(s)
Forkhead Transcription Factors/physiology , Homeostasis/genetics , Forkhead Transcription Factors/chemistry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Homeostasis/physiology , Humans , Models, Biological , Oxidative Stress/genetics , Oxidative Stress/physiology , Protein Processing, Post-Translational/physiology , Signal Transduction/genetics , Stress, Physiological/genetics , Stress, Physiological/physiology , Structure-Activity Relationship
4.
Nature ; 543(7645): 424-427, 2017 03 16.
Article in English | MEDLINE | ID: mdl-28273069

ABSTRACT

The small intestinal epithelium self-renews every four or five days. Intestinal stem cells (Lgr5+ crypt base columnar cells (CBCs)) sustain this renewal and reside between terminally differentiated Paneth cells at the bottom of the intestinal crypt. Whereas the signalling requirements for maintaining stem cell function and crypt homeostasis have been well studied, little is known about how metabolism contributes to epithelial homeostasis. Here we show that freshly isolated Lgr5+ CBCs and Paneth cells from the mouse small intestine display different metabolic programs. Compared to Paneth cells, Lgr5+ CBCs display high mitochondrial activity. Inhibition of mitochondrial activity in Lgr5+ CBCs or inhibition of glycolysis in Paneth cells strongly affects stem cell function, as indicated by impaired organoid formation. In addition, Paneth cells support stem cell function by providing lactate to sustain the enhanced mitochondrial oxidative phosphorylation in the Lgr5+ CBCs. Mechanistically, we show that oxidative phosphorylation stimulates p38 MAPK activation by mitochondrial reactive oxygen species signalling, thereby establishing the mature crypt phenotype. Together, our results reveal a critical role for the metabolic identity of Lgr5+ CBCs and Paneth cells in supporting optimal stem cell function, and we identify mitochondria and reactive oxygen species signalling as a driving force of cellular differentiation.


Subject(s)
Cell Self Renewal , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestine, Small/cytology , Intestine, Small/metabolism , Stem Cells/cytology , Animals , Cell Differentiation , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/pharmacology , Glycolysis , Homeostasis , Lactic Acid/metabolism , Mice , Mitochondria/metabolism , Organoids/cytology , Organoids/drug effects , Organoids/metabolism , Oxidative Phosphorylation , Paneth Cells/cytology , Paneth Cells/metabolism , Reactive Oxygen Species/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Stem Cells/physiology , Wnt3A Protein/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Mol Cell ; 49(4): 730-42, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23333309

ABSTRACT

Forkhead box O (FOXO; DAF-16 in worms) transcription factors, which are of vital importance in cell-cycle control, stress resistance, tumor suppression, and organismal lifespan, are largely regulated through nucleo-cytoplasmic shuttling. Insulin signaling keeps FOXO/DAF-16 cytoplasmic, and hence transcriptionally inactive. Conversely, as in loss of insulin signaling, reactive oxygen species (ROS) can activate FOXO/DAF-16 through nuclear accumulation. How ROS regulate the nuclear translocation of FOXO/DAF-16 is largely unknown. Cysteine oxidation can stabilize protein-protein interactions through the formation of disulfide-bridges when cells encounter ROS. Using a proteome-wide screen that identifies ROS-induced mixed disulfide-dependent complexes, we discovered several interaction partners of FOXO4, one of which is the nuclear import receptor transportin-1. We show that disulfide formation with transportin-1 is required for nuclear localization and the activation of FOXO4/DAF-16 induced by ROS, but not by the loss of insulin signaling. This molecular mechanism for nuclear shuttling is conserved in C. elegans and directly connects redox signaling to the longevity protein FOXO/DAF-16.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Transcription Factors/metabolism , beta Karyopherins/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Amino Acid Substitution , Animals , Caenorhabditis elegans/cytology , Cell Cycle Proteins , Cell Nucleus/metabolism , Cystine/metabolism , Forkhead Transcription Factors , HEK293 Cells , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Oxidation-Reduction , Protein Binding , Reactive Oxygen Species/metabolism , Transcription Factors/genetics , beta Karyopherins/physiology
6.
J Biol Chem ; 294(4): 1128-1141, 2019 01 25.
Article in English | MEDLINE | ID: mdl-30530489

