Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Protein Sci ; 33(9): e5137, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39150085

ABSTRACT

Nuclear factor erythroid 2-related factor 2 (Nrf2) is a major transcription factor that functions in maintaining redox homeostasis in cells. It mediates the transcription of cytoprotective genes in response to environmental and endogenous stresses to prevent oxidative damage. Thus, Nrf2 plays a significant role in chemoprevention. However, aberrant activation of Nrf2 has been shown to protect cancer cells from apoptosis and contribute to their chemoresistance. The interaction between Nrf2 and CBP is critical for the gene transcription activation. CBP and its homologue p300 interact with two transactivation domains in Nrf2, Neh4, and Neh5 domains through their TAZ1 and TAZ2 domains. To date, the molecular basis of this crucial interaction is not known, hindering a more detailed understanding of the regulation of Nrf2. To close this knowledge gap, we have used a set of biophysical experiments to dissect the Nrf2-CBP/p300 interactions. Structural properties of Neh4 and Neh5 and their binding with the TAZ1 and TAZ2 domains of CBP/p300 were characterized. Our results show that the Neh4 and Neh5 domains of Nrf2 are intrinsically disordered, and they both can bind the TAZ1 and TAZ2 domains of CBP/p300 with micromolar affinities. The findings provide molecular insight into the regulation of Nrf2 by CBP/p300 through multi-domain interactions.


Subject(s)
NF-E2-Related Factor 2 , Oxidative Stress , Protein Domains , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/chemistry , NF-E2-Related Factor 2/genetics , Humans , E1A-Associated p300 Protein/metabolism , E1A-Associated p300 Protein/chemistry , E1A-Associated p300 Protein/genetics , p300-CBP Transcription Factors/metabolism , p300-CBP Transcription Factors/chemistry , p300-CBP Transcription Factors/genetics , Protein Binding
2.
Biomater Sci ; 12(11): 2951-2959, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38656316

ABSTRACT

The development of targeted anti-cancer therapeutics offers the potential for increased efficacy of drugs and diagnostics. Utilizing modalities agnostic to tumor type, such as the hypoxic tumor microenvironment (TME), may assist in the development of universal tumor targeting agents. The hypoxia-inducible factor (HIF), in particular HIF1, plays a key role in tumor adaptation to hypoxia, and inhibiting its interaction with p300 has been shown to provide therapeutic potential. Using a multivalent assembled protein (MAP) approach based on the self-assembly of the cartilage oligomeric matrix protein coiled-coil (COMPcc) domain fused to the critical residues of the C-terminal transactivation domain (C-TAD) of the α subunit of HIF1 (HIF1α), we generate HIF1α-MAP (H-MAP). The resulting H-MAP demonstrates picomolar binding affinity to p300, the ability to downregulate hypoxia-inducible genes, and in vivo tumor targeting capability.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit , Protein Engineering , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Humans , Animals , Protein Domains , Mice , Cell Line, Tumor , Cartilage Oligomeric Matrix Protein/chemistry , Cartilage Oligomeric Matrix Protein/metabolism , Tumor Microenvironment , E1A-Associated p300 Protein/metabolism , E1A-Associated p300 Protein/chemistry
3.
Biochemistry ; 62(7): 1321-1329, 2023 04 04.
Article in English | MEDLINE | ID: mdl-36883372

ABSTRACT

The Myb transcription factor is involved in the proliferation of hematopoietic cells, and deregulation of its expression can lead to cancers such as leukemia. Myb interacts with various proteins, including the histone acetyltransferases p300 and CBP. Myb binds to a small domain of p300, the KIX domain (p300KIX), and inhibiting this interaction is a potential new drug discovery strategy in oncology. The available structures show that Myb binds to a very shallow pocket of the KIX domain, indicating that it might be challenging to identify inhibitors of this interaction. Here, we report the design of Myb-derived peptides which interact with p300KIX. We show that by mutating only two Myb residues that bind in or near a hotspot at the surface of p300KIX, it is possible to obtain single-digit nanomolar peptidic inhibitors of the Myb/p300KIX interaction that bind 400-fold tighter to p300KIX than wildtype Myb. These findings suggest that it might also be possible to design potent low molecular-weight compounds to disrupt the Myb/p300KIX interaction.


Subject(s)
E1A-Associated p300 Protein , Peptides , Proto-Oncogene Proteins c-myb , Peptides/pharmacology , Protein Binding , Proto-Oncogene Proteins c-myb/antagonists & inhibitors , Proto-Oncogene Proteins c-myb/chemistry , E1A-Associated p300 Protein/antagonists & inhibitors , E1A-Associated p300 Protein/chemistry
4.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35064087

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is associated with extensive dysregulation of the epigenome and epigenetic regulators, such as bromodomain and extraterminal motif (BET) proteins, have been suggested as potential targets for therapy. However, single-agent BET inhibition has shown poor efficacy in clinical trials, and no epigenetic approaches are currently used in PDAC. To circumvent the limitations of the current generation of BET inhibitors, we developed the compound XP-524 as an inhibitor of the BET protein BRD4 and the histone acetyltransferase EP300/CBP, both of which are ubiquitously expressed in PDAC tissues and cooperate to enhance tumorigenesis. XP-524 showed increased potency and superior tumoricidal activity than the benchmark BET inhibitor JQ-1 in vitro, with comparable efficacy to higher-dose JQ-1 combined with the EP300/CBP inhibitor SGC-CBP30. We determined that this is in part due to the epigenetic silencing of KRAS in vitro, with similar results observed using ex vivo slice cultures of human PDAC tumors. Accordingly, XP-524 prevented KRAS-induced, neoplastic transformation in vivo and extended survival in two transgenic mouse models of aggressive PDAC. In addition to the inhibition of KRAS/MAPK signaling, XP-524 also enhanced the presentation of self-peptide and tumor recruitment of cytotoxic T lymphocytes, though these lymphocytes remained refractory from full activation. We, therefore, combined XP-524 with an anti-PD-1 antibody in vivo, which reactivated the cytotoxic immune program and extended survival well beyond XP-524 in monotherapy. Pending a comprehensive safety evaluation, these results suggest that XP-524 may benefit PDAC patients and warrant further exploration, particularly in combination with immune checkpoint inhibition.


Subject(s)
Antineoplastic Agents/pharmacology , E1A-Associated p300 Protein/antagonists & inhibitors , Immune Checkpoint Inhibitors/pharmacology , Proteins/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Disease Models, Animal , Drug Synergism , E1A-Associated p300 Protein/chemistry , Gene Expression Regulation , Humans , Kaplan-Meier Estimate , Mice , Models, Molecular , Molecular Conformation , Molecular Structure , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/chemistry , Structure-Activity Relationship , Transcription Factors/genetics , Transcription Factors/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays
5.
Chem Commun (Camb) ; 57(88): 11709-11712, 2021 Nov 04.
Article in English | MEDLINE | ID: mdl-34693944

ABSTRACT

We demonstrate for the first time the simultaneous measurement of the acetyltransferase (HAT) and crotonyltransferase (HCT) activities of histone acetylation writer p300 by integrating antibody-based fluorescence labeling with single molecule detection. This methods exhibits good specificity and high sensitivity. Moreover, it can accurately evaluate the kinetic parameters of both the HAT and HCT activities of p300 and screen inhibitors.


Subject(s)
Acetyltransferases/analysis , E1A-Associated p300 Protein/metabolism , Acetylation , Acetyltransferases/metabolism , E1A-Associated p300 Protein/chemistry , Histones/chemistry , Histones/metabolism , Humans
6.
Nat Commun ; 12(1): 4618, 2021 07 29.
Article in English | MEDLINE | ID: mdl-34326347

ABSTRACT

The transcriptional co-activator and acetyltransferase p300 is required for fundamental cellular processes, including differentiation and growth. Here, we report that p300 forms phase separated condensates in the cell nucleus. The phase separation ability of p300 is regulated by autoacetylation and relies on its catalytic core components, including the histone acetyltransferase (HAT) domain, the autoinhibition loop, and bromodomain. p300 condensates sequester chromatin components, such as histone H3 tail and DNA, and are amplified through binding of p300 to the nucleosome. The catalytic HAT activity of p300 is decreased due to occlusion of the active site in the phase separated droplets, a large portion of which co-localizes with chromatin regions enriched in H3K27me3. Our findings suggest a model in which p300 condensates can act as a storage pool of the protein with reduced HAT activity, allowing p300 to be compartmentalized and concentrated at poised or repressed chromatin regions.


Subject(s)
Cell Nucleus/metabolism , Chromatin/metabolism , E1A-Associated p300 Protein/metabolism , Histone Acetyltransferases/metabolism , Histones/metabolism , Transcription Factors/metabolism , Acetylation , Cells, Cultured , E1A-Associated p300 Protein/chemistry , Humans , Protein Domains
7.
Mol Cell ; 79(5): 812-823.e4, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32668201

ABSTRACT

Steroid receptors activate gene transcription by recruiting coactivators to initiate transcription of their target genes. For most nuclear receptors, the ligand-dependent activation function domain-2 (AF-2) is a primary contributor to the nuclear receptor (NR) transcriptional activity. In contrast to other steroid receptors, such as ERα, the activation function of androgen receptor (AR) is largely dependent on its ligand-independent AF-1 located in its N-terminal domain (NTD). It remains unclear why AR utilizes a different AF domain from other receptors despite that NRs share similar domain organizations. Here, we present cryoelectron microscopy (cryo-EM) structures of DNA-bound full-length AR and its complex structure with key coactivators, SRC-3 and p300. AR dimerization follows a unique head-to-head and tail-to-tail manner. Unlike ERα, AR directly contacts a single SRC-3 and p300. The AR NTD is the primary site for coactivator recruitment. The structures provide a basis for understanding assembly of the AR:coactivator complex and its domain contributions for coactivator assembly and transcriptional regulation.


Subject(s)
DNA/chemistry , E1A-Associated p300 Protein/metabolism , Nuclear Receptor Coactivator 3/metabolism , Receptors, Androgen/metabolism , Cryoelectron Microscopy , DNA/metabolism , E1A-Associated p300 Protein/chemistry , HEK293 Cells , Humans , Nuclear Receptor Coactivator 3/chemistry , Nucleic Acid Conformation , Protein Conformation , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
8.
Chemistry ; 26(34): 7638-7646, 2020 Jun 18.
Article in English | MEDLINE | ID: mdl-32307728

ABSTRACT

Protein-protein interactions (PPIs) control virtually all cellular processes and have thus emerged as potential targets for development of molecular therapeutics. Peptide-based inhibitors of PPIs are attractive given that they offer recognition potency and selectivity features that are ideal for function, yet, they do not predominantly populate the bioactive conformation, frequently suffer from poor cellular uptake and are easily degraded, for example, by proteases. The constraint of peptides in a bioactive conformation has emerged as a promising strategy to mitigate against these liabilities. In this work, using peptides derived from hypoxia-inducible factor 1 (HIF-1α) together with dibromomaleimide stapling, we identify constrained peptide inhibitors of the HIF-1α/p300 interaction that are more potent than their unconstrained sequences. Contrary to expectation, the increased potency does not correlate with an increased population of an α-helical conformation in the unbound state as demonstrated by experimental circular dichroism analysis. Rather, the ability of the peptide to adopt a bioactive α-helical conformation in the p300 bound state is better supported in the constrained variant as demonstrated by molecular dynamics simulations and circular dichroism difference spectra.


Subject(s)
E1A-Associated p300 Protein/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Peptides/chemistry , Circular Dichroism , E1A-Associated p300 Protein/antagonists & inhibitors , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Protein Conformation, alpha-Helical
9.
J Biol Chem ; 295(13): 4303-4315, 2020 03 27.
Article in English | MEDLINE | ID: mdl-32098872

ABSTRACT

The E-protein transcription factors guide immune cell differentiation, with E12 and E47 (hereafter called E2A) being essential for B-cell specification and maturation. E2A and the oncogenic chimera E2A-PBX1 contain three transactivation domains (ADs), with AD1 and AD2 having redundant, independent, and cooperative functions in a cell-dependent manner. AD1 and AD2 both mediate their functions by binding to the KIX domain of the histone acetyltransferase paralogues CREB-binding protein (CBP) and E1A-binding protein P300 (p300). This interaction is necessary for B-cell maturation and oncogenesis by E2A-PBX1 and occurs through conserved ΦXXΦΦ motifs (with Φ denoting a hydrophobic amino acid) in AD1 and AD2. However, disruption of this interaction via mutation of the KIX domain in CBP/p300 does not completely abrogate binding of E2A and E2A-PBX1. Here, we determined that E2A-AD1 and E2A-AD2 also interact with the TAZ2 domain of CBP/p300. Characterization of the TAZ2:E2A-AD1(1-37) complex indicated that E2A-AD1 adopts an α-helical structure and uses its ΦXXΦΦ motif to bind TAZ2. Whereas this region overlapped with the KIX recognition region, key KIX-interacting E2A-AD1 residues were exposed, suggesting that E2A-AD1 could simultaneously bind both the KIX and TAZ2 domains. However, we did not detect a ternary complex involving E2A-AD1, KIX, and TAZ2 and found that E2A containing both intact AD1 and AD2 is required to bind to CBP/p300. Our findings highlight the structural plasticity and promiscuity of E2A-AD1 and suggest that E2A binds both the TAZ2 and KIX domains of CBP/p300 through AD1 and AD2.


Subject(s)
CREB-Binding Protein/chemistry , E1A-Associated p300 Protein/genetics , Protein Domains/genetics , Transcription Factor 3/chemistry , B-Lymphocytes/chemistry , B-Lymphocytes/metabolism , CREB-Binding Protein/genetics , CREB-Binding Protein/ultrastructure , E1A-Associated p300 Protein/chemistry , E1A-Associated p300 Protein/ultrastructure , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Homeodomain Proteins/ultrastructure , Humans , Mutation/genetics , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/ultrastructure , Protein Binding/genetics , Protein Conformation , Transcription Factor 3/genetics , Transcription Factor 3/ultrastructure
10.
J Biomol Struct Dyn ; 38(1): 1-12, 2020 01.
Article in English | MEDLINE | ID: mdl-30784357

ABSTRACT

Hypoxia-inducible factor-1 (HIF-1) is a transcription factor that plays an important role in the expression of genes, whose function is exerted through protein-protein interactions (PPIs), such as the transcriptional co-activator (CREB)-binding protein (CBP) and p300. Under hypoxic conditions, HIF-1is stabilized and translocated to CBP or p300, leading to the hypoxic response cascade. Furthermore, the PPI between HIF and p300/CBP is a potential cancer target for their role in the hypoxic response. In this study, molecular dynamics (MD) simulation was used to explore the conformational change for the p300 binding to one subunit of HIF-1, namely HIF-1α. Results indicated that HIF-1α-p300 complex was stable during MD simulation. New H-bonds were made in the intra-chain of p300 with HIF-1α binding. Inhibiting the HIF-1α-p300 interaction modulated the HIF-1α identification of selective molecules, which may indicate the target metabolic and cellular processes that enable the survival and growth of tumors in cancer chemotherapy. CAVER 3.0 results suggested that three main tunnels were present, according to helices 1, 2 and 3 of p300. To explore the unbinding pathway for HIF-1α via p300, we selected helices 1, 2 and 3 on the HIF-1α as a new ligand to explore the unbinding pathway via its own tunnel. For helix 1, R368 in p300 formed a H-bond with E816 in HIF1-α. A345 and D346 in p300 formed H-bonds with N803 in HIF-1α. A H-bond existed between K351(p300) and E789 (Hif1-α). These molecules may be the key residues in the unbinding pathway via its tunnel.Communicated by Ramaswamy H. Sarma.


Subject(s)
E1A-Associated p300 Protein/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Algorithms , Amino Acids/chemistry , Binding Sites , Catalytic Domain , E1A-Associated p300 Protein/metabolism , Humans , Hydrogen Bonding , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Structure-Activity Relationship , Zinc/chemistry
11.
Acta Pharmacol Sin ; 40(11): 1436-1447, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31097763

ABSTRACT

In advanced prostate cancer, CREB (cAMP-responsive element-binding protein) binding protein (CBP) and its homolog EP300 are highly expressed; targeting the bromodomain of CBP is a new strategy for the treatment of prostate cancer. In the current study we identified Y08197, a novel 1-(indolizin-3-yl) ethanone derivative, as a selective inhibitor of CBP/EP300 bromodomain and explored its antitumor activity against prostate cancer cell lines in vitro. In the AlphaScreen assay, we demonstrated that Y08197 dose-dependently inhibited the CBP bromodomain with an IC50 value at 100.67 ± 3.30 nM. Y08197 also exhibited high selectivity for CBP/EP300 over other bromodomain-containing proteins. In LNCaP, 22Rv1 and VCaP prostate cancer cells, treatment with Y08197 (1, 5 µM) strongly affected downstream signaling transduction, thus markedly inhibiting the expression of androgen receptor (AR)-regulated genes PSA, KLK2, TMPRSS2, and oncogenes C-MYC and ERG. Notably, Y08197 potently inhibited cell growth in several AR-positive prostate cancer cell lines including LNCaP, 22Rv1, VCaP, and C4-2B. In 22Rv1 prostate cancer cells, treatment with Y08197 (1, 4, 16 µM) dose-dependently induced G0/G1 phase arrest and apoptosis. Furthermore, treatment with Y08197 (5 µM) significantly decreased ERG-induced invasive capacity of 22Rv1 prostate cancer cells detected in wound-healing assay and cell migration assay. Taken together, CBP/EP300 inhibitor Y08197 represents a promising lead compound for development as new therapeutics for the treatment of castration-resistant prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , CREB-Binding Protein/metabolism , E1A-Associated p300 Protein/metabolism , Indolizines/pharmacology , Protein Domains/drug effects , Pyrazoles/pharmacology , CREB-Binding Protein/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , E1A-Associated p300 Protein/chemistry , Humans , Male , Prostatic Neoplasms/drug therapy , Receptors, Androgen/metabolism , Signal Transduction
12.
Mol Cell ; 74(2): 268-283.e5, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30902546

ABSTRACT

Linker histone H1 has been correlated with transcriptional inhibition, but the mechanistic basis of the inhibition and its reversal during gene activation has remained enigmatic. We report that H1-compacted chromatin, reconstituted in vitro, blocks transcription by abrogating core histone modifications by p300 but not activator and p300 binding. Transcription from H1-bound chromatin is elicited by the H1 chaperone NAP1, which is recruited in a gene-specific manner through direct interactions with activator-bound p300 that facilitate core histone acetylation (by p300) and concomitant eviction of H1 and H2A-H2B. An analysis in B cells confirms the strong dependency on NAP1-mediated H1 eviction for induction of the silent CD40 gene and further demonstrates that H1 eviction, seeded by activator-p300-NAP1-H1 interactions, is propagated over a CCCTC-binding factor (CTCF)-demarcated region through a distinct mechanism that also involves NAP1. Our results confirm direct transcriptional inhibition by H1 and establish a gene-specific H1 eviction mechanism through an activator→p300→NAP1→H1 pathway.


Subject(s)
CCCTC-Binding Factor/genetics , E1A-Associated p300 Protein/genetics , Proteins/genetics , Transcription, Genetic , Acetylation , B-Lymphocytes/chemistry , Binding Sites , CCCTC-Binding Factor/chemistry , CD40 Antigens/genetics , Chromatin/chemistry , Chromatin/genetics , E1A-Associated p300 Protein/chemistry , Histone Code , Histones/chemistry , Histones/genetics , Humans , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Nucleosomes/chemistry , Nucleosomes/genetics , Promoter Regions, Genetic , Protein Binding/genetics , Proteins/chemistry , tRNA Methyltransferases
13.
Structure ; 26(8): 1091-1100.e4, 2018 08 07.
Article in English | MEDLINE | ID: mdl-30099987

ABSTRACT

Despite high sequence homology among the p53 family members, the regulation of their transactivation potential is based on strikingly different mechanisms. Previous studies revealed that the activity of TAp63α is regulated via an autoinhibitory mechanism that keeps inactive TAp63α in a dimeric conformation. While all p73 isoforms are constitutive tetramers, their basal activity is much lower compared with tetrameric TAp63. We show that the dimeric state of TAp63α not only reduces DNA binding affinity, but also suppresses interaction with the acetyltransferase p300. Exchange of the transactivation domains is sufficient to transfer the regulatory characteristics between p63 and p73. Structure determination of the transactivation domains of p63 and p73 in complex with the p300 Taz2 domain further revealed that, in contrast to p53 and p73, p63 has a single transactivation domain. Sequences essential for stabilizing the closed dimer of TAp63α have evolved into a second transactivation domain in p73 and p53.


Subject(s)
DNA/chemistry , E1A-Associated p300 Protein/chemistry , Transcription Factors/chemistry , Transcriptional Activation , Tumor Protein p73/chemistry , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Proteins/chemistry , Amino Acid Sequence , Binding Sites , Cell Line, Tumor , Cloning, Molecular , DNA/genetics , DNA/metabolism , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Models, Molecular , Neurons , Osteoblasts , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Thermodynamics , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Protein p73/genetics , Tumor Protein p73/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
14.
Epigenetics Chromatin ; 11(1): 30, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29884215

ABSTRACT

BACKGROUND: The reported antitumor activity of the BET family bromodomain inhibitors has prompted the development of inhibitors against other bromodomains. However, the human genome encodes more than 60 different bromodomains and most of them remain unexplored. RESULTS: We report that the bromodomains of the histone acetyltransferases CREBBP/EP300 are critical to sustain the proliferation of human leukemia and lymphoma cell lines. EP300 is very abundant at super-enhancers in K562 and is coincident with sites of GATA1 and MYC occupancy. In accordance, CREBBP/EP300 bromodomain inhibitors interfere with GATA1- and MYC-driven transcription, causing the accumulation of cells in the G0/G1 phase of the cell cycle. The CREBBP/CBP30 bromodomain inhibitor CBP30 displaces CREBBP and EP300 from GATA1 and MYC binding sites at enhancers, resulting in a decrease in the levels of histone acetylation at these regulatory regions and consequently reduced gene expression of critical genes controlled by these transcription factors. CONCLUSIONS: Our data shows that inhibition of CREBBP/EP300 bromodomains can interfere with oncogene-driven transcriptional programs in cancer cells and consequently hold therapeutic potential.


Subject(s)
CREB-Binding Protein/chemistry , E1A-Associated p300 Protein/chemistry , GATA1 Transcription Factor/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Proto-Oncogene Proteins c-myc/metabolism , CREB-Binding Protein/metabolism , Cell Cycle , Cell Proliferation , E1A-Associated p300 Protein/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Protein Domains , Transcription, Genetic
15.
Biochim Biophys Acta Gen Subj ; 1862(8): 1729-1741, 2018 08.
Article in English | MEDLINE | ID: mdl-29746960

ABSTRACT

BACKGROUND: p300 (KAT3B) lysine acetyltransferase activity is modulated under different physiological and pathological contexts through the induction of trans-autoacetylation. This phenomenon is mediated by several factors, mechanisms of which are not fully understood. METHODS: Through acetyltransferase assays using full-length, baculovirus-expressed KATs, the specificity of NPM1-mediated enhancement of p300 autoacetylation was tested. Chaperone assays and tryptophan fluorescence studies were performed to evaluate the NPM1-induced protein folding. The NPM1 oligomer-defective mutant characterization was done by glutaraldehyde-crosslinking. The small-molecule inhibitor of NPM1 oligomerization was used to confirm the absolute requirement of multimeric NPM1 in vivo. Immunohistochemistry analysis of oral cancer patient samples was done to uncover the pathophysiological significance of NPM1-induced p300 autoacetylation. RESULTS: We find that the histone chaperone NPM1 is a specific inducer of p300 autoacetylation. Distinct from its histone chaperone activity, NPM1 is a molecular chaperone of p300. The biophysical experiments suggest that there is a reversible binding between NPM1 and p300 which can modulate p300 acetyltransferase activity. Disruption of NPM1 oligomerization suggests that oligomeric NPM1 is essential for the induction of p300 autoacetylation. Significantly, we observe a concomitant hyper-autoacetylation of p300 with overexpression of NPM1 in oral cancer samples. CONCLUSION: NPM1 can specifically modulate p300 acetyltransferase activity through the enhancement of autoacetylation. The molecular chaperone activity and oligomerization of NPM1 play a pivotal role in this phenomenon. GENERAL SIGNIFICANCE: NPM1 is overexpressed in several solid cancers, the significance of which is unknown. Induction of p300 autoacetylation could be the cause of NPM1-mediated tumorigenicity.


Subject(s)
E1A-Associated p300 Protein/chemistry , E1A-Associated p300 Protein/metabolism , Histones/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Protein Folding , Protein Multimerization , Tongue Neoplasms/metabolism , Acetylation , Humans , Nucleophosmin , Protein Binding , Protein Conformation , Tongue Neoplasms/pathology , Tumor Cells, Cultured
16.
ACS Chem Biol ; 13(2): 455-460, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29309117

ABSTRACT

Lysine acetyltransferases (KATs) play a critical role in the regulation of transcription and other genomic functions. However, a persistent challenge is the development of assays capable of defining KAT activity directly in living cells. Toward this goal, here we report the application of a previously reported dCas9-p300 fusion as a transcriptional reporter of KAT activity. First, we benchmark the activity of dCas9-p300 relative to other dCas9-based transcriptional activators and demonstrate its compatibility with second generation short guide RNA architectures. Next, we repurpose this technology to rapidly identify small molecule inhibitors of acetylation-dependent gene expression. These studies validate a recently reported p300 inhibitor chemotype and reveal a role for p300s bromodomain in dCas9-p300-mediated transcriptional activation. Comparison with other CRISPR-Cas9 transcriptional activators highlights the inherent ligand tunable nature of dCas9-p300 fusions, suggesting new opportunities for orthogonal gene expression control. Overall, our studies highlight dCas9-p300 as a powerful tool for studying gene expression mechanisms in which acetylation plays a causal role and provide a foundation for future applications requiring spatiotemporal control over acetylation at specific genomic loci.


Subject(s)
CRISPR-Cas Systems/genetics , E1A-Associated p300 Protein/metabolism , Acetylation , Azepines/pharmacology , Benzimidazoles/pharmacology , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Proteins/genetics , Capsid Proteins/genetics , Cytomegalovirus/genetics , E1A-Associated p300 Protein/antagonists & inhibitors , E1A-Associated p300 Protein/chemistry , E1A-Associated p300 Protein/genetics , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Hydantoins/pharmacology , Interleukin 1 Receptor Antagonist Protein/genetics , Isoxazoles/pharmacology , Protein Domains , RNA, Guide, Kinetoplastida/genetics , Recombinant Fusion Proteins , Spiro Compounds/pharmacology , Streptococcus pyogenes/enzymology , Transcription, Genetic/genetics , Transcriptional Activation/genetics , Triazoles/pharmacology
17.
J Biomol Struct Dyn ; 36(12): 3246-3264, 2018 Sep.
Article in English | MEDLINE | ID: mdl-28948877

ABSTRACT

Histone acetyltransferases (HATs) and histone deacetylases (HDACs) are enzymes that exhibit an important transcription activity. Dysfunction of these enzymes may lead to different diseases including cancer, cardiovascular, and other diseases. Therefore, these enzymes are the potential target for the generation of new therapeutics. C646 is a synthetic p300 HAT inhibitor; its structural and the electrostatic properties are the paradigm to understand its activity in the active site of p300 HAT enzyme. The docked C646 molecule in the active site forms expected key intermolecular interactions with the amino acid residues Trp1436, Tyr1467, and one water molecule (W1861); and these interactions are important for acetylation reaction. When compare the active site structure of C646 with the gas-phase structure, it is confirmed that the electron density distribution of polar bonds are highly altered, when the molecule present in the active site. In the gas-phase structure of C646, a large negative regions of electrostatic potential is found at the vicinity of O(4), O(5), and O(6) atoms; whereas, the negative region of these atoms are reduced in the active site. The molecular dynamics (MD) simulation also performed, it reveals the conformational stability and the intermolecular interactions of C646 molecule in the active site of p300.


Subject(s)
E1A-Associated p300 Protein/chemistry , Histone Acetyltransferases/chemistry , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/chemistry , Acetylation , Benzoates/chemistry , Benzoates/pharmacology , Catalytic Domain , Cell Line, Tumor , E1A-Associated p300 Protein/genetics , Histone Acetyltransferases/genetics , Histone Deacetylases/genetics , Humans , Molecular Dynamics Simulation , Protein Binding/drug effects , Protein Stability/drug effects , Pyrazoles/chemistry , Pyrazoles/pharmacology , Static Electricity
18.
Mol Cell ; 67(5): 733-743.e4, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28844863

ABSTRACT

Nuclear receptors recruit multiple coactivators sequentially to activate transcription. This "ordered" recruitment allows different coactivator activities to engage the nuclear receptor complex at different steps of transcription. Estrogen receptor (ER) recruits steroid receptor coactivator-3 (SRC-3) primary coactivator and secondary coactivators, p300/CBP and CARM1. CARM1 recruitment lags behind the binding of SRC-3 and p300 to ER. Combining cryo-electron microscopy (cryo-EM) structure analysis and biochemical approaches, we demonstrate that there is a close crosstalk between early- and late-recruited coactivators. The sequential recruitment of CARM1 not only adds a protein arginine methyltransferase activity to the ER-coactivator complex, it also alters the structural organization of the pre-existing ERE/ERα/SRC-3/p300 complex. It induces a p300 conformational change and significantly increases p300 HAT activity on histone H3K18 residues, which, in turn, promotes CARM1 methylation activity on H3R17 residues to enhance transcriptional activity. This study reveals a structural role for a coactivator sequential recruitment and biochemical process in ER-mediated transcription.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , E1A-Associated p300 Protein/metabolism , Estrogen Receptor alpha/metabolism , Guanylate Cyclase/metabolism , Nuclear Receptor Coactivator 3/metabolism , Transcription, Genetic , Acetylation , Binding Sites , CARD Signaling Adaptor Proteins/chemistry , CARD Signaling Adaptor Proteins/genetics , Cryoelectron Microscopy , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , E1A-Associated p300 Protein/chemistry , E1A-Associated p300 Protein/genetics , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/genetics , Guanylate Cyclase/chemistry , Guanylate Cyclase/genetics , HEK293 Cells , HeLa Cells , Histones/chemistry , Histones/metabolism , Humans , MCF-7 Cells , Methylation , Models, Molecular , Multiprotein Complexes , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Receptor Coactivator 3/chemistry , Nuclear Receptor Coactivator 3/genetics , Promoter Regions, Genetic , Protein Binding , Protein Interaction Domains and Motifs , Structure-Activity Relationship , Time Factors , Transcription Factors , Transcriptional Activation , Transfection
19.
J Biol Chem ; 292(28): 11873-11885, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28539359

ABSTRACT

RNA polymerase II (pol II) is required for the transcription of all protein-coding genes and as such represents a major enzyme whose activity is tightly regulated. Transcriptional initiation therefore requires numerous general transcriptional factors and cofactors that associate with pol II at the core promoter to form a pre-initiation complex. Transcription factor IIA (TFIIA) is a general cofactor that binds TFIID and stabilizes the TFIID-DNA complex during transcription initiation. Previous studies showed that TFIIA can make contact with the DNA sequence upstream or downstream of the TATA box, and that the region bound by TFIIA could overlap with the elements recognized by another factor, TFIIB, at adenovirus major late core promoter. Whether core promoters contain a DNA motif recognized by TFIIA remains unknown. Here we have identified a core promoter element upstream of the TATA box that is recognized by TFIIA. A search of the human promoter database revealed that many natural promoters contain a TFIIA recognition element (IIARE). We show that the IIARE enhances TFIIA-promoter binding and enhances the activity of TATA-containing promoters, but represses or activates promoters that lack a TATA box. Chromatin immunoprecipitation assays revealed that the IIARE activates transcription by increasing the recruitment of pol II, TFIIA, TAF4, and P300 at TATA-dependent promoters. These findings extend our understanding of the role of TFIIA in transcription, and provide new insights into the regulatory mechanism of core promoter elements in gene transcription by pol II.


Subject(s)
Gene Expression Regulation , Promoter Regions, Genetic , RNA Polymerase II/metabolism , Response Elements , TATA Box , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIIA/metabolism , Transcription Factor TFIID/metabolism , Binding Sites , Chromatin Immunoprecipitation , DNA, Recombinant , E1A-Associated p300 Protein/chemistry , E1A-Associated p300 Protein/metabolism , Genes, Reporter , HEK293 Cells , Humans , Mutagenesis, Site-Directed , Mutation , Nucleotide Motifs , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , RNA Polymerase II/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , TATA-Binding Protein Associated Factors/chemistry , TATA-Box Binding Protein/chemistry , TATA-Box Binding Protein/genetics , TATA-Box Binding Protein/metabolism , Transcription Factor TFIIA/chemistry , Transcription Factor TFIIA/genetics , Transcription Factor TFIID/chemistry , Upstream Stimulatory Factors/chemistry , Upstream Stimulatory Factors/genetics , Upstream Stimulatory Factors/metabolism
20.
Sci Rep ; 7: 42860, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28225008

ABSTRACT

The hypoxia inducible factor (HIF) pathway has been considered to be an attractive anti-cancer target. One strategy to inhibit HIF activity is through the disruption of the HIF-1α-p300 protein-protein interaction. We report herein the identification of an osmium(II) complex as the first metal-based inhibitor of the HIF-1α-p300 interaction. We evaluated the effect of complex 1 on HIF-1α signaling pathway in vitro and in cellulo by using the dual luciferase reporter assay, co-immunoprecipitation assay, and immunoblot assay. Complex 1 exhibited a dose-dependent inhibition of HRE-driven luciferase activity, with an IC50 value of 1.22 µM. Complex 1 interfered with the HIF-1α-p300 interaction as revealed by a dose-dependent reduction of p300 co-precipitated with HIF-1α as the concentration of complex 1 was increased. Complex 1 repressed the phosphorylation of SRC, AKT and STAT3, and had no discernible effect on the activity of NF-κB. We anticipate that complex 1 could be utilized as a promising scaffold for the further development of more potent HIF-1α inhibitors for anti-cancer treatment.


Subject(s)
Coordination Complexes/chemical synthesis , E1A-Associated p300 Protein/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Osmium/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Dose-Response Relationship, Drug , E1A-Associated p300 Protein/chemistry , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Mice , Molecular Structure , Phosphorylation/drug effects , Protein Binding/drug effects , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL