Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 561
Filter
Add more filters

Publication year range
1.
Cell ; 186(11): 2438-2455.e22, 2023 05 25.
Article in English | MEDLINE | ID: mdl-37178687

ABSTRACT

The generation of distinct messenger RNA isoforms through alternative RNA processing modulates the expression and function of genes, often in a cell-type-specific manner. Here, we assess the regulatory relationships between transcription initiation, alternative splicing, and 3' end site selection. Applying long-read sequencing to accurately represent even the longest transcripts from end to end, we quantify mRNA isoforms in Drosophila tissues, including the transcriptionally complex nervous system. We find that in Drosophila heads, as well as in human cerebral organoids, 3' end site choice is globally influenced by the site of transcription initiation (TSS). "Dominant promoters," characterized by specific epigenetic signatures including p300/CBP binding, impose a transcriptional constraint to define splice and polyadenylation variants. In vivo deletion or overexpression of dominant promoters as well as p300/CBP loss disrupted the 3' end expression landscape. Our study demonstrates the crucial impact of TSS choice on the regulation of transcript diversity and tissue identity.


Subject(s)
Alternative Splicing , RNA Isoforms , Transcription Initiation Site , Humans , Polyadenylation , Promoter Regions, Genetic , RNA Isoforms/metabolism , RNA, Messenger/metabolism
2.
Immunity ; 57(2): 364-378.e9, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38301651

ABSTRACT

Mutations of the CBP/p300 histone acetyltransferase (HAT) domain can be linked to leukemic transformation in humans, suggestive of a checkpoint of leukocyte compartment sizes. Here, we examined the impact of reversible inhibition of this domain by the small-molecule A485. We found that A485 triggered acute and transient mobilization of leukocytes from the bone marrow into the blood. Leukocyte mobilization by A485 was equally potent as, but mechanistically distinct from, granulocyte colony-stimulating factor (G-CSF), which allowed for additive neutrophil mobilization when both compounds were combined. These effects were maintained in models of leukopenia and conferred augmented host defenses. Mechanistically, activation of the hypothalamus-pituitary-adrenal gland (HPA) axis by A485 relayed shifts in leukocyte distribution through corticotropin-releasing hormone receptor 1 (CRHR1) and adrenocorticotropic hormone (ACTH), but independently of glucocorticoids. Our findings identify a strategy for rapid expansion of the blood leukocyte compartment via a neuroendocrine loop, with implications for the treatment of human pathologies.


Subject(s)
Bone Marrow , Histone Acetyltransferases , Humans , Histone Acetyltransferases/metabolism , Bone Marrow/metabolism , Histones/metabolism , Neutrophils/metabolism , Hypothalamo-Hypophyseal System/metabolism
3.
Cell ; 174(1): 231-244.e12, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29804834

ABSTRACT

The acetyltransferases CBP and p300 are multifunctional transcriptional co-activators. Here, we combined quantitative proteomics with CBP/p300-specific catalytic inhibitors, bromodomain inhibitor, and gene knockout to reveal a comprehensive map of regulated acetylation sites and their dynamic turnover rates. CBP/p300 acetylates thousands of sites, including signature histone sites and a multitude of sites on signaling effectors and enhancer-associated transcriptional regulators. Time-resolved acetylome analyses identified a subset of CBP/p300-regulated sites with very rapid (<30 min) acetylation turnover, revealing a dynamic balance between acetylation and deacetylation. Quantification of acetylation, mRNA, and protein abundance after CBP/p300 inhibition reveals a kinetically competent network of gene expression that strictly depends on CBP/p300-catalyzed rapid acetylation. Collectively, our in-depth acetylome analyses reveal systems attributes of CBP/p300 targets, and the resource dataset provides a framework for investigating CBP/p300 functions and for understanding the impact of small-molecule inhibitors targeting its catalytic and bromodomain activities.


Subject(s)
Acetyltransferases/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation/drug effects , Acetyltransferases/antagonists & inhibitors , Animals , Cell Line , Gene Knockout Techniques , Half-Life , Heterocyclic Compounds, 4 or More Rings/chemistry , Heterocyclic Compounds, 4 or More Rings/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Histones/metabolism , Humans , Isotope Labeling , Kinetics , Mass Spectrometry , Mice , Peptides/analysis , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Signal Transduction , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Transcriptome/drug effects , p300-CBP Transcription Factors/antagonists & inhibitors , p300-CBP Transcription Factors/genetics
4.
Cell ; 168(1-2): 135-149.e22, 2017 Jan 12.
Article in English | MEDLINE | ID: mdl-28086087

ABSTRACT

CBP/p300 are transcription co-activators whose binding is a signature of enhancers, cis-regulatory elements that control patterns of gene expression in multicellular organisms. Active enhancers produce bi-directional enhancer RNAs (eRNAs) and display CBP/p300-dependent histone acetylation. Here, we demonstrate that CBP binds directly to RNAs in vivo and in vitro. RNAs bound to CBP in vivo include a large number of eRNAs. Using steady-state histone acetyltransferase (HAT) assays, we show that an RNA binding region in the HAT domain of CBP-a regulatory motif unique to CBP/p300-allows RNA to stimulate CBP's HAT activity. At enhancers where CBP interacts with eRNAs, stimulation manifests in RNA-dependent changes in the histone acetylation mediated by CBP, such as H3K27ac, and by corresponding changes in gene expression. By interacting directly with CBP, eRNAs contribute to the unique chromatin structure at active enhancers, which, in turn, is required for regulation of target genes.


Subject(s)
Histone Acetyltransferases/metabolism , RNA, Untranslated/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation , Animals , Cell Line , Enhancer Elements, Genetic , Fibroblasts/metabolism , Histones/metabolism , Mice
5.
Mol Cell ; 84(10): 1855-1869.e5, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38593804

ABSTRACT

RNA transcribed from enhancers, i.e., eRNA, has been suggested to directly activate transcription by recruiting transcription factors and co-activators. Although there have been specific examples of eRNA functioning in this way, it is not clear how general this may be. We find that the AT-hook of SWI/SNF preferentially binds RNA and, as part of the esBAF complex, associates with eRNA transcribed from intronic and intergenic regions. Our data suggest that SWI/SNF is globally recruited in cis by eRNA to cell-type-specific enhancers, representative of two distinct stages that mimic early mammalian development, and not at enhancers that are shared between the two stages. In this manner, SWI/SNF facilitates recruitment and/or activation of MLL3/4, p300/CBP, and Mediator to stage-specific enhancers and super-enhancers that regulate the transcription of metabolic and cell lineage priming-related genes. These findings highlight a connection between ATP-dependent chromatin remodeling and eRNA in cell identity and typical- and super-enhancer activation.


Subject(s)
Cell Lineage , DNA Helicases , Enhancer Elements, Genetic , Nuclear Proteins , Transcription Factors , Transcription Factors/metabolism , Transcription Factors/genetics , DNA Helicases/metabolism , DNA Helicases/genetics , Cell Lineage/genetics , Animals , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Humans , Mice , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics
6.
Mol Cell ; 83(2): 186-202.e11, 2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36669479

ABSTRACT

PGC-1α is well established as a metazoan transcriptional coactivator of cellular adaptation in response to stress. However, the mechanisms by which PGC-1α activates gene transcription are incompletely understood. Here, we report that PGC-1α serves as a scaffold protein that physically and functionally connects the DNA-binding protein estrogen-related receptor α (ERRα), cap-binding protein 80 (CBP80), and Mediator to overcome promoter-proximal pausing of RNAPII and transcriptionally activate stress-response genes. We show that PGC-1α promotes pausing release in a two-arm mechanism (1) by recruiting the positive transcription elongation factor b (P-TEFb) and (2) by outcompeting the premature transcription termination complex Integrator. Using mice homozygous for five amino acid changes in the CBP80-binding motif (CBM) of PGC-1α that destroy CBM function, we show that efficient differentiation of primary myoblasts to myofibers and timely skeletal muscle regeneration after injury require PGC-1α binding to CBP80. Our findings reveal how PGC-1α activates stress-response gene transcription in a previously unanticipated pre-mRNA quality-control pathway.


Subject(s)
RNA Precursors , Transcription Factors , Animals , Mice , DNA-Binding Proteins/genetics , Muscle, Skeletal/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Promoter Regions, Genetic , RNA Cap-Binding Proteins/genetics , RNA Polymerase II/metabolism , RNA Precursors/metabolism , Transcription Factors/metabolism , Transcription, Genetic
7.
Mol Cell ; 82(19): 3580-3597.e9, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36206738

ABSTRACT

Maintenance of appropriate cell states involves epigenetic mechanisms, including Polycomb-group (PcG)-mediated transcriptional repression. While PcG proteins are known to induce chromatin compaction, how PcG proteins gain access to DNA in compact chromatin to achieve long-term silencing is poorly understood. Here, we show that the p300/CREB-binding protein (CBP) co-activator is associated with two-thirds of PcG regions and required for PcG occupancy at many of these in Drosophila and mouse cells. CBP stabilizes RNA polymerase II (Pol II) at PcG-bound repressive sites and promotes Pol II pausing independently of its histone acetyltransferase activity. CBP and Pol II pausing are necessary for RNA-DNA hybrid (R-loop) formation and nucleosome depletion at Polycomb Response Elements (PREs), whereas transcription beyond the pause region is not. These results suggest that non-enzymatic activities of the CBP co-activator have been repurposed to support PcG-mediated silencing, revealing how chromatin regulator interplay maintains transcriptional states.


Subject(s)
Drosophila Proteins , Nucleosomes , Animals , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Chromatin/genetics , Chromatin/metabolism , DNA/metabolism , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Mice , Nucleosomes/genetics , Nucleosomes/metabolism , Polycomb Repressive Complex 1/genetics , Polycomb-Group Proteins/metabolism , Protein Binding , RNA/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism
8.
Mol Cell ; 81(10): 2166-2182.e6, 2021 05 20.
Article in English | MEDLINE | ID: mdl-33765415

ABSTRACT

The metazoan-specific acetyltransferase p300/CBP is involved in activating signal-induced, enhancer-mediated transcription of cell-type-specific genes. However, the global kinetics and mechanisms of p300/CBP activity-dependent transcription activation remain poorly understood. We performed genome-wide, time-resolved analyses to show that enhancers and super-enhancers are dynamically activated through p300/CBP-catalyzed acetylation, deactivated by the opposing deacetylase activity, and kinetic acetylation directly contributes to maintaining cell identity at very rapid (minutes) timescales. The acetyltransferase activity is dispensable for the recruitment of p300/CBP and transcription factors but essential for promoting the recruitment of TFIID and RNAPII at virtually all enhancers and enhancer-regulated genes. This identifies pre-initiation complex assembly as a dynamically controlled step in the transcription cycle and reveals p300/CBP-catalyzed acetylation as the signal that specifically promotes transcription initiation at enhancer-regulated genes. We propose that p300/CBP activity uses a "recruit-and-release" mechanism to simultaneously promote RNAPII recruitment and pause release and thereby enables kinetic activation of enhancer-mediated transcription.


Subject(s)
Enhancer Elements, Genetic , RNA Polymerase II/metabolism , Transcription Initiation, Genetic , p300-CBP Transcription Factors/metabolism , Acetylation , Animals , Biocatalysis , Chromatin/metabolism , Down-Regulation/genetics , Histone Deacetylases/metabolism , Histones/metabolism , Lysine/metabolism , Mice , Models, Biological , Nuclear Proteins/metabolism , Protein Binding , Transcription Factor TFIID/metabolism , Transcription Factors/metabolism
9.
Mol Cell ; 81(10): 2183-2200.e13, 2021 05 20.
Article in English | MEDLINE | ID: mdl-34019788

ABSTRACT

To separate causal effects of histone acetylation on chromatin accessibility and transcriptional output, we used integrated epigenomic and transcriptomic analyses following acute inhibition of major cellular lysine acetyltransferases P300 and CBP in hematological malignancies. We found that catalytic P300/CBP inhibition dynamically perturbs steady-state acetylation kinetics and suppresses oncogenic transcriptional networks in the absence of changes to chromatin accessibility. CRISPR-Cas9 screening identified NCOR1 and HDAC3 transcriptional co-repressors as the principal antagonists of P300/CBP by counteracting acetylation turnover kinetics. Finally, deacetylation of H3K27 provides nucleation sites for reciprocal methylation switching, a feature that can be exploited therapeutically by concomitant KDM6A and P300/CBP inhibition. Overall, this study indicates that the steady-state histone acetylation-methylation equilibrium functions as a molecular rheostat governing cellular transcription that is amenable to therapeutic exploitation as an anti-cancer regimen.


Subject(s)
Biocatalysis , Histones/metabolism , Oncogenes , Transcription, Genetic , p300-CBP Transcription Factors/metabolism , Acetylation , Cell Line , Chromatin/metabolism , Co-Repressor Proteins/metabolism , Conserved Sequence , Evolution, Molecular , Gene Regulatory Networks , Genome , Histone Deacetylases/metabolism , Humans , Kinetics , Methylation , Models, Biological , RNA Polymerase II/metabolism
10.
Genes Dev ; 35(1-2): 65-81, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33334824

ABSTRACT

During developmental progression the genomes of immune cells undergo large-scale changes in chromatin folding. However, insights into signaling pathways and epigenetic control of nuclear architecture remain rudimentary. Here, we found that in activated neutrophils calcium influx rapidly recruited the cohesin-loading factor NIPBL to thousands of active enhancers and promoters to dictate widespread changes in compartment segregation. NIPBL recruitment to enhancers and promoters occurred with distinct kinetics. The induction of NIPBL-binding was coordinate with increased P300, BRG1 and RNA polymerase II occupancy. NIPBL-bound enhancers were associated with NFAT, PU.1, and CEBP cis elements, whereas NIPBL-bound promoters were enriched for GC-rich DNA sequences. Using an acute degradation system, we found that the histone acetyltransferases P300 and CBP maintained H3K27ac abundance and facilitated NIPBL occupancy at enhancers and that active transcriptional elongation is essential to maintain H3K27ac abundance. Chromatin remodelers, containing either of the mutually exclusive BRG1 and BRM ATPases, promoted NIPBL recruitment at active enhancers. Conversely, at active promoters, depletion of BRG1 and BRM showed minimal effect on NIPBL occupancy. Finally, we found that calcium signaling in both primary innate and adaptive immune cells swiftly induced NIPBL occupancy. Collectively, these data reveal how transcriptional regulators, histone acetyltransferases, chromatin remodelers, and transcription elongation promote NIPBL occupancy at active enhancers while the induction of NIPLB occupancy at promoters is primarily associated with GC-rich DNA sequences.


Subject(s)
Calcium/metabolism , Cell Cycle Proteins/metabolism , Enhancer Elements, Genetic/physiology , Genome/physiology , Promoter Regions, Genetic/physiology , Signal Transduction/physiology , Animals , Cell Cycle Proteins/immunology , Cells, Cultured , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/metabolism , DNA-Directed DNA Polymerase/metabolism , Histone Acetyltransferases/metabolism , Histones/metabolism , Mice , NFATC Transcription Factors/metabolism , Neutrophils/cytology , Protein Transport , Transcription Elongation, Genetic
11.
Genes Dev ; 34(17-18): 1113-1127, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32873578

ABSTRACT

The largely nuclear cap-binding complex (CBC) binds to the 5' caps of RNA polymerase II (RNAPII)-synthesized transcripts and serves as a dynamic interaction platform for a myriad of RNA processing factors that regulate gene expression. While influence of the CBC can extend into the cytoplasm, here we review the roles of the CBC in the nucleus, with a focus on protein-coding genes. We discuss differences between CBC function in yeast and mammals, covering the steps of transcription initiation, release of RNAPII from pausing, transcription elongation, cotranscriptional pre-mRNA splicing, transcription termination, and consequences of spurious transcription. We describe parameters known to control the binding of generic or gene-specific cofactors that regulate CBC activities depending on the process(es) targeted, illustrating how the CBC is an ever-changing choreographer of gene expression.


Subject(s)
Gene Expression Regulation , RNA Cap-Binding Proteins/metabolism , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional/genetics , Animals , Cell Nucleus/metabolism , Humans , Open Reading Frames/genetics , Saccharomyces cerevisiae
12.
Genes Dev ; 33(13-14): 828-843, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31171701

ABSTRACT

Adenovirus transformed cells have a dedifferentiated phenotype. Eliminating E1A in transformed human embryonic kidney cells derepressed ∼2600 genes, generating a gene expression profile closely resembling mesenchymal stem cells (MSCs). This was associated with a dramatic change in cell morphology from one with scant cytoplasm and a globular nucleus to one with increased cytoplasm, extensive actin stress fibers, and actomyosin-dependent flattening against the substratum. E1A-induced hypoacetylation at histone H3 Lys27 and Lys18 (H3K27/18) was reversed. Most of the increase in H3K27/18ac was in enhancers near TEAD transcription factors bound by Hippo signaling-regulated coactivators YAP and TAZ. E1A causes YAP/TAZ cytoplasmic sequestration. After eliminating E1A, YAP/TAZ were transported into nuclei, where they associated with poised enhancers with DNA-bound TEAD4 and H3K4me1. This activation of YAP/TAZ required RHO family GTPase signaling and caused histone acetylation by p300/CBP, chromatin remodeling, and cohesin loading to establish MSC-associated enhancers and then superenhancers. Consistent results were also observed in primary rat embryo kidney cells, human fibroblasts, and human respiratory tract epithelial cells. These results together with earlier studies suggest that YAP/TAZ function in a developmental checkpoint controlled by signaling from the actin cytoskeleton that prevents differentiation of a progenitor cell until it is in the correct cellular and tissue environment.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adenovirus E1A Proteins/metabolism , Cell Differentiation/genetics , Gene Silencing , Intracellular Signaling Peptides and Proteins/genetics , Phosphoproteins/genetics , Actin Cytoskeleton/metabolism , Adenoviridae , Animals , Cells, Cultured , HEK293 Cells , Humans , Rats , Signal Transduction , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
13.
EMBO J ; 41(22): e112012, 2022 11 17.
Article in English | MEDLINE | ID: mdl-36215692

ABSTRACT

Epigenome reprogramming after fertilization enables transcriptionally quiescent maternal and paternal chromatin to acquire a permissive state for subsequent zygotic genome activation (ZGA). H3K27 acetylation (H3K27ac) is a well-established chromatin marker of active enhancers and promoters. However, reprogramming dynamics of H3K27ac during maternal-to-zygotic transition (MZT) in mammalian embryos are not well-studied. By profiling the allelic landscape of H3K27ac during mouse MZT, we show that H3K27ac undergoes three waves of rapid global transitions between oocyte stage and 2-cell stage. Notably, germinal vesicle oocyte and zygote chromatin are globally hyperacetylated, with noncanonical, broad H3K27ac domains that correlate with broad H3K4 trimethylation (H3K4me3) and open chromatin. H3K27ac marks genomic regions primed for activation including ZGA genes, retrotransposons, and active alleles of imprinted genes. We show that CBP/p300 and HDAC activities play important roles in regulating H3K27ac dynamics and are essential for preimplantation development. Specifically, CBP/p300 acetyltransferase broadly deposits H3K27ac in zygotes to induce the opening of condensed chromatin at putative enhancers and ensure proper ZGA. On the contrary, HDACs revert broad H3K27ac domains to canonical domains and safeguard ZGA by preventing premature expression of developmental genes. In conclusion, coordinated activities of CBP/p300 and HDACs during mouse MZT are essential for ZGA and preimplantation development.


Subject(s)
Gene Expression Regulation, Developmental , Zygote , Mice , Animals , Zygote/metabolism , Acetylation , Blastocyst/metabolism , Chromatin/genetics , Chromatin/metabolism , Mammals/genetics
14.
Mol Cell ; 72(2): 316-327.e5, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30340023

ABSTRACT

Primary cilia are required for Smoothened to transduce vertebrate Hedgehog signals, but how Smoothened accumulates in cilia and is activated is incompletely understood. Here, we identify cilia-associated oxysterols that promote Smoothened accumulation in cilia and activate the Hedgehog pathway. Our data reveal that cilia-associated oxysterols bind to two distinct Smoothened domains to modulate Smoothened accumulation in cilia and tune the intensity of Hedgehog pathway activation. We find that the oxysterol synthase HSD11ß2 participates in the production of Smoothened-activating oxysterols and promotes Hedgehog pathway activity. Inhibiting oxysterol biosynthesis impedes oncogenic Hedgehog pathway activation and attenuates the growth of Hedgehog pathway-associated medulloblastoma, suggesting that targeted inhibition of Smoothened-activating oxysterol production may be therapeutically useful for patients with Hedgehog-associated cancers.


Subject(s)
Cilia/drug effects , Cilia/metabolism , Oxysterols/pharmacology , Animals , Cell Line , HEK293 Cells , Hedgehog Proteins/metabolism , Humans , Mice , NIH 3T3 Cells , Signal Transduction/drug effects
15.
Genes Dev ; 32(7-8): 555-567, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29654059

ABSTRACT

Although peroxisome proliferator-activated receptor-γ (PPARγ) coactivator 1α (PGC-1α) is a well-established transcriptional coactivator for the metabolic adaptation of mammalian cells to diverse physiological stresses, the molecular mechanism by which it functions is incompletely understood. Here we used in vitro binding assays, X-ray crystallography, and immunoprecipitations of mouse myoblast cell lysates to define a previously unknown cap-binding protein 80 (CBP80)-binding motif (CBM) in the C terminus of PGC-1α. We show that the CBM, which consists of a nine-amino-acid α helix, is critical for the association of PGC-1α with CBP80 at the 5' cap of target transcripts. Results from RNA sequencing demonstrate that the PGC-1α CBM promotes RNA synthesis from promyogenic genes. Our findings reveal a new conduit between DNA-associated and RNA-associated proteins that functions in a cap-binding protein surveillance mechanism, without which efficient differentiation of myoblasts to myotubes fails to occur.


Subject(s)
Nuclear Cap-Binding Protein Complex/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/chemistry , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Transcriptional Activation , Animals , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Differentiation , Humans , MCF-7 Cells , Mice , Muscle Fibers, Skeletal/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , RNA Caps/metabolism , RNA-Binding Proteins , Transcription, Genetic
16.
Dev Biol ; 505: 130-140, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37981061

ABSTRACT

The Trithorax group (trxG) proteins counteract the repressive effect of Polycomb group (PcG) complexes and maintain transcriptional memory of active states of key developmental genes. Although chromatin structure and modifications appear to play a fundamental role in this process, it is not clear how trxG prevents PcG-silencing and heritably maintains an active gene expression state. Here, we report a hitherto unknown role of Drosophila Multiple ankyrin repeats single KH domain (Mask), which emerged as one of the candidate trxG genes in our reverse genetic screen. The genome-wide binding profile of Mask correlates with known trxG binding sites across the Drosophila genome. In particular, the association of Mask at chromatin overlaps with CBP and H3K27ac, which are known hallmarks of actively transcribed genes by trxG. Importantly, Mask predominantly associates with actively transcribed genes in Drosophila. Depletion of Mask not only results in the downregulation of trxG targets but also correlates with diminished levels of H3K27ac. The fact that Mask positively regulates H3K27ac levels in flies was also found to be conserved in human cells. Strong suppression of Pc mutant phenotype by mutation in mask provides physiological relevance that Mask contributes to the anti-silencing effect of trxG, maintaining expression of key developmental genes. Since Mask is a downstream effector of multiple cell signaling pathways, we propose that Mask may connect cell signaling with chromatin mediated epigenetic cell memory governed by trxG.


Subject(s)
Chromatin , Drosophila Proteins , Animals , Humans , Chromatin/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Polycomb Repressive Complex 1/genetics , Chromosomes , Drosophila/metabolism , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , DNA-Binding Proteins/metabolism
17.
Cancer Metastasis Rev ; 43(1): 441-456, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37792222

ABSTRACT

Sam68 (Src associated in mitosis of 68 kDa) is an RNA-binding and multifunctional protein extensively characterized in numerous cellular functions, such as RNA processing, cell cycle regulation, kinase- and growth factor signaling. Recent investigations highlighted Sam68 as a primary target of a class of reverse-turn peptidomimetic drugs, initially developed as inhibitors of Wnt/ß-catenin mediated transcription. Further investigations on such compounds revealed their capacity to selectively eliminate cancer stem cell (CSC) activity upon engaging Sam68. This work highlighted previously unappreciated roles for Sam68 in the maintenance of neoplastic self-renewal and tumor-initiating functions. Here, we discuss the implication of Sam68 in tumorigenesis, where central findings support its contribution to chromatin regulation processes essential to CSCs. We also review advances in CSC-targeting drug discovery aiming to modulate Sam68 cellular distribution and protein-protein interactions. Ultimately, Sam68 constitutes a vulnerability point of CSCs and an attractive therapeutic target to impede neoplastic stemness in human tumors.


Subject(s)
Adaptor Proteins, Signal Transducing , Neoplasms , Humans , Adaptor Proteins, Signal Transducing/metabolism , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , RNA-Binding Proteins/metabolism
18.
J Cell Sci ; 136(12)2023 06 15.
Article in English | MEDLINE | ID: mdl-37314181

ABSTRACT

As one of the major acetyltransferases in mammalian cells, p300 (also known as EP300) and its highly related protein CBP (also known as CREBBP), collectively termed p300/CBP, is characterized as a key regulator in gene transcription by modulating the acetylation of histones. In recent decades, proteomic analyses have revealed that p300 is also involved in the regulation of various cellular processes by acetylating many non-histone proteins. Among the identified substrates, some are key players involved in different autophagy steps, which together establish p300 as a master regulator of autophagy. Accumulating evidence has shown that p300 activity is controlled by many distinct cellular pathways to regulate autophagy in response to cellular or environmental stimuli. In addition, several small molecules have been shown to regulate autophagy by targeting p300, suggesting that manipulation of p300 activity is sufficient for controlling autophagy. Importantly, dysfunction of p300-regulated autophagy has been implicated in a number of human disorders, such as cancer, aging and neurodegeneration, highlighting p300 as a promising target for the drug development of autophagy-related human disorders. Here, we focus on the roles of p300-mediated protein acetylation in the regulation of autophagy and discuss implications for autophagy-related human disorders.


Subject(s)
Autophagy , CREB-Binding Protein , E1A-Associated p300 Protein , Proteomics , Humans , Acetylation , Acetyltransferases , Histones , E1A-Associated p300 Protein/metabolism , CREB-Binding Protein/metabolism
19.
FASEB J ; 38(13): e23780, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38948938

ABSTRACT

Aerobic training (AT), an effective form of cardiac rehabilitation, has been shown to be beneficial for cardiac repair and remodeling after myocardial infarction (MI). The p300/CBP-associated factor (PCAF) is one of the most important lysine acetyltransferases and is involved in various biological processes. However, the role of PCAF in AT and AT-mediated cardiac remodeling post-MI has not been determined. Here, we found that the PCAF protein level was significantly increased after MI, while AT blocked the increase in PCAF. AT markedly improved cardiac remodeling in mice after MI by reducing endoplasmic reticulum stress (ERS). In vivo, similar to AT, pharmacological inhibition of PCAF by Embelin improved cardiac recovery and attenuated ERS in MI mice. Furthermore, we observed that both IGF-1, a simulated exercise environment, and Embelin protected from H2O2-induced cardiomyocyte injury, while PCAF overexpression by viruses or the sirtuin inhibitor nicotinamide eliminated the protective effect of IGF-1 in H9C2 cells. Thus, our data indicate that maintaining low PCAF levels plays an essential role in AT-mediated cardiac protection, and PCAF inhibition represents a promising therapeutic target for attenuating cardiac remodeling after MI.


Subject(s)
Myocardial Infarction , Physical Conditioning, Animal , Ventricular Remodeling , p300-CBP Transcription Factors , Animals , p300-CBP Transcription Factors/metabolism , p300-CBP Transcription Factors/antagonists & inhibitors , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Mice , Ventricular Remodeling/drug effects , Ventricular Remodeling/physiology , Male , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Endoplasmic Reticulum Stress/drug effects
20.
J Pathol ; 263(2): 242-256, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38578195

ABSTRACT

There are diverse phenotypes of castration-resistant prostate cancer, including neuroendocrine disease, that vary in their sensitivity to drug treatment. The efficacy of BET and CBP/p300 inhibitors in prostate cancer is attributed, at least in part, to their ability to decrease androgen receptor (AR) signalling. However, the activity of BET and CBP/p300 inhibitors in prostate cancers that lack the AR is unclear. In this study, we showed that BRD4, CBP, and p300 were co-expressed in AR-positive and AR-null prostate cancer. A combined inhibitor of these three proteins, NEO2734, reduced the growth of both AR-positive and AR-null organoids, as measured by changes in viability, size, and composition. NEO2734 treatment caused consistent transcriptional downregulation of cell cycle pathways. In neuroendocrine models, NEO2734 treatment reduced ASCL1 levels and other neuroendocrine markers, and reduced tumour growth in vivo. Collectively, these results show that epigenome-targeted inhibitors cause decreased growth and phenotype-dependent disruption of lineage regulators in neuroendocrine prostate cancer, warranting further development of compounds with this activity in the clinic. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
E1A-Associated p300 Protein , Receptors, Androgen , Signal Transduction , Male , Humans , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Animals , E1A-Associated p300 Protein/metabolism , E1A-Associated p300 Protein/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Mice , Xenograft Model Antitumor Assays , Bromodomain Containing Proteins , CREB-Binding Protein
SELECTION OF CITATIONS
SEARCH DETAIL