Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Epigenetics Chromatin ; 16(1): 46, 2023 Nov 28.
Article in English | MEDLINE | ID: mdl-38017471

ABSTRACT

Chromatin plays a central role in the conversion of energy in cells: alteration of chromatin structure to make DNA accessible consumes energy, and compaction of chromatin preserves energy. Alteration of chromatin structure uses energy sources derived from carbon metabolism such as ATP and acetyl-CoA; conversely, chromatin compaction and epigenetic modification feedback to metabolism and energy homeostasis by controlling gene expression and storing metabolites. Coordination of these dual chromatin events must be flexibly modulated in response to environmental changes such as during development and exposure to stress. Aging also alters chromatin structure and the coordination of metabolism, chromatin dynamics, and other cell processes. Noncoding RNAs and other RNA species that associate directly with chromatin or with chromatin modifiers contribute to spatiotemporal control of transcription and energy conversion. The time required for generating the large amounts of RNAs and chromatin modifiers observed in super-enhancers may be critical for regulation of transcription and may be impacted by aging. Here, taking into account these factors, we review alterations of chromatin that are fundamental to cell responses to metabolic changes due to stress and aging to maintain redox and energy homeostasis. We discuss the relationship between spatiotemporal control of energy and chromatin function, as this emerging concept must be considered to understand how cell homeostasis is maintained.


Subject(s)
Chromatin , Epigenesis, Genetic , Oxidation-Reduction , Homeostasis
2.
Molecules ; 27(12)2022 Jun 10.
Article in English | MEDLINE | ID: mdl-35744859

ABSTRACT

Molybdenum cofactor (Moco) biosynthesis requires iron, copper, and ATP. The Moco-containing enzyme sulfite oxidase catalyzes terminal oxidation in oxidative cysteine catabolism, and another Moco-containing enzyme, xanthine dehydrogenase, functions in purine catabolism. Thus, molybdenum enzymes participate in metabolic pathways that are essential for cellular detoxication and energy dynamics. Studies of the Moco biosynthetic enzymes MoaE (in the Ada2a-containing (ATAC) histone acetyltransferase complex) and MOCS2 have revealed that Moco biosynthesis and molybdenum enzymes align to regulate signaling and metabolism via control of transcription and translation. Disruption of these functions is involved in the onset of dementia and neurodegenerative disease. This review provides an overview of the roles of MoaE and MOCS2 in normal cellular processes and neurodegenerative disease, as well as directions for future research.


Subject(s)
Metalloproteins , Neurodegenerative Diseases , Sulfite Oxidase , Coenzymes/metabolism , Humans , Molybdenum/metabolism , Molybdenum Cofactors , Sulfite Oxidase/metabolism , Sulfurtransferases , Xanthine Dehydrogenase/metabolism
3.
Redox Biol ; 51: 102270, 2022 05.
Article in English | MEDLINE | ID: mdl-35189552

ABSTRACT

Overproduction of reactive oxygen species (ROS) drives inflammation and mutagenesis. However, the role of the DNA damage response in immune responses remains largely unknown. Here we found that stabilization of the mismatch repair (MMR) protein MSH6 in response to alkylation damage requires interactions with the molybdopterin synthase associating complex (MPTAC) and Ada2a-containing histone acetyltransferase complex (ATAC). Furthermore, MSH6 promotes sterol biosynthesis via the mevalonate pathway in a MPTAC- and ATAC-dependent manner. MPTAC reduces the source of alkylating agents (ROS). Therefore, the association between MMR proteins, MPTAC, and ATAC promotes anti-inflammation response and reduces alkylating agents. The inflammatory responses measured by xanthine oxidase activity are elevated in Lymphoblastoid Cell Lines (LCLs) from some Fragile X-associated disorders (FXD) patients, suggesting that alkylating agents are increased in these FXD patients. However, MPTAC is disrupted in LCLs from some FXD patients. In LCLs from other FXD patients, interaction between MSH6 and ATAC was lost, destabilizing MSH6. Thus, impairment of MPTAC and ATAC may cause alkylation damage resistance in some FXD patients.


Subject(s)
DNA Damage , DNA-Binding Proteins , Alkylating Agents/pharmacology , Alkylation , DNA Repair , DNA-Binding Proteins/genetics , Humans , Reactive Oxygen Species , Sterols
5.
Int J Mol Sci ; 22(19)2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34638614

ABSTRACT

Inflammation is the body's means of defense against harmful stimuli, with the ultimate aim being to restore homeostasis. Controlled acute inflammation transiently activates an immune response and can be beneficial as protection against infection or injury. However, dysregulated inflammatory responses, including chronic inflammation, disrupt the immune system's ability to maintain homeostatic balance, leading to increased susceptibility to infection, continuous tissue damage, and dysfunction. Aging is a risk factor for chronic inflammation; their coincidence is termed "inflammaging". Metabolic disorders including obesity, neurodegenerative diseases, and atherosclerosis are often encountered in old age. Therefore, it is important to understand the mechanistic relationship between aging, chronic inflammation, and metabolism. It has been established that the expression of inflammatory mediators is transcriptionally and translationally regulated. In addition, the post-translational modification of the mediators plays a crucial role in the response to inflammatory signaling. Chromatin regulation responds to metabolic status and controls homeostasis. However, chromatin structure is also changed by aging. In this review, we discuss the functional contributions of chromatin regulation to inflammaging.


Subject(s)
Aging/immunology , Aging/metabolism , Chromatin/metabolism , Macrophages/immunology , Macrophages/metabolism , Animals , Histone Code , Humans , Inflammation/immunology , Inflammation/metabolism , Models, Biological , Nucleosomes/immunology , Nucleosomes/metabolism
6.
Front Endocrinol (Lausanne) ; 12: 731648, 2021.
Article in English | MEDLINE | ID: mdl-34526971

ABSTRACT

The mechanisms of epigenetic gene regulation-histone modifications, chromatin remodeling, DNA methylation, and noncoding RNA-use metabolites as enzymatic cofactors and substrates in reactions that allow chromatin formation, nucleotide biogenesis, transcription, RNA processing, and translation. Gene expression responds to demands from cellular processes that use specific metabolites and alters or maintains cellular metabolic status. However, the roles of metabolites-particularly nucleotides-as regulatory molecules in epigenetic regulation and biological processes remain largely unknown. Here we review the crosstalk between gene expression, nucleotide metabolism, and cellular processes, and explore the role of metabolism in epigenetics as a critical regulator of biological events.


Subject(s)
Epigenesis, Genetic/physiology , Nucleotides/metabolism , Animals , Chromatin/metabolism , Chromatin Assembly and Disassembly/genetics , Gene Expression/physiology , Histones/metabolism , Humans , Protein Processing, Post-Translational/genetics
7.
J Cell Biochem ; 120(9): 16088-16107, 2019 09.
Article in English | MEDLINE | ID: mdl-31090959

ABSTRACT

NUAK1 is a serine/threonine kinase member of the AMPK-α family. NUAK1 regulates several processes in tumorigenesis; however, its regulation and molecular targets are still poorly understood. Bioinformatics analysis predicted that the majority of NUAK1 localizes in the nucleus. However, there are no studies about the regulation of NUAK1 subcellular distribution. Here, we analyzed NUAK1 localization in several human cell lines, mouse embryo fibroblasts, and normal mouse tissues. We found that NUAK1 is located in the nucleus and also in the cytoplasm. Through bioinformatics analysis and studies comparing subcellular localization of wild type and NUAK1 mutants, we identified a conserved bipartite nuclear localization signal at the N-terminal domain of NUAK1. Based on mass spectrometry analysis, we found that NUAK1 interacts with importin-ß members including importin-ß1 (KPNB1), importin-7 (IPO7), and importin-9 (IPO9). We confirmed that importin-ß members are responsible for NUAK1 nuclear import through the inhibition of importin-ß by Importazole and the knockdown of either IPO7 or IPO9. In addition, we found that oxidative stress induces NUAK1 cytoplasmic accumulation, indicating that oxidative stress affects NUAK1 nuclear transport. Thus, our study is the first evidence of an active nuclear transport mechanism regulating NUAK1 subcellular localization. These data will lead to investigations of the molecular targets of NUAK1 according to its subcellular distribution, which could be new biomarkers or targets for cancer therapies.


