Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Cell ; 132(2): 233-46, 2008 Jan 25.
Article in English | MEDLINE | ID: mdl-18243099

ABSTRACT

Maintenance of chromosomal stability relies on coordination between various processes that are critical for proper chromosome segregation in mitosis. Here we show that monopolar spindle 1 (Mps1) kinase, which is essential for the mitotic checkpoint, also controls correction of improper chromosome attachments. We report that Borealin/DasraB, a member of the complex that regulates the Aurora B kinase, is directly phosphorylated by Mps1 on residues that are crucial for Aurora B activity and chromosome alignment. As a result, cells lacking Mps1 kinase activity fail to efficiently align chromosomes due to impaired Aurora B function at centromeres, leaving improper attachments uncorrected. Strikingly, Borealin/DasraB bearing phosphomimetic mutations restores Aurora B activity and alignment in Mps1-depleted cells. Mps1 thus coordinates attachment error correction and checkpoint signaling, two crucial responses to unproductive chromosome attachments.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , Chromosomes, Human/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Alleles , Aurora Kinase B , Aurora Kinases , Cell Cycle Proteins/genetics , Cell Line, Tumor , Enzyme Activation , HeLa Cells , Humans , Kinetochores/metabolism , Microtubules/metabolism , Mutation , Phosphorylation , Plasmids , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases , RNA, Small Interfering/metabolism , Recombinant Proteins/metabolism , Spindle Apparatus/metabolism , Transfection
2.
Immunity ; 39(2): 259-71, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23973222

ABSTRACT

Stable Foxp3 expression is required for the development of functional regulatory T (Treg) cells. Here, we demonstrate that the expression of the transcription factor Foxp3 can be regulated through the polyubiquitination of multiple lysine residues, resulting in proteasome-mediated degradation. Expression of the deubiquitinase (DUB) USP7 was found to be upregulated and active in Treg cells, being associated with Foxp3 in the nucleus. Ectopic expression of USP7 decreased Foxp3 polyubiquitination and increased Foxp3 expression. Conversely, either treatment with DUB inhibitor or USP7 knockdown decreased endogenous Foxp3 protein expression and decreased Treg-cell-mediated suppression in vitro. Furthermore, in a murine adoptive-transfer-induced colitis model, either inhibition of DUB activity or USP7 knockdown in Treg cells abrogated their ability to resolve inflammation in vivo. Our data reveal a molecular mechanism in which rapid temporal control of Foxp3 expression in Treg cells can be regulated by USP7, thereby modulating Treg cell numbers and function.


Subject(s)
Colitis/immunology , Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Ubiquitin Thiolesterase/metabolism , Adoptive Transfer , Animals , Cell Differentiation/immunology , Cell Line , DNA-Binding Proteins/genetics , Disease Models, Animal , Endopeptidases/metabolism , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Immediate-Early Proteins/antagonists & inhibitors , Immediate-Early Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , RNA Interference , RNA, Small Interfering , Ubiquitin Thiolesterase/genetics , Ubiquitin-Specific Peptidase 7 , Ubiquitination
3.
Mol Cell ; 41(4): 445-57, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21329882

ABSTRACT

Expression of the Myc oncoprotein is downregulated in response to stress signals to allow cells to cease proliferation and escape apoptosis, but the mechanisms involved in this process are poorly understood. Cell cycle arrest in response to DNA damage requires downregulation of Myc via a p53-independent signaling pathway. Here we have used siRNA screening of the human kinome to identify MAPKAPK5 (MK5, PRAK) as a negative regulator of Myc expression. MK5 regulates translation of Myc, since it is required for expression of miR-34b and miR-34c that bind to the 3'UTR of MYC. MK5 activates miR-34b/c expression via phosphorylation of FoxO3a, thereby promoting nuclear localization of FoxO3a and enabling it to induce miR-34b/c expression and arrest proliferation. Expression of MK5 in turn is directly activated by Myc, forming a negative feedback loop. MK5 is downregulated in colon carcinomas, arguing that this feedback loop is disrupted during colorectal tumorigenesis.


Subject(s)
Colorectal Neoplasms/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/genetics , Cell Line, Tumor , Colorectal Neoplasms/enzymology , Down-Regulation , Feedback, Physiological , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , HCT116 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
4.
Blood ; 125(11): 1782-92, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25568349

ABSTRACT

C/EBPε, a member of the CCAAT/enhancer binding protein (C/EBP) family of transcription factors, is exclusively expressed in myeloid cells and regulates transition from the promyelocytic stage to the myelocytic stage of neutrophil development, being indispensable for secondary and tertiary granule formation. Knowledge concerning the functional role of C/EBPε posttranslational modifications is limited to studies concerning phosphorylation and sumoylation. In the current study, using ectopic expression and ex vivo differentiation of CD34(+) hematopoietic progenitor cells, we demonstrate that C/EBPε is acetylated, which was confirmed by mass spectrometry analysis, identifying 4 acetylated lysines in 3 distinct functional domains. Regulation of C/EBPε acetylation levels by the p300 acetyltransferase and the sirtuin 1 deacetylase controls transcriptional activity, which can at least in part be explained by modulation of DNA binding. During neutrophil development, acetylation of lysines 121 and 198 were found to be crucial for terminal neutrophil differentiation and the expression of neutrophil-specific granule proteins, including lactoferrin and collagenase. Taken together, our data illustrate a critical role for acetylation in the functional regulation of C/EBPε activity during terminal neutrophil development.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Neutrophils/cytology , Neutrophils/metabolism , Acetylation , Animals , CCAAT-Enhancer-Binding Proteins/chemistry , CCAAT-Enhancer-Binding Proteins/genetics , COS Cells , Cell Differentiation , Cell Line, Tumor , Chlorocebus aethiops , Collagenases/metabolism , HL-60 Cells , Humans , Lactoferrin/metabolism , Lysine/chemistry , Myelopoiesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sirtuin 1/metabolism , Transcription, Genetic , p300-CBP Transcription Factors/metabolism
5.
J Immunol ; 194(4): 1856-66, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25582855

ABSTRACT

During secondary immune responses, Ab-opsonized bacteria are efficiently taken up via FcRs by dendritic cells. We now demonstrate that this process induces cross-talk between FcRs and TLRs, which results in synergistic release of several inflammatory cytokines, as well as altered lipid metabolite profiles. This altered inflammatory profile redirects Th1 polarization toward Th17 cell responses. Interestingly, GM-CSF-producing Th cells were synergistically evoked as well, which suggests the onset of polyfunctional Th17 cells. Synergistic cytokine release was dependent on activation via MyD88 and ITAM signaling pathways through TLRs and FcRs, respectively. Cytokine regulation occurred via transcription-dependent mechanisms for TNF-α and IL-23 and posttranscriptional mechanisms for caspase-1-dependent release of IL-1ß. Furthermore, cross-talk between TLRs and FcRs was not restricted to dendritic cells. In conclusion, our results support that bacteria alone initiate fundamentally different immune responses compared with Ab-opsonized bacteria through the combined action of two classes of receptors and, ultimately, may refine new therapies for inflammatory diseases.


Subject(s)
Dendritic Cells/immunology , Receptor Cross-Talk/immunology , Receptors, Fc/immunology , T-Lymphocytes, Helper-Inducer/immunology , Toll-Like Receptors/immunology , Antibodies, Bacterial/immunology , Blotting, Western , Cell Differentiation/immunology , Cell Separation , Enterobacteriaceae Infections/immunology , Enzyme-Linked Immunosorbent Assay , Escherichia coli/immunology , Flow Cytometry , Humans , Immunologic Memory/immunology , Inflammation/immunology , Lymphocyte Activation/immunology , Phenotype , Real-Time Polymerase Chain Reaction , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology
6.
Mol Syst Biol ; 9: 679, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23820781

ABSTRACT

Mutations in the daf-2 gene of the conserved Insulin/Insulin-like Growth Factor (IGF-1) pathway double the lifespan of the nematode Caenorhabditis elegans. This phenotype is completely suppressed by deletion of Forkhead transcription factor daf-16. To uncover regulatory mechanisms coordinating this extension of life, we employed a quantitative proteomics strategy with daf-2 mutants in comparison with N2 and daf-16; daf-2 double mutants. This revealed a remarkable longevity-specific decrease in proteins involved in mRNA processing and transport, the translational machinery, and protein metabolism. Correspondingly, the daf-2 mutants display lower amounts of mRNA and 20S proteasome activity, despite maintaining total protein levels equal to that observed in wild types. Polyribosome profiling in the daf-2 and daf-16;daf-2 double mutants confirmed a daf-16-dependent reduction in overall translation, a phenotype reminiscent of Dietary Restriction-mediated longevity, which was independent of germline activity. RNA interference (RNAi)-mediated knockdown of proteins identified by our approach resulted in modified C. elegans lifespan confirming the importance of these processes in Insulin/IGF-1-mediated longevity. Together, the results demonstrate a role for the metabolism of proteins in the Insulin/IGF-1-mediated extension of life.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Insulin-Like Growth Factor I/genetics , Insulin/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , Receptor, Insulin/genetics , Transcription Factors/genetics , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/metabolism , Forkhead Transcription Factors , Gene Expression Regulation , Genotype , Insulin-Like Growth Factor I/metabolism , Longevity/genetics , Mutation , Phenotype , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Receptor, Insulin/antagonists & inhibitors , Receptor, Insulin/metabolism , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism
7.
Biochem J ; 451(1): 45-53, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23320500

ABSTRACT

Reversible phosphorylation is a widespread molecular mechanism to regulate the function of cellular proteins, including transcription factors. Phosphorylation of the nuclear receptor PPARγ (peroxisome-proliferator-activated receptor γ) at two conserved serine residue (Ser(112) and Ser(273)) results in an altered transcriptional activity of this transcription factor. So far, only a very limited number of cellular enzymatic activities has been described which can dephosphorylate nuclear receptors. In the present study we used immunoprecipitation assays coupled to tandem MS analysis to identify novel PPARγ-regulating proteins. We identified the serine/threonine phosphatase PPM1B [PP (protein phosphatase), Mg(2+)/Mn(2+) dependent, 1B; also known as PP2Cß] as a novel PPARγ-interacting protein. Endogenous PPM1B protein is localized in the nucleus of mature 3T3-L1 adipocytes where it can bind to PPARγ. Furthermore we show that PPM1B can directly dephosphorylate PPARγ, both in intact cells and in vitro. In addition PPM1B increases PPARγ-mediated transcription via dephosphorylation of Ser(112). Finally, we show that knockdown of PPM1B in 3T3-L1 adipocytes blunts the expression of some PPARγ target genes while leaving others unaltered. These findings qualify the phosphatase PPM1B as a novel selective modulator of PPARγ activity.


Subject(s)
Adipocytes/metabolism , Cell Nucleus/metabolism , PPAR gamma/metabolism , Phosphoprotein Phosphatases/metabolism , Transcription, Genetic/physiology , 3T3-L1 Cells , Active Transport, Cell Nucleus/physiology , Adipocytes/cytology , Animals , Cell Line, Tumor , Cell Nucleus/genetics , Humans , Magnesium/metabolism , Manganese/metabolism , Mice , PPAR gamma/genetics , Phosphoprotein Phosphatases/genetics , Phosphorylation/physiology , Protein Phosphatase 2C
8.
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
9.
J Exp Med ; 203(6): 1419-25, 2006 Jun 12.
Article in English | MEDLINE | ID: mdl-16754721

ABSTRACT

Collagens are the most abundant proteins in the human body, important in maintenance of tissue structure and hemostasis. Here we report that collagens are high affinity ligands for the broadly expressed inhibitory leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1). The interaction is dependent on the conserved Gly-Pro-Hyp collagen repeats. Antibody cross-linking of LAIR-1 is known to inhibit immune cell function in vitro. We now show that collagens are functional ligands for LAIR-1 and directly inhibit immune cell activation in vitro. Thus far, all documented ligands for immune inhibitory receptors are membrane molecules, implying a regulatory role in cell-cell interaction. Our data reveal a novel mechanism of peripheral immune regulation by inhibitory immune receptors binding to extracellular matrix collagens.


Subject(s)
Collagen/immunology , Receptors, Immunologic/physiology , Amino Acid Sequence , Collagen/metabolism , Humans , K562 Cells , Ligands , Oligopeptides/chemistry , Oligopeptides/metabolism , Receptors, Collagen/immunology , Receptors, Collagen/physiology , Receptors, Immunologic/antagonists & inhibitors , Transfection
10.
Blood ; 115(5): 965-74, 2010 Feb 04.
Article in English | MEDLINE | ID: mdl-19996091

ABSTRACT

Regulatory T cells (Tregs) are a specific subset of lymphocytes that are critical for the maintenance of self-tolerance. Expression levels of the transcription factor Foxp3 have been causally associated with Treg differentiation and function. Recent studies show that Foxp3 can also be transiently expressed in effector T cells; however, stable Foxp3 expression is required for development of a functional Treg suppressor phenotype. Here, we demonstrate that Foxp3 is acetylated, and this can be reciprocally regulated by the histone acetyltransferase p300 and the histone deacetylase SIRT1. Hyperacetylation of Foxp3 prevented polyubiquitination and proteasomal degradation, therefore dramatically increasing stable Foxp3 protein levels. Moreover, using mouse splenocytes, human peripheral blood mononuclear cells, T cell clones, and skin-derived T cells, we demonstrate that treatment with histone deacetylase inhibitors resulted in significantly increased numbers of functional Treg cells. Taken together, our data demonstrate that modulation of the acetylation state of Foxp3 provides a novel molecular mechanism for assuring rapid temporal control of Foxp3 levels in T cells, thereby regulating Treg numbers and functionality. Manipulating Foxp3 acetylation levels could therefore provide a new therapeutic strategy to control inappropriate (auto)immune responses.


Subject(s)
Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation/drug effects , Animals , Cell Line , Cells, Cultured , Flow Cytometry , Forkhead Transcription Factors/genetics , Histone Deacetylase Inhibitors/pharmacology , Humans , Immunoblotting , Immunoprecipitation , Mice , Mice, Inbred C57BL , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Sirtuin 1/genetics , Sirtuin 1/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/cytology , Transfection , p300-CBP Transcription Factors/genetics
11.
Nat Chem Biol ; 5(9): 664-72, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19648934

ABSTRACT

Cellular damage invoked by reactive oxygen species plays a key role in the pathobiology of cancer and aging. Forkhead box class O (FoxO) transcription factors are involved in various cellular processes including cell cycle regulation, apoptosis and resistance to reactive oxygen species, and studies in animal models have shown that these transcription factors are of vital importance in tumor suppression, stem cell maintenance and lifespan extension. Here we report that the activity of FoxO in human cells is directly regulated by the cellular redox state through a unique mechanism in signal transduction. We show that reactive oxygen species induce the formation of cysteine-thiol disulfide-dependent complexes of FoxO and the p300/CBP acetyltransferase, and that modulation of FoxO biological activity by p300/CBP-mediated acetylation is fully dependent on the formation of this redox-dependent complex. These findings directly link cellular redox status to the activity of the longevity protein FoxO.


Subject(s)
Cysteine/metabolism , Reactive Oxygen Species/metabolism , Transcription Factors/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation , Animals , Cell Cycle Proteins , Cell Line , Cell Survival , Cysteine/genetics , Forkhead Transcription Factors , Humans , Lysine/genetics , Lysine/metabolism , Mice , Mutation , Oxidation-Reduction , Peroxides/pharmacology , Signal Transduction , Thioredoxins/pharmacology , Transcription Factors/genetics , p300-CBP Transcription Factors/genetics
12.
FASEB J ; 24(11): 4271-80, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20570964

ABSTRACT

In this study, we searched for proteins regulating the tumor suppressor and life-span regulator FOXO4. Through an unbiased tandem-affinity purification strategy combined with mass spectrometry, we identified the heterodimer Ku70/Ku80 (Ku), a DNA double-strand break repair component. Using biochemical interaction studies, we found Ku70 to be necessary and sufficient for the interaction. FOXO4 mediates its tumor-suppressive function in part through transcriptional regulation of the cell cycle arrest p27(kip1) gene. Immunoblotting, luciferase reporter assays, and flow cytometry showed that Ku70 inhibited FOXO4-mediated p27(kip1) transcription and cell cycle arrest induction by >40%. In contrast, Ku70 RNAi but not control RNAi significantly increased p27(kip1) transcription. In addition, in contrast to wild-type mouse embryonic stem (ES) cells, Ku70(-/-) ES cells showed significantly increased FOXO activity, which was rescued by Ku70 reexpression. Immunofluorescence studies demonstrated that Ku70 sequestered FOXO4 in the nucleus. Interestingly, the Ku70-FOXO4 interaction stoichiometry followed a nonlinear dose-response curve by hydrogen peroxide-generated oxidative stress. Low levels of oxidative stress increased interaction stoichiometry up to 75%, peaking at 50 µM, after which dissociation occurred. Because the Ku70 ortholog in the roundworm Caenorhabditis elegans was shown to regulate life span involving C. elegans FOXO, our findings suggest a conserved critical Ku70 role for FOXO function toward coordination of a survival program, regulated by the magnitude of oxidative damage.


Subject(s)
Antigens, Nuclear/metabolism , DNA-Binding Proteins/metabolism , Forkhead Transcription Factors/metabolism , Multiprotein Complexes , Stress, Physiological , Transcription Factors/metabolism , Animals , Antigens, Nuclear/genetics , Cell Cycle/physiology , Cell Cycle Proteins , Cell Line , DNA-Binding Proteins/genetics , Forkhead Transcription Factors/genetics , Gene Knockout Techniques , HEK293 Cells , HeLa Cells , Humans , Ku Autoantigen , Mice , Multiprotein Complexes/pharmacology , Oxidative Stress/drug effects , Tandem Mass Spectrometry
13.
Nucleic Acids Res ; 35(7): 2428-39, 2007.
Article in English | MEDLINE | ID: mdl-17392337

ABSTRACT

Efficient transcription is linked to modification of chromatin. For instance, tri-methylation of lysine 4 on histone H3 (H3K4) strongly correlates with transcriptional activity and is regulated by the Bur1/2 kinase complex. We found that the evolutionarily conserved Ccr4-Not complex is involved in establishing H3K4 tri-methylation in Saccharomyces cerevisiae. We observed synthetic lethal interactions of Ccr4-Not components with BUR1 and BUR2. Further analysis indicated that the genes encoding the Not-proteins are essential for efficient regulation of H3K4me3, but not H3K4me1/2, H3K36me2 or H3K79me2/3 levels. Moreover, regulation of H3K4me3 levels by NOT4 is independent of defects in RNA polymerase II loading. We found NOT4 to be important for ubiquitylation of histone H2B via recruitment of the PAF complex, but not for recruitment or activation of the Bur1/2 complex. These results suggest a mechanism in which the Ccr4-Not complex functions parallel to or downstream of the Bur1/2 kinase to facilitate H3K4me3 via PAF complex recruitment.


Subject(s)
Histones/metabolism , Ribonucleases/physiology , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/enzymology , Ubiquitin-Protein Ligases/physiology , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cyclins/genetics , Cyclins/metabolism , DNA-Binding Proteins/metabolism , DNA-Directed RNA Polymerases/metabolism , Gene Deletion , Gene Expression Regulation, Fungal , Histone-Lysine N-Methyltransferase/metabolism , Histones/chemistry , Lysine/metabolism , Methylation , Nuclear Proteins/metabolism , Repressor Proteins , Ribonucleases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics
14.
Endocrinology ; 149(4): 1840-9, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18096664

ABSTRACT

The transcription factor peroxisome proliferator-activated receptor gamma (PPARgamma) plays a key role in the regulation of lipid and glucose metabolism in adipocytes, by regulating their differentiation, maintenance, and function. The transcriptional activity of PPARgamma is dictated by the set of proteins with which this nuclear receptor interacts under specific conditions. Here we identify the HIV-1 Tat-interacting protein 60 (Tip60) as a novel positive regulator of PPARgamma transcriptional activity. Using tandem mass spectrometry, we found that PPARgamma and the acetyltransferase Tip60 interact in cells, and through use of chimeric proteins, we established that coactivation by Tip60 critically depends on the N-terminal activation function 1 of PPARgamma, a domain involved in isotype-specific gene expression and adipogenesis. Chromatin immunoprecipitation experiments showed that the endogenous Tip60 protein is recruited to PPARgamma target genes in mature 3T3-L1 adipocytes but not in preadipocytes, indicating that Tip60 requires PPARgamma for its recruitment to PPARgamma target genes. Importantly, we show that in common with disruption of PPARgamma function, small interfering RNA-mediated reduction of Tip60 protein impairs differentiation of 3T3-L1 preadipocytes. Taken together, these findings qualify the acetyltransferase Tip60 as a novel adipogenic factor.


Subject(s)
Adipogenesis , Histone Acetyltransferases/physiology , PPAR gamma/physiology , 3T3-L1 Cells , Amino Acid Sequence , Animals , Cell Line, Tumor , Humans , Lysine Acetyltransferase 5 , Mice , Molecular Sequence Data , PPAR gamma/chemistry , Protein Structure, Tertiary , Transcription, Genetic
15.
Fertil Steril ; 107(3): 699-706.e6, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28259259

ABSTRACT

OBJECTIVE: To identify metabolites that are associated with and predict the presence of endometriosis. DESIGN: Metabolomics study using state-of-the-art mass spectrometry approaches. SETTING: University hospital and universities. PATIENT(S): Twenty-five women with laparoscopically confirmed endometriosis (cases) and 19 women with laparoscopically documented absence of endometriosis (controls). None of the women included in this study had received oral contraception or GnRH agonists for a minimum of 1 month before blood collection. INTERVENTION(S): Plasma collection. MAIN OUTCOME MEASURE(S): Metabolite profiles were generated and interrogated using multiple mass spectrometry methods, that is, high performance liquid chromatography coupled with negative mode electrospray ionization tandem mass spectrometry, UPLC-MS/MS, and ultra performance liquid chromatography-electroSpray ionization-quadrupole time-of-flight (UPLC-ESI-Q-TOF). Metabolite groups investigated included phospholipids, glycerophospholipids, ether-phospholipids, cholesterol-esters, triacylglycerol, sphingolipids, free fatty acids, steroids, eicosanoids, and acylcarnitines. RESULT(S): A panel of acylcarnitines predicted the presence of endometriosis with 88.9% specificity and 81.5% sensitivity in human plasma, with a positive predictive value of 75%. However, due to data limitations the outcome of the receiver operating characteristic curve analysis was not significant. CONCLUSION(S): A diagnostic model based on acylcarnitines has the potential to predict the presence and stage of endometriosis.


Subject(s)
Carnitine/analogs & derivatives , Endometriosis/blood , Lipids/blood , Metabolomics , Adult , Area Under Curve , Belgium , Biomarkers/blood , Carnitine/blood , Case-Control Studies , Chromatography, High Pressure Liquid , Endometriosis/diagnosis , Female , Hospitals, University , Humans , Laparoscopy , Metabolomics/methods , Pilot Projects , Predictive Value of Tests , ROC Curve , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
16.
Trends Mol Med ; 9(11): 464-7, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14604822

ABSTRACT

The FoxO transcription factors (FoxO1a, 3a and 4) comprise a small subfamily of the Forkhead transcription factor family. An increasing number of studies has provided genetic evidence showing that Forkhead transcription factors control crucial steps in embryogenesis and are essential for the development of all germ layers and organs (for a recent review, see Ref. ). A recent study by Castrillon et al. has now added a function for FoxO3a in the control of follicular development.


Subject(s)
Aging/physiology , DNA-Binding Proteins/physiology , Fertility/physiology , Ovarian Follicle/physiology , Transcription Factors/physiology , Animals , Cell Membrane/physiology , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors , Humans
17.
Sci Rep ; 5: 10944, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-26091097

ABSTRACT

Regular exercise has emerged as one of the best therapeutic strategies to prevent and treat type-2-diabetes. Exercise-induced changes in the muscle secretome, consisting of myokines and metabolites, may underlie the inter-organ communication between muscle and other organs. To investigate this crosstalk, we developed an in vitro system in which mouse C2C12 myotubes underwent electric pulse stimulation (EPS) to induce contraction. Subsequently the effects of EPS-conditioned media (EPS-CM) on hepatocytes were investigated. Here, we demonstrate that EPS-CM induces Metallothionein 1/2 and Slc30a2 gene expression and reduces Cyp2a3 gene expression in rat hepatocytes. When testing EPS-CM that was generated in the absence of C2C12 myotubes (non-cell EPS-CM) no decrease in Cyp2a3 expression was detected. However, similar inductions in hepatic Mt1/2 and Slc30a2 expression were observed. Non-cell EPS-CM were also applied to C2C12 myotubes and compared to C2C12 myotubes that underwent EPS: here changes in AMPK phosphorylation and myokine secretion largely depended on EPS-induced contraction. Taken together, these findings indicate that EPS can alter C2C12 myotube function and thereby affect gene expression in cells subjected to EPS-CM (Cyp2a3). However, EPS can also generate non-cell-mediated changes in cell culture media, which can affect gene expression in cells subjected to EPS-CM too. While EPS clearly represents a valuable tool in exercise research, care should be taken in experimental design to control for non-cell-mediated effects.


Subject(s)
Gene Expression Regulation , Muscle Contraction , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/metabolism , Animals , Cell Line , Electric Stimulation , Mice , Muscle Fibers, Skeletal/cytology , Rats
18.
Cell Rep ; 10(3): 339-345, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25600869

ABSTRACT

The biogenesis of ribosomes and their coordination of protein translation consume an enormous amount of cellular energy. As such, it has been established that the inhibition of either process can extend eukaryotic lifespan. Here, we used next-generation sequencing to compare ribosome-associated RNAs from normal strains of Caenorhabditis elegans to those carrying the life-extending daf-2 mutation. We found a long noncoding RNA (lncRNA), transcribed telomeric sequence 1 (tts-1), on ribosomes of the daf-2 mutant. Depleting tts-1 in daf-2 mutants increases ribosome levels and significantly shortens their extended lifespan. We find tts-1 is also required for the longer lifespan of the mitochondrial clk-1 mutants but not the feeding-defective eat-2 mutants. In line with this, the clk-1 mutants express more tts-1 and fewer ribosomes than the eat-2 mutants. Our results suggest that the expression of tts-1 functions in different longevity pathways to reduce ribosome levels in a way that promotes life extension.

19.
PLoS One ; 9(5): e98438, 2014.
Article in English | MEDLINE | ID: mdl-24875183

ABSTRACT

The Forkhead transcription factor FOXA2 plays a fundamental role in controlling metabolic homeostasis in the liver during fasting. The precise molecular regulation of FOXA2 in response to nutrients is not fully understood. Here, we studied whether FOXA2 could be controlled at a post-translational level by acetylation. By means of LC-MS/MS analyses, we identified five acetylated residues in FOXA2. Sirtuin family member SIRT1 was found to interact with and deacetylate FOXA2, the latter process being dependent on the NAD+-binding catalytic site of SIRT1. Deacetylation by SIRT1 reduced protein stability of FOXA2 by targeting it towards proteasomal degradation, and inhibited transcription from the FOXA2-driven G6pase and CPT1a promoters. While mutation of the five identified acetylated residues weakly affected protein acetylation and stability, mutation of at least seven additional lysine residues was required to abolish acetylation and reduce protein levels of FOXA2. The importance of acetylation of FOXA2 became apparent upon changes in nutrient levels. The interaction of FOXA2 and SIRT1 was strongly reduced upon nutrient withdrawal in cell culture, while enhanced Foxa2 acetylation levels were observed in murine liver in vivo after starvation for 36 hours. Collectively, this study demonstrates that SIRT1 controls the acetylation level of FOXA2 in a nutrient-dependent manner and in times of nutrient shortage the interaction between SIRT1 and FOXA2 is reduced. As a result, FOXA2 is protected from degradation by enhanced acetylation, hence enabling the FOXA2 transcriptional program to be executed to maintain metabolic homeostasis.


Subject(s)
Animal Feed , Hepatocyte Nuclear Factor 3-beta/metabolism , Sirtuin 1/metabolism , Acetylation , Active Transport, Cell Nucleus , Animals , Catalysis , Cell Line , Gene Expression Regulation , Hepatocyte Nuclear Factor 3-beta/genetics , Humans , Liver/metabolism , Mice , Protein Binding , Protein Processing, Post-Translational , Protein Transport , Starvation , Transcription, Genetic
20.
Obesity (Silver Spring) ; 22(5): 1296-308, 2014 May.
Article in English | MEDLINE | ID: mdl-24339422

ABSTRACT

OBJECTIVE: Extracellular vesicles (EVs) released by human adipocytes or adipose tissue (AT)-explants play a role in the paracrine interaction between adipocytes and macrophages, a key mechanism in AT inflammation, leading to metabolic complications like insulin resistance (IR) were determined. METHODS: EVs released from in vitro differentiated adipocytes and AT-explants ex vivo were characterized by electron microscopy, Western blot, multiplex adipokine-profiling, and quantified by flow cytometry. Primary monocytes were stimulated with EVs from adipocytes, subcutaneous (SCAT) or omental-derived AT (OAT), and phenotyped. Macrophage supernatant was subsequently used to assess the effect on insulin signaling in adipocytes. RESULTS: Adipocyte and AT-derived EVs differentiated monocytes into macrophages characteristic of human adipose tissue macrophages (ATM), defined by release of both pro- and anti-inflammatory cytokines. The adiponectin-positive subset of AT-derived EVs, presumably representing adipocyte-derived EVs, induced a more pronounced ATM-phenotype than the adiponectin-negative AT-EVs. This effect was more evident for OAT-EVs versus SCAT-EVs. Furthermore, supernatant of macrophages pre-stimulated with AT-EVs interfered with insulin signaling in human adipocytes. Finally, the number of OAT-derived EVs correlated positively with patients HOMA-IR. CONCLUSIONS: A possible role for human AT-EVs in a reciprocal pro-inflammatory loop between adipocytes and macrophages, with the potential to aggravate local and systemic IR was demonstrated.


Subject(s)
Adipocytes/ultrastructure , Cell Communication , Macrophages/metabolism , Adipokines/metabolism , Adiponectin/metabolism , Adipose Tissue/metabolism , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Humans , Immunologic Factors/pharmacology , Inflammation/metabolism , Insulin/metabolism , Insulin Resistance , Monocytes/cytology , Monocytes/metabolism , Obesity/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL