Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(14)2022 Jul 11.
Article in English | MEDLINE | ID: mdl-35887017

ABSTRACT

The Jumonji-C (JmjC) family of lysine demethylases (KDMs) (JMJC-KDMs) plays an essential role in controlling gene expression and chromatin structure. In most cases, their function has been attributed to the demethylase activity. However, accumulating evidence demonstrates that these proteins play roles distinct from histone demethylation. This raises the possibility that they might share domains that contribute to their functional outcome. Here, we show that the JMJC-KDMs contain low-complexity domains and intrinsically disordered regions (IDR), which in some cases reached 70% of the protein. Our data revealed that plant homeodomain finger protein (PHF2), KDM2A, and KDM4B cluster by phase separation. Moreover, our molecular analysis implies that PHF2 IDR contributes to transcription regulation. These data suggest that clustering via phase separation is a common feature that JMJC-KDMs utilize to facilitate their functional responses. Our study uncovers a novel potential function for the JMJC-KDM family that sheds light on the mechanisms to achieve the competent concentration of molecules in time and space within the cell nucleus.


Subject(s)
Histone Demethylases , Jumonji Domain-Containing Histone Demethylases , Cell Nucleus/metabolism , Demethylation , Histone Demethylases/genetics , Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Lysine/metabolism
2.
Nat Commun ; 13(1): 3263, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35672304

ABSTRACT

Enhancers are key regulatory elements that govern gene expression programs in response to developmental signals. However, how multiple enhancers arrange in the 3D-space to control the activation of a specific promoter remains unclear. To address this question, we exploited our previously characterized TGFß-response model, the neural stem cells, focusing on a ~374 kb locus where enhancers abound. Our 4C-seq experiments reveal that the TGFß pathway drives the assembly of an enhancer-cluster and precise gene activation. We discover that the TGFß pathway coactivator JMJD3 is essential to maintain these structures. Using live-cell imaging techniques, we demonstrate that an intrinsically disordered region contained in JMJD3 is involved in the formation of phase-separated biomolecular condensates, which are found in the enhancer-cluster. Overall, in this work we uncover novel functions for the coactivator JMJD3, and we shed light on the relationships between the 3D-conformation of the chromatin and the TGFß-driven response during mammalian neurogenesis.


Subject(s)
Neural Stem Cells , Transforming Growth Factor beta , Animals , Chromatin/genetics , Chromatin/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression , Genome , Mammals/genetics , Neural Stem Cells/metabolism , Transcriptional Activation/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
3.
Development ; 148(12)2021 06 15.
Article in English | MEDLINE | ID: mdl-34081130

ABSTRACT

Epigenetic factors have been shown to play a crucial role in X-linked intellectual disability (XLID). Here, we investigate the contribution of the XLID-associated histone demethylase PHF8 to astrocyte differentiation and function. Using genome-wide analyses and biochemical assays in mouse astrocytic cultures, we reveal a regulatory crosstalk between PHF8 and the Notch signaling pathway that balances the expression of the master astrocytic gene Nfia. Moreover, PHF8 regulates key synaptic genes in astrocytes by maintaining low levels of H4K20me3. Accordingly, astrocytic-PHF8 depletion has a striking effect on neuronal synapse formation and maturation in vitro. These data reveal that PHF8 is crucial in astrocyte development to maintain chromatin homeostasis and limit heterochromatin formation at synaptogenic genes. Our studies provide insights into the involvement of epigenetics in intellectual disability.


Subject(s)
Astrocytes/metabolism , Cell Differentiation , Gene Expression Regulation , Histone Demethylases/genetics , Transcription Factors/genetics , Animals , Astrocytes/cytology , Binding Sites , Biomarkers , Cell Differentiation/genetics , Cell Proliferation , Gene Expression Profiling , Histone Demethylases/metabolism , Histones/metabolism , Mice , Models, Biological , Neurogenesis , Neurons/metabolism , Protein Binding , Synapses/metabolism , Transcription Factors/metabolism , Transcription, Genetic
4.
Proc Natl Acad Sci U S A ; 116(39): 19464-19473, 2019 09 24.
Article in English | MEDLINE | ID: mdl-31488723

ABSTRACT

Histone H3 lysine 9 methylation (H3K9me) is essential for cellular homeostasis; however, its contribution to development is not well established. Here, we demonstrate that the H3K9me2 demethylase PHF2 is essential for neural progenitor proliferation in vitro and for early neurogenesis in the chicken spinal cord. Using genome-wide analyses and biochemical assays we show that PHF2 controls the expression of critical cell cycle progression genes, particularly those related to DNA replication, by keeping low levels of H3K9me3 at promoters. Accordingly, PHF2 depletion induces R-loop accumulation that leads to extensive DNA damage and cell cycle arrest. These data reveal a role of PHF2 as a guarantor of genome stability that allows proper expansion of neural progenitors during development.


Subject(s)
DNA Damage , Histone Demethylases/metabolism , Homeodomain Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Chick Embryo , DNA Methylation , Embryonic Stem Cells , Epigenesis, Genetic , Genome-Wide Association Study , Histone Demethylases/genetics , Histones/metabolism , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Neural Stem Cells/enzymology , Neurogenesis/physiology , Promoter Regions, Genetic , Transcription Factors/metabolism
5.
Elife ; 72018 08 10.
Article in English | MEDLINE | ID: mdl-30095408

ABSTRACT

Class II HLH proteins heterodimerize with class I HLH/E proteins to regulate transcription. Here, we show that E proteins sharpen neurogenesis by adjusting the neurogenic strength of the distinct proneural proteins. We find that inhibiting BMP signaling or its target ID2 in the chick embryo spinal cord, impairs the neuronal production from progenitors expressing ATOH1/ASCL1, but less severely that from progenitors expressing NEUROG1/2/PTF1a. We show this context-dependent response to result from the differential modulation of proneural proteins' activity by E proteins. E proteins synergize with proneural proteins when acting on CAGSTG motifs, thereby facilitating the activity of ASCL1/ATOH1 which preferentially bind to such motifs. Conversely, E proteins restrict the neurogenic strength of NEUROG1/2 by directly inhibiting their preferential binding to CADATG motifs. Since we find this mechanism to be conserved in corticogenesis, we propose this differential co-operation of E proteins with proneural proteins as a novel though general feature of their mechanism of action.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA/metabolism , Gene Expression Regulation, Developmental , Neurogenesis , Animals , Binding Sites , Chick Embryo , Protein Binding
6.
Nucleic Acids Res ; 46(7): 3351-3365, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29438503

ABSTRACT

During neurogenesis, dynamic developmental cues, transcription factors and histone modifying enzymes regulate the gene expression programs by modulating the activity of neural-specific enhancers. How transient developmental signals coordinate transcription factor recruitment to enhancers and to which extent chromatin modifiers contribute to enhancer activity is starting to be uncovered. Here, we take advantage of neural stem cells as a model to unravel the mechanisms underlying neural enhancer activation in response to the TGFß signaling. Genome-wide experiments demonstrate that the proneural factor ASCL1 assists SMAD3 in the binding to a subset of enhancers. Once located at the enhancers, SMAD3 recruits the histone demethylase JMJD3 and the remodeling factor CHD8, creating the appropriate chromatin landscape to allow enhancer transcription and posterior gene activation. Finally, to analyze the phenotypical traits owed to cis-regulatory regions, we use CRISPR-Cas9 technology to demonstrate that the TGFß-responsive Neurog2 enhancer is essential for proper neuronal polarization.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Enhancer Elements, Genetic/genetics , Neurogenesis/genetics , Smad3 Protein/genetics , Transforming Growth Factor beta/genetics , Animals , CRISPR-Cas Systems/genetics , Cell Lineage/genetics , Cell Polarity/genetics , DNA-Binding Proteins/genetics , Epigenesis, Genetic , Jumonji Domain-Containing Histone Demethylases/genetics , Mice , Nerve Tissue Proteins/genetics , Neural Stem Cells/metabolism , Promoter Regions, Genetic , Signal Transduction/genetics , Transcription Factors/genetics
7.
Nucleic Acids Res ; 45(7): 3800-3811, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28100697

ABSTRACT

A precise immune response is essential for cellular homeostasis and animal survival. The paramount importance of its control is reflected by the fact that its non-specific activation leads to inflammatory events that ultimately contribute to the appearance of many chronic diseases. However, the molecular mechanisms preventing non-specific activation and allowing a quick response upon signal activation are not yet fully understood. In this paper we uncover a new function of PHF8 blocking signal independent activation of immune gene promoters. Affinity purifications coupled with mass spectrometry analysis identified SIN3A and HDAC1 corepressors as new PHF8 interacting partners. Further molecular analysis demonstrated that prior to interferon gamma (IFNγ) stimulation, PHF8 is bound to a subset of IFNγ-responsive promoters. Through the association with HDAC1 and SIN3A, PHF8 keeps the promoters in a silent state, maintaining low levels of H4K20me1. Upon IFNγ treatment, PHF8 is phosphorylated by ERK2 and evicted from the promoters, correlating with an increase in H4K20me1 and transcriptional activation. Our data strongly indicate that in addition to its well-characterized function as a coactivator, PHF8 safeguards transcription to allow an accurate immune response.


Subject(s)
Histone Demethylases/metabolism , Interferon-gamma/pharmacology , Transcription Factors/metabolism , Transcriptional Activation , Cell Line , Chromatin/metabolism , Gene Silencing , Histone Deacetylase 1/metabolism , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Promoter Regions, Genetic , Repressor Proteins/metabolism , Sin3 Histone Deacetylase and Corepressor Complex
8.
Open Biol ; 6(4): 150227, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27248655

ABSTRACT

The function of EZH2 as a transcription repressor is well characterized. However, its role during vertebrate development is still poorly understood, particularly in neurogenesis. Here, we uncover the role of EZH2 in controlling the integrity of the neural tube and allowing proper progenitor proliferation. We demonstrate that knocking down the EZH2 in chick embryo neural tubes unexpectedly disrupts the neuroepithelium (NE) structure, correlating with alteration of the Rho pathway, and reduces neural progenitor proliferation. Moreover, we use transcriptional profiling and functional assays to show that EZH2-mediated repression of p21(WAF1/CIP1) contributes to both processes. Accordingly, overexpression of cytoplasmic p21(WAF1/CIP1) induces NE structural alterations and p21(WAF1/CIP1) suppression rescues proliferation defects and partially compensates for the structural alterations and the Rho activity. Overall, our findings describe a new role of EZH2 in controlling the NE integrity in the neural tube to allow proper progenitor proliferation.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Neuroepithelial Cells/cytology , Neuroepithelial Cells/metabolism , Repressor Proteins/metabolism , Animals , Cell Polarity , Cell Proliferation , Chick Embryo , Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Expression Regulation, Developmental , Humans , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Tube/cytology , Neural Tube/metabolism , Promoter Regions, Genetic/genetics
9.
Neurogenesis (Austin) ; 3(1): e1250034, 2016.
Article in English | MEDLINE | ID: mdl-28090544

ABSTRACT

During early stages of neural development, neuroepithelial cells translocate their nuclei along the apicobasal axis in a harmonized manner with the cell cycle. How cell cycle progression and neuroepithelium polarity are coordinated remains unclear. It has been proposed that developmental cues, epigenetic mechanisms and cell cycle regulators must be linked in order to orchestrate these processes. We have recently discovered that a master epigenetic factor, EZH2 is essential to coordinate these events. EZH2 directly represses the cell cycle regulator p21WAF1/CIP in the chicken spinal cord. By doing so, EZH2 controls neural progenitor cell renewal and fine-tunes Rho signaling pathway, which is essential to maintain neuroepithelial structure. Our findings point to a new role of EZH2 during development that could have potential implication in other areas as cancer.

10.
Cell Tissue Res ; 359(1): 87-98, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24950624

ABSTRACT

Central nervous system (CNS) development is driven by coordinated actions of developmental signals and chromatin regulators that precisely regulate gene expression patterns. Histone methylation is a regulatory mechanism that controls transcriptional programs. In the last 10 years, several histone demethylases (HDM) have been identified as important players in neural development, and their implication in cell fate decisions is beginning to be recognized. Identification of the physiological roles of these enzymes and their molecular mechanisms of action will be necessary for completely understanding the process that ultimately generates different neural cells in the CNS. In this review, we provide an overview of the Jumonji family of HDMs involved in neurodevelopment, and we discuss their roles during neural fate establishment and neuronal differentiation.


Subject(s)
Histone Demethylases/metabolism , Nervous System/growth & development , Neurogenesis , Animals , Humans , Models, Biological , Multigene Family , Nervous System/pathology , Nervous System Diseases/enzymology , Nervous System Diseases/pathology
11.
Carcinogenesis ; 35(10): 2194-202, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24853677

ABSTRACT

Cell transformation is clearly linked to epigenetic changes. However, the role of the histone-modifying enzymes in this process is still poorly understood. In this study, we investigated the contribution of the histone acetyltransferase (HAT) enzymes to Ras-mediated transformation. Our results demonstrated that lysine acetyltransferase 5, also known as Tip60, facilitates histone acetylation of bulk chromatin in Ras-transformed cells. As a consequence, global H4 acetylation (H4K8ac and H4K12ac) increases in Ras-transformed cells, rendering a more decompacted chromatin than in parental cells. Furthermore, low levels of CREB-binding protein (CBP) lead to hypoacetylation of retinoblastoma 1 (Rb1) and cyclin-dependent kinase inhibitor 1B (Cdkn1b or p27Kip1) tumour suppressor gene promoters to facilitate Ras-mediated transformation. In agreement with these data, overexpression of Cbp counteracts Ras transforming capability in a HAT-dependent manner. Altogether our results indicate that CBP and Tip60 coordinate histone acetylation at both local and global levels to facilitate Ras-induced transformation.


Subject(s)
CREB-Binding Protein/metabolism , Cell Transformation, Neoplastic/genetics , Genes, ras , Histone Acetyltransferases/metabolism , Histones/metabolism , Trans-Activators/metabolism , Acetylation , Animals , CREB-Binding Protein/genetics , Chromatin/metabolism , Chromatin/ultrastructure , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Histone Acetyltransferases/genetics , Lysine Acetyltransferase 5 , Mice , NIH 3T3 Cells/pathology , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic , Signal Transduction , Trans-Activators/genetics
12.
Neurobiol Dis ; 67: 49-56, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24657916

ABSTRACT

Epigenetic mechanisms are fundamental for shaping the activity of the central nervous system (CNS). Methyl-CpG binding protein 2 (MECP2) acts as a bridge between methylated DNA and transcriptional effectors responsible for differentiation programs in neurons. The importance of MECP2 dosage in CNS is evident in Rett Syndrome and MECP2 duplication syndrome, which are neurodevelopmental diseases caused by loss-of-function mutations or duplication of the MECP2 gene, respectively. Although many studies have been performed on Rett syndrome models, little is known about the effects of an increase in MECP2 dosage. Herein, we demonstrate that MECP2 overexpression affects neural tube formation, leading to a decrease in neuroblast proliferation in the neural tube ventricular zone. Furthermore, an increase in MECP2 dose provokes premature differentiation of neural precursors accompanied by greater cell death, resulting in a loss of neuronal populations. Overall, our data indicate that correct MECP2 expression levels are required for proper nervous system development.


Subject(s)
Methyl-CpG-Binding Protein 2/genetics , Neural Tube/growth & development , Neural Tube/metabolism , Amino Acid Sequence , Animals , Apoptosis , Chickens , Gene Dosage , Humans , Methyl-CpG-Binding Protein 2/metabolism , Molecular Sequence Data , Spinal Cord/growth & development , Spinal Cord/metabolism
13.
Mol Biol Cell ; 24(3): 351-60, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23243002

ABSTRACT

JMJD3 H3K27me3 demethylase plays an important role in the transcriptional response to different signaling pathways; however, the mechanism by which it facilitates transcription has been unclear. Here we show that JMJD3 regulates transcription of transforming growth factor ß (TGFß)-responsive genes by promoting RNA polymerase II (RNAPII) progression along the gene bodies. Using chromatin immunoprecipitation followed by sequencing experiments, we show that, upon TGFß treatment, JMJD3 and elongating RNAPII colocalize extensively along the intragenic regions of TGFß target genes. According to these data, genome-wide analysis shows that JMJD3-dependent TGFß target genes are enriched in H3K27me3 before TGFß signaling pathway activation. Further molecular analyses demonstrate that JMJD3 demethylates H3K27me3 along the gene bodies, paving the way for the RNAPII progression. Overall these findings uncover the mechanism by which JMJD3 facilitates transcriptional activation.


Subject(s)
Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/physiology , Protein Processing, Post-Translational , RNA Polymerase II/metabolism , Transcription, Genetic , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chromatin Immunoprecipitation , Cyclin-Dependent Kinase 9/metabolism , Genome , HEK293 Cells , Humans , Methylation , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Sequence Analysis, DNA , Smad3 Protein/metabolism , Transcription Initiation Site , Transforming Growth Factor beta/physiology
14.
Nucleic Acids Res ; 40(19): 9429-40, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22850744

ABSTRACT

PHF8 is a histone demethylase associated with X-linked mental retardation. It has been described as a transcriptional co-activator involved in cell cycle progression, but its physiological role is still poorly understood. Here we show that PHF8 controls the expression of genes involved in cell adhesion and cytoskeleton organization such as RhoA, Rac1 and GSK3ß. A lack of PHF8 not only results in a cell cycle delay but also in a disorganized actin cytoskeleton and impaired cell adhesion. Our data demonstrate that PHF8 directly regulates the expression of these genes by demethylating H4K20me1 at promoters. Moreover, c-Myc transcription factor cooperates with PHF8 to regulate the analysed promoters. Further analysis in neurons shows that depletion of PHF8 results in down-regulation of cytoskeleton genes and leads to a deficient neurite outgrowth. Overall, our results suggest that the mental retardation phenotype associated with loss of function of PHF8 could be due to abnormal neuronal connections as a result of alterations in cytoskeleton function.


Subject(s)
Actin Cytoskeleton/ultrastructure , Gene Expression Regulation , Histone Demethylases/physiology , Transcription Factors/physiology , Transcription, Genetic , Animals , Cell Adhesion/genetics , Cell Line , Cells, Cultured , Down-Regulation , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , HeLa Cells , Histone Demethylases/antagonists & inhibitors , Humans , Mice , Neurites/ultrastructure , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/antagonists & inhibitors , alpha Catenin/genetics , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
15.
Development ; 139(15): 2681-91, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22782721

ABSTRACT

Neural development requires crosstalk between signaling pathways and chromatin. In this study, we demonstrate that neurogenesis is promoted by an interplay between the TGFß pathway and the H3K27me3 histone demethylase (HDM) JMJD3. Genome-wide analysis showed that JMJD3 is targeted to gene promoters by Smad3 in neural stem cells (NSCs) and is essential to activate TGFß-responsive genes. In vivo experiments in chick spinal cord revealed that the generation of neurons promoted by Smad3 is dependent on JMJD3 HDM activity. Overall, these findings indicate that JMJD3 function is required for the TGFß developmental program to proceed.


Subject(s)
Developmental Biology/methods , Gene Expression Regulation , Genome-Wide Association Study , Jumonji Domain-Containing Histone Demethylases/metabolism , Neurons/metabolism , Smad3 Protein/metabolism , Animals , Chick Embryo , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Mice , Models, Biological , Oligonucleotide Array Sequence Analysis , Phosphorylation , Spinal Cord/embryology , Transforming Growth Factor beta/metabolism
16.
Development ; 137(17): 2915-25, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20667911

ABSTRACT

During spinal cord development, the combination of secreted signaling proteins and transcription factors provides information for each neural type differentiation. Studies using embryonic stem cells show that trimethylation of lysine 27 of histone H3 (H3K27me3) contributes to repression of many genes key for neural development. However, it remains unclear how H3K27me3-mediated mechanisms control neurogenesis in developing spinal cord. Here, we demonstrate that H3K27me3 controls dorsal interneuron generation by regulation of BMP activity. Our study indicates that expression of Noggin, a BMP extracellular inhibitor, is repressed by H3K27me3. Moreover, we show that Noggin expression is induced by BMP pathway signaling, generating a negative-feedback regulatory loop. In response to BMP pathway activation, JMJD3 histone demethylase interacts with the Smad1/Smad4 complex to demethylate and activate the Noggin promoter. Together, our data reveal how the BMP signaling pathway restricts its own activity in developing spinal cord by modulating H3K27me3 levels at the Noggin promoter.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Histones/metabolism , Spinal Cord/embryology , Spinal Cord/metabolism , Animals , Animals, Genetically Modified , Base Sequence , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Differentiation , Cell Proliferation , Chick Embryo , DNA Primers/genetics , Epigenesis, Genetic , Histones/chemistry , Humans , Methylation , Models, Neurological , Neurogenesis , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Smad Proteins/genetics , Smad Proteins/metabolism , Spinal Cord/cytology
17.
J Biol Chem ; 284(3): 1343-52, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19015268

ABSTRACT

Acetylation is a posttranslational modification that alters the biological activities of proteins by affecting their association with other proteins or DNA, their catalytic activities, or their subcellular distribution. The acetyltransferase P/CAF is autoacetylated and acetylated by p300 in vivo. P/CAF autoacetylation is an intramolecular or intermolecular event. Intramolecular acetylation targets five lysines within the nuclear localization signal at the P/CAF C terminus. We analyzed how the subcellular distribution of P/CAF is regulated by intramolecular autoacetylation and found that a P/CAF mutant lacking histone acetyltransferase activity accumulated primarily in the cytoplasm. This cytoplasmic fraction of P/CAF is enriched for nonautoacetylated P/CAF. In addition, P/CAF deacetylation by HDAC3 and in a minor degree by HDAC1, HDAC2, or HDAC4 leads to cytoplasmic accumulation of P/CAF. Importantly, our data show that P/CAF accumulates in the cytoplasm during apoptosis. These results reveal the molecular mechanism of autoacetylation control of P/CAF nuclear translocation and suggest a novel pathway by which P/CAF activity is controlled in vivo.


Subject(s)
Apoptosis/physiology , Cell Nucleus/enzymology , Cytoplasm/enzymology , Nuclear Localization Signals/metabolism , Protein Processing, Post-Translational/physiology , p300-CBP Transcription Factors/metabolism , Acetylation , Active Transport, Cell Nucleus/physiology , Animals , Cell Nucleus/genetics , Cytoplasm/genetics , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Humans , Mice , Mutation , NIH 3T3 Cells , Nuclear Localization Signals/genetics , Protein Structure, Tertiary/physiology , p300-CBP Transcription Factors/genetics
18.
Nucleic Acids Res ; 35(6): 1958-68, 2007.
Article in English | MEDLINE | ID: mdl-17341466

ABSTRACT

Simian Virus 40 (SV40) large T antigen (T Ag) is a multifunctional viral oncoprotein that regulates viral and cellular transcriptional activity. However, the mechanisms by which such regulation occurs remain unclear. Here we show that T antigen represses CBP-mediated transcriptional activity. This repression is concomitant with histone H3 deacetylation and is TSA sensitive. Moreover, our results demonstrate that T antigen interacts with HDAC1 in vitro in an Rb-independent manner. In addition, the overexpression of HDAC1 cooperates with T antigen to antagonize CBP transactivation function and correlates with chromatin deacetylation of the TK promoter. Finally, decreasing HDAC1 levels with small interfering RNA (siRNA) partially abolishes T antigen-induced repression. These findings highlight the importance of the histone acetylation/deacetylation balance in the cellular transformation mediated by oncoviral proteins.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , CREB-Binding Protein/antagonists & inhibitors , Gene Expression Regulation , Histone Deacetylases/metabolism , Histones/metabolism , Acetylation , Animals , Chromatin/enzymology , Humans , Repressor Proteins/metabolism , Transcription, Genetic
19.
Biochem J ; 398(2): 215-24, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16704373

ABSTRACT

The CBP [CREB (cAMP-response-element-binding protein)-binding protein]/p300 acetyltransferases function as transcriptional co-activators and play critical roles in cell differentiation and proliferation. Accumulating evidence shows that alterations of the CBP/p300 protein levels are linked to human tumours. In the present study, we show that the levels of the CBP/p300 co-activators are decreased dramatically by continuous PDGF (platelet-derived growth factor) and Ras signalling pathway activation in NIH 3T3 fibroblasts. This effect occurs by reducing the expression levels of the CBP/p300 genes. In addition, CBP and p300 are degraded by the 26 S proteasome pathway leading to an overall decrease in the levels of the CBP/p300 proteins. Furthermore, we provide evidence that Mdm2 (murine double minute 2), in the presence of active H-Ras or N-Ras, induces CBP/p300 degradation in NIH 3T3 cells. These findings support a novel mechanism for modulating other signalling transduction pathways that require these common co-activators.


Subject(s)
Cell Cycle Proteins/metabolism , Histone Acetyltransferases/metabolism , Signal Transduction , Transcription Factors/metabolism , ras Proteins/metabolism , Animals , Cell Cycle Proteins/genetics , Enzyme Activation/drug effects , Gene Expression , Histone Acetyltransferases/genetics , Mice , NIH 3T3 Cells , Platelet-Derived Growth Factor/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Transcription Factors/genetics , Ubiquitin/metabolism , Valine/genetics , Valine/metabolism , p300-CBP Transcription Factors , ras Proteins/genetics
20.
J Biol Chem ; 280(52): 42592-600, 2005 Dec 30.
Article in English | MEDLINE | ID: mdl-16199528

ABSTRACT

The global inhibition of transcription at the mitotic phase of the cell cycle occurs together with the general displacement of transcription factors from the mitotic chromatin. Nevertheless, the DNase- and potassium permanganate-hypersensitive sites are maintained on potentially active promoters during mitosis, helping to mark active genes at this stage of the cell cycle. Our study focuses on the role of histone acetylation and H3 (Lys-4) methylation in the maintenance of the competency of these active genes during mitosis. To this end we have analyzed histone modifications across the promoters and coding regions of constitutively active, inducible, and inactive genes in mitotic arrested cells. Our results show that basal histone modifications are maintained during mitosis at promoters and coding regions of the active and inducible RNA polymerase II-transcribed genes. In addition we have demonstrated that, together with H3 acetylation and H3 (Lys-4) methylation, H4 (Lys-12) acetylation at the coding regions contributes to the formation of a stable mark on active genes at this stage of the cell cycle. Finally, analysis of cyclin B1 gene activation during mitosis revealed that the former occurs with a strong increase of H3 (Lys-4) trimethylation but not H3 or H4 acetylation, suggesting that histone methyltransferases are active during this stage. These data demonstrate a critical role of histone acetylation and H3 (Lys-4) methylation during mitosis in marking and activating genes during the mitotic stage of the cell cycle.


Subject(s)
Histones/chemistry , Mitosis , Acetylation , Cell Cycle , Cell Nucleus/metabolism , Cell Nucleus Division , Chromatin/chemistry , Chromatin/metabolism , Chromatin Immunoprecipitation , Cyclin B/metabolism , Cyclin B1 , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation , HeLa Cells , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Humans , Immunoblotting , Interphase , Lysine/chemistry , Methylation , Nucleosomes/metabolism , Polymerase Chain Reaction , Promoter Regions, Genetic , Protein Methyltransferases , Protein Processing, Post-Translational , RNA Polymerase II/metabolism , Temperature , Transcription, Genetic , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...