Subject(s)
Nuclear Localization Signals/metabolism , Protein Kinases/metabolism , Repressor Proteins/metabolism , beta Karyopherins/metabolism , Active Transport, Cell Nucleus , Animals , Cell Line , Cytoplasm/metabolism , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Mice , Oxidative Stress , Protein Kinases/genetics , Repressor Proteins/genetics
8.
Cell Rep ; 24(6): 1585-1596, 2018 08 07.
Article in English | MEDLINE | ID: mdl-30089268

ABSTRACT

Metabolic disorder has been suggested to underlie Alzheimer's disease (AD). However, the decisive molecular linkages remain unclear. We discovered that human Molybdopterin Synthase Associating Complex, MPTAC, promotes sulfur amino acid catabolism to prevent oxidative damage from excess sulfur amino acids, which, in turn, advances fatty acid oxidation and acetyl coenzyme A (acetyl-CoA) synthesis. The association of MPTAC with Protein arginine (R) Methyltransferase 5 (PRMT5) complex and small nuclear ribonucleoprotein (SNRP) splicing factors enables SNRPs to sense metabolic states through their methylation. This promotes the splicing fidelity of amyloid precursor protein (APP) pre-mRNA and proper APP fragmentation, abnormalities of which have been observed in the platelets of AD patients. The functions of MPTAC are crucial to maintain expression of drebrin 1, which is required for synaptic plasticity, through prevention from oxidative damage. Thus, adjustment of sulfur amino acid catabolism by MPTAC prevents events that occur early in the onset of AD.


Subject(s)
Alzheimer Disease/metabolism , Amino Acids, Sulfur/metabolism , Amyloid beta-Protein Precursor/metabolism , Sulfurtransferases/metabolism , HEK293 Cells , Humans , Neuronal Plasticity
9.
Elife ; 72018 06 25.
Article in English | MEDLINE | ID: mdl-29938647

ABSTRACT

Upon nutritional stress, the metabolic status of cells is changed by nutrient signaling pathways to ensure survival. Altered metabolism by nutrient signaling pathways has been suggested to influence cellular lifespan. However, it remains unclear how chromatin regulation is involved in this process. Here, we found that histone H3 threonine 11 phosphorylation (H3pT11) functions as a marker for nutritional stress and aging. Sch9 and CK2 kinases cooperatively regulate H3pT11 under stress conditions. Importantly, H3pT11 defective mutants prolonged chronological lifespan (CLS) by altering nutritional stress responses. Thus, the phosphorylation of H3T11 by Sch9 and CK2 links a nutritional stress response to chromatin in the regulation of CLS.


Subject(s)
Casein Kinase II/metabolism , Gene Expression Regulation, Fungal , Histones/metabolism , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Stress, Physiological/genetics , Acetic Acid/metabolism , Acetic Acid/pharmacology , Casein Kinase II/genetics , Cell Division , Chromatin/chemistry , Chromatin/metabolism , Culture Media/pharmacology , Glucose/deficiency , Glucose/pharmacology , Histones/genetics , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Subunits/genetics , Protein Subunits/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction , Threonine/metabolism
10.
Annu Rev Biochem ; 87: 27-49, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29925263

ABSTRACT

Chromatin is a mighty consumer of cellular energy generated by metabolism. Metabolic status is efficiently coordinated with transcription and translation, which also feed back to regulate metabolism. Conversely, suppression of energy utilization by chromatin processes may serve to preserve energy resources for cell survival. Most of the reactions involved in chromatin modification require metabolites as their cofactors or coenzymes. Therefore, the metabolic status of the cell can influence the spectra of posttranslational histone modifications and the structure, density and location of nucleosomes, impacting epigenetic processes. Thus, transcription, translation, and DNA/RNA biogenesis adapt to cellular metabolism. In addition to dysfunctions of metabolic enzymes, imbalances between metabolism and chromatin activities trigger metabolic disease and life span alteration. Here, we review the synthesis of the metabolites and the relationships between metabolism and chromatin function. Furthermore, we discuss how the chromatin response feeds back to metabolic regulation in biological processes.


Subject(s)
Chromatin/metabolism , Aging/genetics , Aging/metabolism , Animals , Chromatin/genetics , Chromatin Assembly and Disassembly , Energy Metabolism , Epigenesis, Genetic , Histone Code , Humans , Longevity/genetics , Longevity/physiology , Models, Biological
12.
J Mol Cell Biol ; 8(1): 44-50, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26705305

ABSTRACT

Molybdenum cofactor (Moco) biosynthesis is linked to c-Jun N-terminal kinase (JNK) signaling in Drosophila through MoaE, a molybdopterin (MPT) synthase subunit that is also a component of the Ada Two A containing (ATAC) acetyltransferase complex. Here, we show that human MPT synthase and ATAC inhibited PKR, a double-stranded RNA-dependent protein kinase, to facilitate translation initiation of iron-responsive mRNA. MPT synthase and ATAC directly interacted with PKR and suppressed latent autophosphorylation of PKR and its downstream phosphorylation of JNK and eukaryotic initiation factor 2α (eIF2α). The suppression of eIF2α phosphorylation via MPT synthase and ATAC prevented sequestration of the guanine nucleotide exchange factor eIF2B, which recycles eIF2-GDP to eIF2-GTP, resulting in the promotion of translation initiation. Indeed, translation of the iron storage protein, ferritin, was reduced in the absence of MPT synthase or ATAC subunits. Thus, MPT synthase and ATAC regulate latent PKR signaling and link transcription and translation initiation.


Subject(s)
Acetyltransferases/metabolism , Coenzymes/biosynthesis , Metalloproteins/biosynthesis , eIF-2 Kinase/metabolism , Animals , Cell Line , Eukaryotic Initiation Factor-2/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Molybdenum Cofactors , Phosphorylation , Protein Biosynthesis , Pteridines , Sulfurtransferases/metabolism
13.
Mol Cell ; 60(3): 408-21, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26527276

ABSTRACT

Pyruvate kinase M2 (PKM2) is a key enzyme for glycolysis and catalyzes the conversion of phosphoenolpyruvate (PEP) to pyruvate, which supplies cellular energy. PKM2 also phosphorylates histone H3 threonine 11 (H3T11); however, it is largely unknown how PKM2 links cellular metabolism to chromatin regulation. Here, we show that the yeast PKM2 homolog, Pyk1, is a part of a novel protein complex named SESAME (Serine-responsive SAM-containing Metabolic Enzyme complex), which contains serine metabolic enzymes, SAM (S-adenosylmethionine) synthetases, and an acetyl-CoA synthetase. SESAME interacts with the Set1 H3K4 methyltransferase complex, which requires SAM synthesized from SESAME, and recruits SESAME to target genes, resulting in phosphorylation of H3T11. SESAME regulates the crosstalk between H3K4 methylation and H3T11 phosphorylation by sensing glycolysis and glucose-derived serine metabolism. This leads to auto-regulation of PYK1 expression. Thus, our study provides insights into the mechanism of regulating gene expression, responding to cellular metabolism via chromatin modifications.


Subject(s)
Chromatin/metabolism , Histones/metabolism , Multiprotein Complexes/metabolism , Protein-Tyrosine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Chromatin/genetics , Gene Expression Regulation, Enzymologic/physiology , Gene Expression Regulation, Fungal/physiology , Histones/genetics , Multiprotein Complexes/genetics , Phosphorylation/physiology , Protein-Tyrosine Kinases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
14.
Biochim Biophys Acta ; 1829(11): 1218-24, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23891849

ABSTRACT

C-Jun is a major transcription factor belonging to the activating protein 1 (AP-1) family. Phosphorylation has been shown to be critical for c-Jun activation and stability. Here, we report that Jra, the Drosophila Jun protein, is acetylated in vivo. We demonstrate that the acetylation of Jra leads to its rapid degradation in response to osmotic stress. Intriguingly, we also found that Jra phosphorylation antagonized its acetylation, indicating the opposite roles of acetylation and phosphorylation in Jra degradation process under osmotic stress. Our results provide new insights into how c-Jun proteins are precisely regulated by the interplay of different posttranslational modifications.


Subject(s)
Drosophila/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Acetylation , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Phosphorylation , Proto-Oncogene Proteins c-jun/chemistry , Sequence Homology, Amino Acid , Ubiquitination
15.
Curr Opin Cell Biol ; 25(3): 322-6, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23498660

ABSTRACT

Signaling involves the coordinated action of multiple molecules including stimuli, receptors and enzymes part of which interact with the transcriptional machinery and target chromatin. Signaling systems regulate the cell events responsible for survival, development and homeostasis. Many of the signaling pathways induce target gene activation through interaction with the transcription machinery, including RNA polymerase II, and with histone modifying complexes. These studies are having a broad impact on chromatin biology. Recent studies suggest that chromatin itself receives the signals. Increasing examples are illustrating novel regulatory mechanisms that promote our understanding of development and disease.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/metabolism , Signal Transduction , Animals , Histones/metabolism , Humans , Mitogen-Activated Protein Kinase Kinases/metabolism , Nucleosomes/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Transcription, Genetic
16.
J Mol Cell Biol ; 4(5): 348-50, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22831833

ABSTRACT

Signal transduction pathways alter the gene expression program in response to extracellular or intracellular cues. Mitogen-activated protein kinases (MAPKs) govern numerous cellular processes including cell growth, stress response, apoptosis, and differentiation. In the past decade, MAPKs have been shown to regulate the transcription machinery and associate with chromatin-modifying complexes. Moreover, recent studies demonstrate that several MAPKs bind directly to chromatin at target genes. This review highlights the recent discoveries of MAPK signaling in regard to histone modifications and chromatin regulation. Evidence suggesting that further unknown mechanisms integrate signal transduction with chromatin biology is discussed.


Subject(s)
Histones/metabolism , Mitogen-Activated Protein Kinases/metabolism , Animals , Humans , Signal Transduction
17.
Mol Cell Proteomics ; 11(5): 90-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22345504

ABSTRACT

Molybdopterin (MPT) synthase is an essential enzyme involved in the synthesis of the molybdenum cofactor precursor molybdopterin. The molybdenum cofactor biosynthetic pathway is conserved from prokaryotes to Metazoa. CG10238 is the Drosophila homolog of the MoaE protein, a subunit of MPT synthase, and is found in a fusion with the mitogen-activated protein kinase (MAPK)-upstream protein kinase-binding inhibitory protein (MBIP). This fused protein inhibits the activation of c-Jun N-terminal kinase (JNK). dMoaE (CG10238) carries out this function as a subunit of the ATAC histone acetyltransferase complex. In this study, we demonstrate that Drosophila MoaE (CG10238) also interacts with Drosophila MoaD and with itself to form a complex with stoichiometry identical to the MPT synthase holoenzyme in addition to its function in ATAC. We also show that sequence determinants that regulate MAPK signaling are located within the MoaE region of dMoaE (CG10238). Analysis of other metazoan MBIPs reveals that MBIP protein sequences have an N-terminal region that appears to have been derived from the MoaE protein, although it has lost residues responsible for catalytic activity. Thus, intact and modified copies of the MoaE protein may have been conscripted to play a new, noncatalytic role in MAPK signaling in Metazoa as part of the ATAC complex.


Subject(s)
Drosophila melanogaster/enzymology , MAP Kinase Signaling System , Protein Subunits/physiology , Sulfurtransferases/genetics , Sulfurtransferases/physiology , Algorithms , Animals , Cell Line , Conserved Sequence , Enzyme Activation , Evolution, Molecular , Immunoprecipitation , Molecular Sequence Data , Phylogeny , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Protein Subunits/isolation & purification , Protein Subunits/metabolism , Sequence Analysis, Protein , Sulfurtransferases/isolation & purification , Sulfurtransferases/metabolism
18.
Biochemistry ; 51(5): 952-62, 2012 Feb 07.
Article in English | MEDLINE | ID: mdl-22242598

ABSTRACT

Transcription factor C/EBPß is involved in several cellular processes, such as proliferation, differentiation, and energy metabolism. This factor exerts its activity through recruitment of different proteins or protein complexes, including the ATP-dependent chromatin remodeling complex SWI/SNF. The C/EBPß protein is found as three major isoforms, C/EBPß1, -2, and -3. They are generated by translation at alternative AUG initiation codons of a unique mRNA, C/EBPß1 being the full-length isoform. It has been found that C/EBPß1 participates in terminal differentiation processes. Conversely, C/EBPß2 and -3 promote cell proliferation and are involved in malignant progression in a number of tissues. The mechanisms by which C/EBPß2 and -3 promote cell proliferation and tumor progression are not fully understood. In this work, we sought to identify proteins interacting with hC/EBPß using a proteomics approach. We found that all three isoforms interact with hSNF2H and hACF, components of ACF and CHRAC chromatin remodeling complexes, which belong to the imitation switch subfamily. Additional protein-protein interaction studies confirmed this finding and also showed that hC/EBPß directly interacts with hACF1. By overexpressing hC/EBPß, hSNF2H, and hACF1 in HepG2 cells and analyzing variations in expression of cyclin D1 and other C/EBPß target genes, we observed a functional interaction between C/EBPß and SNF2H/ACF1, characterized mainly by suppression of C/EBPß transactivation activity in the presence of SNF2H and ACF1. Consistent with these findings, induction of differentiation of HepG2 cells by 1% DMSO was accompanied by a reduction in the level of cyclin D1 expression and the appearance of hC/EBPß, hSNF2H, and hACF1 on the promoter region of this gene.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/chemistry , CCAAT-Enhancer-Binding Protein-beta/metabolism , Chromatin Assembly and Disassembly , Genes, Switch , Protein Interaction Mapping , Proteomics/methods , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , CCAAT-Enhancer-Binding Protein-beta/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/metabolism , HeLa Cells , Hep G2 Cells , Humans , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Repressor Proteins/chemistry , Trans-Activators/chemistry , Trans-Activators/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism
19.
Annu Rev Biochem ; 80: 473-99, 2011.
Article in English | MEDLINE | ID: mdl-21529160

ABSTRACT

Alterations of chromatin structure have been shown to be crucial for response to cell signaling and for programmed gene expression in development. Posttranslational histone modifications influence changes in chromatin structure both directly and by targeting or activating chromatin-remodeling complexes. Histone modifications intersect with cell signaling pathways to control gene expression and can act combinatorially to enforce or reverse epigenetic marks in chromatin. Through their recognition by protein complexes with enzymatic activities cross talk is established between different modifications and with other epigenetic pathways, including noncoding RNAs (ncRNAs) and DNA methylation. Here, we review the functions of histone modifications and their exploitation in the programming of gene expression during several events in development.


Subject(s)
Histones/chemistry , Histones/metabolism , Protein Processing, Post-Translational , Animals , Chromatin/chemistry , Chromatin Assembly and Disassembly , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Histones/genetics , Humans , Models, Biological , Nucleosomes/chemistry , RNA, Untranslated/metabolism , Signal Transduction/physiology
20.
Cell ; 142(5): 726-36, 2010 Sep 03.
Article in English | MEDLINE | ID: mdl-20813260

ABSTRACT

In response to extracellular cues, signal transduction activates downstream transcription factors like c-Jun to induce expression of target genes. We demonstrate that the ATAC (Ada two A containing) histone acetyltransferase (HAT) complex serves as a transcriptional cofactor for c-Jun at the Jun N-terminal kinase (JNK) target genes Jra and chickadee. ATAC subunits are required for c-Jun occupancy of these genes and for H4K16 acetylation at the Jra enhancer, promoter, and transcribed sequences. Under conditions of osmotic stress, ATAC colocalizes with c-Jun, recruits the upstream kinases Misshapen, MKK4, and JNK, and suppresses further activation of JNK. Relocalization of these MAPKs and suppression of JNK activation by ATAC are dependent on the CG10238 subunit of ATAC. Thus, ATAC governs the transcriptional response to MAP kinase signaling by serving as both a coactivator of transcription and as a suppressor of upstream signaling.


Subject(s)
Drosophila/metabolism , Histone Acetyltransferases/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , MAP Kinase Signaling System , Sulfurtransferases/metabolism , Animals , Cell Line , Drosophila/enzymology , Drosophila/genetics , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Osmotic Pressure , Protein Structure, Tertiary , Stress, Physiological , Sulfurtransferases/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...