ABSTRACT

Lysine methylation is an important post-translational modification that is also present on mitochondrial proteins, but the mitochondrial lysine-specific methyltransferases (KMTs) responsible for modification are in most cases unknown. Here, we set out to determine the function of human family with sequence similarity 173 member B (FAM173B), a mitochondrial methyltransferase (MTase) reported to promote chronic pain. Using bioinformatics analyses and biochemical assays, we found that FAM173B contains an atypical, noncleavable mitochondrial targeting sequence responsible for its localization to mitochondria. Interestingly, CRISPR/Cas9-mediated KO of FAM173B in mammalian cells abrogated trimethylation of Lys-43 in ATP synthase c-subunit (ATPSc), a modification previously reported as ubiquitous among metazoans. ATPSc methylation was restored by complementing the KO cells with enzymatically active human FAM173B or with a putative FAM173B orthologue from the nematode Caenorhabditis elegans Interestingly, lack of Lys-43 methylation caused aberrant incorporation of ATPSc into the ATP synthase complex and resulted in decreased ATP-generating ability of the complex, as well as decreased mitochondrial respiration. In summary, we have identified FAM173B as the long-sought KMT responsible for methylation of ATPSc, a key protein in cellular ATP production, and have demonstrated functional significance of ATPSc methylation. We suggest renaming FAM173B to ATPSc-KMT (gene name ATPSCKMT).


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Lysine/metabolism , Mitochondria/enzymology , Mitochondrial Proton-Translocating ATPases/metabolism , Animals , Cell Line , Computational Biology , HeLa Cells , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/genetics , Humans , Methylation , Mice , Mitochondria/metabolism
7.
Rheumatology (Oxford) ; 58(12): 2305-2314, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31131409

ABSTRACT

OBJECTIVE: A considerable body of evidence supports a role for type-I IFN in the pathogenesis of primary SS (pSS). As plasmacytoid dendritic cells (pDCs) are a major source of type-I IFN, we investigated their molecular regulation by measuring expression of a large set of miRNAs. METHODS: pDCs were isolated from peripheral blood of pSS patients (n = 30) and healthy controls (n = 16) divided into two independent cohorts (discovery and replication). Screening of 758 miRNAs was assessed by an OpenArray quantitative PCR-based technique; replication of a set of identified miRNAs was performed by custom array. Functional annotation of miRNA targets was performed using pathway enrichment. Novel targets of miR-29a and miR-29c were identified using a proteomic approach (stable isotope labelling with amino acids in cell culture). RESULTS: In the discovery cohort, 20 miRNAs were differentially expressed in pSS pDCs compared with healthy control pDCs. Of these, differential expression of 10 miRNAs was confirmed in the replication cohort. The dysregulated miRNAs were involved in phosphoinositide 3-kinase-Ak strain transforming and mammalian target of rapamycin signalling, as well as regulation of cell death. In addition, a set of novel protein targets of miR-29a and miR-29c were identified, including five targets that were regulated by both miRs. CONCLUSION: The dysregulated miRNome in pDCs of patients with pSS is associated with aberrant regulation of processes at the centre of pDC function, including type-I IFN production and cell death. As miR-29a and miR-29c are pro-apoptotic factors and several of the novel targets identified here are regulators of apoptosis, their downregulation in patients with pSS is associated with enhanced pDC survival.


Subject(s)
Dendritic Cells/metabolism , Gene Expression Regulation , MicroRNAs/genetics , Sjogren's Syndrome/genetics , Adult , Aged , Cells, Cultured , Dendritic Cells/pathology , Down-Regulation , Female , Humans , Male , MicroRNAs/biosynthesis , Middle Aged , Proteomics/methods , RNA/genetics , Signal Transduction , Sjogren's Syndrome/metabolism , Sjogren's Syndrome/pathology
8.
Gastroenterology ; 152(6): 1462-1476.e10, 2017 05.
Article in English | MEDLINE | ID: mdl-28130067

ABSTRACT

BACKGROUND & AIMS: The nuclear receptor subfamily 1 group H member 4 (NR1H4 or farnesoid X receptor [FXR]) regulates bile acid synthesis, transport, and catabolism. FXR also regulates postprandial lipid and glucose metabolism. We performed quantitative proteomic analyses of liver tissues from mice to evaluate these functions and investigate whether FXR regulates amino acid metabolism. METHODS: To study the role of FXR in mouse liver, we used mice with a disruption of Nr1h4 (FXR-knockout mice) and compared them with floxed control mice. Mice were gavaged with the FXR agonist obeticholic acid or vehicle for 11 days. Proteome analyses, as well as targeted metabolomics and chromatin immunoprecipitation, were performed on the livers of these mice. Primary rat hepatocytes were used to validate the role of FXR in amino acid catabolism by gene expression and metabolomics studies. Finally, control mice and mice with liver-specific disruption of Nr1h4 (liver FXR-knockout mice) were re-fed with a high-protein diet after 6 hours fasting and gavaged a 15NH4Cl tracer. Gene expression and the metabolome were studied in the livers and plasma from these mice. RESULTS: In livers of control mice and primary rat hepatocytes, activation of FXR with obeticholic acid increased expression of proteins that regulate amino acid degradation, ureagenesis, and glutamine synthesis. We found FXR to bind to regulatory sites of genes encoding these proteins in control livers. Liver tissues from FXR-knockout mice had reduced expression of urea cycle proteins, and accumulated precursors of ureagenesis, compared with control mice. In liver FXR-knockout mice on a high-protein diet, the plasma concentration of newly formed urea was significantly decreased compared with controls. In addition, liver FXR-knockout mice had reduced hepatic expression of enzymes that regulate ammonium detoxification compared with controls. In contrast, obeticholic acid increased expression of genes encoding enzymes involved in ureagenesis compared with vehicle in C57Bl/6 mice. CONCLUSIONS: In livers of mice, FXR regulates amino acid catabolism and detoxification of ammonium via ureagenesis and glutamine synthesis. Failure of the urea cycle and hyperammonemia are common in patients with acute and chronic liver diseases; compounds that activate FXR might promote ammonium clearance in these patients.


Subject(s)
Ammonia/metabolism , Glutamine/biosynthesis , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Urea/metabolism , Animals , Bile Acids and Salts/metabolism , Chenodeoxycholic Acid/analogs & derivatives , Chenodeoxycholic Acid/pharmacology , Dietary Proteins/administration & dosage , Gene Expression , Hepatocytes , Liver/enzymology , Male , Metabolome , Mice , Mice, Inbred C57BL , Mice, Knockout , Proteome , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors
9.
EMBO Rep ; 16(4): 456-66, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25648147

ABSTRACT

FOXO transcription factors are considered bona fide tumor suppressors; however, recent studies showed FOXOs are also required for tumor survival. Here, we identify FOXOs as transcriptional activators of IDH1. FOXOs promote IDH1 expression and thereby maintain the cytosolic levels of α-ketoglutarate and NADPH. In cancer cells carrying mutant IDH1, FOXOs likewise stimulate mutant IDH1 expression and maintain the levels of the oncometabolite 2-hydroxyglutarate, which stimulates cancer cell proliferation and inhibits TET enzymes and histone demethylases. Combined, our data provide a new paradigm for the paradoxical role of FOXOs in both tumor suppression and promotion.


Subject(s)
Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Isocitrate Dehydrogenase/metabolism , Transcription Factors/metabolism , Binding Sites , Cell Cycle Proteins , Cell Line , Cell Proliferation , Citric Acid Cycle/genetics , Enzyme Activation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Glutarates/metabolism , HeLa Cells , Histone Demethylases/genetics , Histone Demethylases/metabolism , Humans , Introns , Isocitrate Dehydrogenase/genetics , Ketoglutaric Acids/metabolism , NADP/metabolism , Protein Binding , Signal Transduction , Transcription Factors/genetics , Transcription, Genetic
10.
Biochem J ; 469(2): 289-98, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25990325

ABSTRACT

Activity of FOXO (forkhead box O) transcription factors is inhibited by growth factor-PI3K (phosphoinositide 3-kinase)-PKB (protein kinase B)/Akt signalling to control a variety of cellular processes including cell cycle progression. Through comparative analysis of a number of microarray datasets we identified a set of genes commonly regulated by FOXO proteins and PI3K-PKB/Akt, which includes CTDSP2 (C-terminal domain small phosphatase 2). We validated CTDSP2 as a genuine FOXO target gene and show that ectopic CTDSP2 can induce cell cycle arrest. We analysed transcriptional regulation after CTDSP2 expression and identified extensive regulation of genes involved in cell cycle progression, which depends on the phosphatase activity of CTDSP2. The most notably regulated gene is the CDK (cyclin-dependent kinase) inhibitor p21(Cip1/Waf1) and in the present study we show that p21(Cip1/Waf1) is partially responsible for the cell cycle arrest through decreasing cyclin-CDK activity. Our data suggest that CTDSP2 induces p21(Cip1/Waf1) through increasing the activity of Ras. As has been described previously, Ras induces p21(Cip1/Waf1) through p53-dependent and p53-independent pathways and indeed both p53 and MEK inhibition can mitigate the CTDSP2-induced p21(Cip1/Waf1) mRNA up-regulation. In support of Ras activation by CTDSP2, depletion of endogenous CTDSP2 results in reduced Ras activity and thus CTDSP2 seems to be part of a larger set of genes regulated by FOXO proteins, which increase growth factor signalling upon FOXO activation.


Subject(s)
Cell Cycle Checkpoints/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Forkhead Transcription Factors/metabolism , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , ras Proteins/metabolism , Animals , Cyclin-Dependent Kinase Inhibitor p21/genetics , Forkhead Transcription Factors/genetics , Gene Expression Regulation/physiology , HEK293 Cells , Humans , Mice , Mice, Knockout , NIH 3T3 Cells , Nuclear Proteins/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoprotein Phosphatases/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Transcription, Genetic/physiology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , ras Proteins/genetics
11.
Biochem J ; 460(1): 25-34, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24762137

ABSTRACT

Growth factors inactivate the FOXO (forkhead box O) transcription factors through PI3K (phosphoinositide 3-kinase) and PKB (protein kinase B). By comparing microarray data from multiple model systems, we identified HBP1 (high-mobility group-box protein 1) as a novel downstream target of this pathway. HBP1 mRNA was down-regulated by PDGF (platelet-derived growth factor), FGF (fibroblast growth factor), PI3K and PKB, whereas it was up-regulated by FOXO factors. This observation was confirmed in human and murine fibroblasts as well as in cell lines derived from leukaemia, breast adenocarcinoma and colon carcinoma. Bioinformatics analysis led to the identification of a conserved consensus FOXO-binding site in the HBP1 promoter. By luciferase activity assay and ChIP, we demonstrated that FOXO bound to this site and regulated the HBP1 promoter activity in a PI3K-dependent manner. Silencing of HBP1 by shRNA increased the proliferation of human fibroblasts in response to growth factors, suggesting that HBP1 limits cell growth. Finally, by analysing a transcriptomics dataset from The Cancer Genome Atlas, we observed that HBP1 expression was lower in breast tumours that had lost FOXO expression. In conclusion, HBP1 is a novel target of the PI3K/FOXO pathway and controls cell proliferation in response to growth factors.


Subject(s)
Down-Regulation/genetics , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , High Mobility Group Proteins/antagonists & inhibitors , High Mobility Group Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Phosphatidylinositol 3-Kinase/genetics , Proto-Oncogene Proteins c-akt/genetics , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Animals , CHO Cells , Cells, Cultured , Conserved Sequence , Cricetinae , Cricetulus , Fibroblasts/drug effects , Fibroblasts/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/biosynthesis , HEK293 Cells , High Mobility Group Proteins/biosynthesis , Humans , MCF-7 Cells , Male , Mice , NIH 3T3 Cells , Phosphatidylinositol 3-Kinase/biosynthesis , Promoter Regions, Genetic , Protein Binding/genetics , Proto-Oncogene Proteins c-akt/biosynthesis , Repressor Proteins/biosynthesis , Signal Transduction/genetics
12.
Nat Rev Immunol ; 4(11): 889-99, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15516968

ABSTRACT

It is more than a decade since the discovery of the first forkhead-box (FOX) transcription factor in the fruit fly Drosophila melanogaster. In the intervening time, there has been an explosion in the identification and characterization of members of this family of proteins. Importantly, in the past few years, it has become clear that members of the FOX family have crucial roles in various aspects of immune regulation, from lymphocyte survival to thymic development. This review focuses on FOXP3, FOXN1, FOXJ1 and members of the FOXO subfamily and their function in the immune system.


Subject(s)
DNA-Binding Proteins/immunology , Immune System/physiology , Trans-Activators/immunology , Transcription Factors/immunology , Animals , Forkhead Transcription Factors , Humans , Immune System/immunology , Lymphocyte Activation/immunology , Mice , T-Lymphocytes, Helper-Inducer/immunology , Thymus Gland/embryology , Thymus Gland/immunology
13.
J Biol Chem ; 288(30): 21729-41, 2013 Jul 26.
Article in English | MEDLINE | ID: mdl-23770673

ABSTRACT

FOXO (forkhead box O) transcription factors are tumor suppressors and increase the life spans of model organisms. Cellular stress, in particular oxidative stress caused by an increase in levels of reactive oxygen species (ROS), activates FOXOs through JNK-mediated phosphorylation. Importantly, JNK regulation of FOXO is evolutionarily conserved. Here we identified the pathway that mediates ROS-induced JNK-dependent FOXO regulation. Following increased ROS, RALA is activated by the exchange factor RLF (RalGDS-like factor), which is in complex with JIP1 (C-Jun-amino-terminal-interacting protein 1) and JNK. Active RALA consequently regulates assembly and activation of MLK3, MKK4, and JNK onto the JIP1 scaffold. Furthermore, regulation of FOXO by RALA and JIP1 is conserved in C. elegans, where both ral-1 and jip-1 depletion impairs heat shock-induced nuclear translocation of the FOXO orthologue DAF16.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Transcription Factors/metabolism , ral GTP-Binding Proteins/metabolism , Active Transport, Cell Nucleus , Adaptor Proteins, Signal Transducing/genetics , Animals , Blotting, Western , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Cycle Proteins , Cell Nucleus/metabolism , Enzyme Activation , Forkhead Transcription Factors , HEK293 Cells , Humans , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mice , Mitogen-Activated Protein Kinase 8/genetics , Mutation , NIH 3T3 Cells , RNA Interference , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , ral GTP-Binding Proteins/genetics
14.
Mol Syst Biol ; 9: 638, 2013.
Article in English | MEDLINE | ID: mdl-23340844

ABSTRACT

Forkhead box O (FOXO) transcription factors are key players in diverse cellular processes affecting tumorigenesis, stem cell maintenance and lifespan. To gain insight into the mechanisms of FOXO-regulated target gene expression, we studied genome-wide effects of FOXO3 activation. Profiling RNA polymerase II changes shows that FOXO3 regulates gene expression through transcription initiation. Correlative analysis of FOXO3 and RNA polymerase II ChIP-seq profiles demonstrates FOXO3 to act as a transcriptional activator. Furthermore, this analysis reveals a significant part of FOXO3 gene regulation proceeds through enhancer regions. FOXO3 binds to pre-existing enhancers and further activates these enhancers as shown by changes in histone acetylation and RNA polymerase II recruitment. In addition, FOXO3-mediated enhancer activation correlates with regulation of adjacent genes and pre-existence of chromatin loops between FOXO3 bound enhancers and target genes. Combined, our data elucidate how FOXOs regulate gene transcription and provide insight into mechanisms by which FOXOs can induce different gene expression programs depending on chromatin architecture.


Subject(s)
Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , RNA Polymerase II/genetics , Cell Line , Chromatin/genetics , Chromatin/ultrastructure , Chromatin Immunoprecipitation , Enhancer Elements, Genetic , Forkhead Box Protein O3 , Gene Expression Profiling , Humans , Promoter Regions, Genetic , RNA Polymerase II/metabolism
15.
Nat Commun ; 15(1): 2725, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38548751

ABSTRACT

Reactive Oxygen Species (ROS) derived from mitochondrial respiration are frequently cited as a major source of chromosomal DNA mutations that contribute to cancer development and aging. However, experimental evidence showing that ROS released by mitochondria can directly damage nuclear DNA is largely lacking. In this study, we investigated the effects of H2O2 released by mitochondria or produced at the nucleosomes using a titratable chemogenetic approach. This enabled us to precisely investigate to what extent DNA damage occurs downstream of near- and supraphysiological amounts of localized H2O2. Nuclear H2O2 gives rise to DNA damage and mutations and a subsequent p53 dependent cell cycle arrest. Mitochondrial H2O2 release shows none of these effects, even at levels that are orders of magnitude higher than what mitochondria normally produce. We conclude that H2O2 released from mitochondria is unlikely to directly damage nuclear genomic DNA, limiting its contribution to oncogenic transformation and aging.


Subject(s)
Hydrogen Peroxide , Mitochondria , Reactive Oxygen Species/metabolism , Hydrogen Peroxide/metabolism , Mitochondria/metabolism , DNA/metabolism , DNA Damage , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism
16.
Nat Cell Biol ; 8(10): 1064-73, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16964248

ABSTRACT

FOXO (Forkhead box O) transcription factors are important regulators of cellular metabolism, cell-cycle progression and cell death. FOXO activity is regulated by multiple post-translational modifications, including phosphorylation, acetylation and polyubiquitination. Here, we show that FOXO becomes monoubiquitinated in response to increased cellular oxidative stress, resulting in its re-localization to the nucleus and an increase in its transcriptional activity. Deubiquitination of FOXO requires the deubiquitinating enzyme USP7/HAUSP (herpesvirus-associated ubiquitin-specific protease), which interacts with and deubiquitinates FOXO in response to oxidative stress. Oxidative stress-induced ubiquitination and deubiquitination by USP7 do not influence FOXO protein half-life. However, USP7 does negatively regulate FOXO transcriptional activity towards endogenous promoters. Our results demonstrate a novel mechanism of FOXO regulation and indicate that USP7 has an important role in regulating FOXO-mediated stress responses.


Subject(s)
Endopeptidases/metabolism , Gene Expression Regulation/physiology , Transcription Factors/genetics , Ubiquitin/metabolism , Animals , Cell Cycle Proteins , Cells, Cultured , Forkhead Transcription Factors , Humans , Hydrogen Peroxide/pharmacology , Kidney/metabolism , Lung Neoplasms/metabolism , Mice , NIH 3T3 Cells , Oxidants/pharmacology , Oxidative Stress , Protein Processing, Post-Translational , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Transfection , Ubiquitin Thiolesterase , Ubiquitin-Specific Peptidase 7 , Ubiquitin-Specific Proteases
17.
Free Radic Biol Med ; 206: 134-142, 2023 09.
Article in English | MEDLINE | ID: mdl-37392950

ABSTRACT

Reactive Oxygen Species (ROS) in the form of H2O2 can act both as physiological signaling molecules as well as damaging agents, depending on their concentration and localization. The downstream biological effects of H2O2 were often studied making use of exogenously added H2O2, generally as a bolus and at supraphysiological levels. But this does not mimic the continuous, low levels of intracellular H2O2 production by for instance mitochondrial respiration. The enzyme d-Amino Acid Oxidase (DAAO) catalyzes H2O2 formation using d-amino acids, which are absent from culture media, as a substrate. Ectopic expression of DAAO has recently been used in several studies to produce inducible and titratable intracellular H2O2. However, a method to directly quantify the amount of H2O2 produced by DAAO has been lacking, making it difficult to assess whether observed phenotypes are the result of physiological or artificially high levels of H2O2. Here we describe a simple assay to directly quantify DAAO activity by measuring the oxygen consumed during H2O2 production. The oxygen consumption rate (OCR) of DAAO can directly be compared to the basal mitochondrial respiration in the same assay, to estimate whether the ensuing level of H2O2 production is within the range of physiological mitochondrial ROS production. In the tested monoclonal RPE1-hTERT cells, addition of 5 mM d-Ala to the culture media amounts to a DAAO-dependent OCR that surpasses ∼5% of the OCR that stems from basal mitochondrial respiration and hence produces supra-physiological levels of H2O2. We show that the assay can also be used to select clones that express differentially localized DAAO with the same absolute level of H2O2 production to be able to discriminate the effects of H2O2 production at different subcellular locations from differences in total oxidative burden. This method therefore greatly improves the interpretation and applicability of DAAO-based models, thereby moving the redox biology field forward.


Subject(s)
Amino Acids , Hydrogen Peroxide , Humans , Hydrogen Peroxide/metabolism , Reactive Oxygen Species/metabolism , Amino Acids/metabolism , Oxygen Consumption , Oxygen
18.
Cell Rep ; 42(6): 112583, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37267106

ABSTRACT

Upon antigen-specific T cell receptor (TCR) engagement, human CD4+ T cells proliferate and differentiate, a process associated with rapid transcriptional changes and metabolic reprogramming. Here, we show that the generation of extramitochondrial pyruvate is an important step for acetyl-CoA production and subsequent H3K27ac-mediated remodeling of histone acetylation. Histone modification, transcriptomic, and carbon tracing analyses of pyruvate dehydrogenase (PDH)-deficient T cells show PDH-dependent acetyl-CoA generation as a rate-limiting step during T activation. Furthermore, T cell activation results in the nuclear translocation of PDH and its association with both the p300 acetyltransferase and histone H3K27ac. These data support the tight integration of metabolic and histone-modifying enzymes, allowing metabolic reprogramming to fuel CD4+ T cell activation. Targeting this pathway may provide a therapeutic approach to specifically regulate antigen-driven T cell activation.


Subject(s)
Chromatin Assembly and Disassembly , Histones , Humans , Histones/metabolism , Acetyl Coenzyme A/metabolism , CD4-Positive T-Lymphocytes/metabolism
20.
Biochim Biophys Acta ; 1813(11): 1926-37, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21539865

ABSTRACT

Aging is characterized by the general decline in tissue and body function and the increased susceptibility to age-related pathologies, such as cancer. To maintain optimal tissue and body function, organisms have developed complex mechanisms for tissue homeostasis. Importantly, it is becoming apparent that these same mechanisms when deregulated also result in the development of age-related disease. The build in fail safe mechanisms of homeostasis, which prevent skewing toward disease, themselves contribute to aspects of aging. Thus, longevity is limited by an intrinsic trade-off between optimal tissue function and disease. Consequently, aging and age-related diseases, such as cancer and diabetes are driven by the same genetic determinants. Illustrative in this respect is the insulin/IGF-1 signaling pathway acting through PI3K/PKB and FOXO. Loss of PKB signaling contributes to diabetes, whereas gain of function of PKB drives cancer. Enhanced FOXO activity, at least in model organism contributes to extended lifespan and acts as a tumor suppressive mechanism. Here, we focus on the linkage between PKB and FOXO as a central switch in contributing to tissue homeostasis and age-related diseases in particular cancer. This article is part of a Special Issue entitled: P13K-AKT-FoxO axis in cancer and aging.


Subject(s)
Aging/metabolism , Forkhead Transcription Factors/metabolism , Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Cycle Proteins/metabolism , Forkhead Box Protein O1 , Humans , Models, Biological , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL