Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Int J Mol Sci ; 22(16)2021 Aug 18.
Article in English | MEDLINE | ID: mdl-34445616

ABSTRACT

Neovascularization restores blood flow recovery after ischemia in peripheral arterial disease. The main two components of neovascularization are angiogenesis and arteriogenesis. Both of these processes contribute to functional improvements of blood flow after occlusion. However, discriminating between the specific contribution of each process is difficult. A frequently used model for investigating neovascularization is the murine hind limb ischemia model (HLI). With this model, it is difficult to determine the role of angiogenesis, because usually the timing for the sacrifice of the mice is chosen to be optimal for the analysis of arteriogenesis. More importantly, the occurring angiogenesis in the distal calf muscles is probably affected by the proximally occurring arteriogenesis. Therefore, to understand and subsequently intervene in the process of angiogenesis, a model is needed which investigates angiogenesis without the influence of arteriogenesis. In this study we evaluated the in vivo Matrigel plug assay in genetic deficient mice to investigate angiogenesis. Mice deficient for interferon regulatory factor (IRF)3, IRF7, RadioProtective 105 (RP105), Chemokine CC receptor CCR7, and p300/CBP-associated factor (PCAF) underwent the in vivo Matrigel model. Histological analysis of the Matrigel plugs showed an increased angiogenesis in mice deficient of IRF3, IRF7, and RP105, and a decreased angiogenesis in PCAF deficient mice. Our results also suggest an involvement of CCR7 in angiogenesis. Comparing our results with results of the HLI model found in the literature suggests that the in vivo Matrigel plug assay is superior in evaluating the angiogenic response after ischemia.


Subject(s)
Antigens, CD/physiology , Hindlimb/blood supply , Interferon Regulatory Factor-3/physiology , Interferon Regulatory Factor-7/physiology , Ischemia/pathology , Neovascularization, Pathologic/pathology , p300-CBP Transcription Factors/physiology , Animals , Collagen , Drug Combinations , Hindlimb/pathology , Ischemia/metabolism , Laminin , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/metabolism , Proteoglycans , Recovery of Function
2.
J Autoimmun ; 123: 102709, 2021 09.
Article in English | MEDLINE | ID: mdl-34304080

ABSTRACT

Chromatin remodeling, and a persistent histone 3 lysine 27 acetylation (H3K27ac) in particular, are associated with a sustained inflammatory response of synovial fibroblasts (SF) in rheumatoid arthritis (RA). Here we investigated individual functions of the writers of H3K27ac marks, the homologues histone acetyl transferases (HAT) CBP and p300, in controlling the constitutive and inflammatory gene expression in RA SF. We applied a silencing strategy, followed by RNA-sequencing and pathway analysis, complemented with the treatment of SF with inhibitors targeting the HAT (C646) or bromo domains (I-CBP) of CBP and p300. We showed that CBP and p300 undertook overlapping and, in particular at gene levels, distinct regulatory functions in SF. p300 is the major HAT for H3K27ac in SF and regulated more diverse pathways than CBP. Whereas both factors regulated genes associated with extracellular matrix remodeling, adhesion and proliferation, p300 specifically controlled developmental genes associated with limb development. Silencing of CBP specifically down regulated the TNF-induced expression of interferon-signature genes. In contrast, silencing of p300 resulted in anti- and pro-inflammatory effects. Integration of data sets derived from RNA-sequencing and chromatin immunoprecipitation sequencing for H3K27ac revealed that changes in gene expression after CBP or p300 silencing could be only partially explained by changes in levels of H3K27ac. Inhibition of CBP/p300 using HAT and bromo domain inhibitors strongly mirrored effects obtained by silencing of p300, including anti- and pro-inflammatory effects, indicating that such inhibitors are not sufficient to be used as anti-inflammatory drugs.


Subject(s)
CREB-Binding Protein/physiology , Inflammation/etiology , p300-CBP Transcription Factors/physiology , Aged , Aged, 80 and over , Animals , CREB-Binding Protein/antagonists & inhibitors , Cell Proliferation , Chromatin Assembly and Disassembly , Extracellular Matrix/physiology , Extremities/embryology , Female , Fibroblasts/physiology , Humans , Male , Middle Aged , Synoviocytes/physiology , p300-CBP Transcription Factors/antagonists & inhibitors
3.
Clin Cancer Res ; 27(7): 2087-2099, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33495313

ABSTRACT

PURPOSE: Abiraterone acetate (AA), an inhibitor of cytochrome P450 17alpha-hydroxylase/17, 20 lyase, is an FDA-approved drug for advanced prostate cancer. However, not all patients respond to AA, and AA resistance ultimately develops in patients who initially respond. We aimed to identify AA resistance mechanisms in prostate cancer cells. EXPERIMENTAL DESIGN: We established several AA-resistant cell lines and performed a comprehensive study on mechanisms involved in AA resistance development. RNA sequencing and phospho-kinase array screenings were performed to discover that the cAMP-response element CRE binding protein 1 (CREB1) was a critical molecule in AA resistance development. RESULTS: The drug-resistant cell lines are phenotypically stable without drug selection, and exhibit permanent global gene expression changes. The phosphorylated CREB1 (pCREB1) is increased in AA-resistant cell lines and is critical in controlling global gene expression. Upregulation of pCREB1 desensitized prostate cancer cells to AA, while blocking CREB1 phosphorylation resensitized AA-resistant cells to AA. AA treatment increases intracellular cyclic AMP (cAMP) levels, induces kinases activity, and leads to the phosphorylation of CREB1, which may subsequently augment the essential role of the CBP/p300 complex in AA-resistant cells because AA-resistant cells exhibit a relatively higher sensitivity to CBP/p300 inhibitors. Further pharmacokinetics studies demonstrated that AA significantly synergizes with CBP/p300 inhibitors in limiting the growth of prostate cancer cells. CONCLUSIONS: Our studies suggest that AA treatment upregulates pCREB1, which enhances CBP/p300 activity, leading to global gene expression alterations, subsequently resulting in drug resistance development. Combining AA with therapies targeting resistance mechanisms may provide a more effective treatment strategy.


Subject(s)
Abiraterone Acetate/therapeutic use , Cyclic AMP Response Element-Binding Protein/metabolism , Prostatic Neoplasms/drug therapy , p300-CBP Transcription Factors/physiology , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Male , Phosphorylation , Prostatic Neoplasms/pathology
4.
Sci Rep ; 10(1): 1817, 2020 02 04.
Article in English | MEDLINE | ID: mdl-32019948

ABSTRACT

Mesenchymal stem cells (MSCs) specifically differentiate into cardiomyocytes as a potential way to reverse myocardial injury diseases, and uncovering this differentiation mechanism is immensely important. We have previously shown that histone acetylation/methylation and DNA methylation are involved in MSC differentiation into cardiomyocytes induced by islet-1. These modifications regulate cardiac-specific genes by interacting with each other in the promoter regions of these genes, but the molecular mechanism of these interactions remains unknown. In this study, we found that the key enzymes that regulate GATA4/Nkx2.5 expression are Gcn5/HDAC1, G9A, and DNMT-1. When α-methylene-γ-butyrolactone 3 (MB-3) was used to inhibit Gcn5 expression, we observed that the interactions among these key enzymes in the GATA4/Nkx2.5 promoters were blocked, and MSCs could not be induced into cardiomyocytes. Our results indicated that islet-1 could induce Gcn5 binding to GATA4/Nkx2.5 promoter regions and induce the interactions among Gcn5, HDAC1, G9A and DNMT-1, which upregulated GATA4/Nkx2.5 expression and promoted MSC differentiation into cardiomyocytes.


Subject(s)
Cell Differentiation , LIM-Homeodomain Proteins/physiology , Mesenchymal Stem Cells/physiology , Myocytes, Cardiac/physiology , Transcription Factors/physiology , p300-CBP Transcription Factors/physiology , Acetylation , Animals , Blotting, Western , Cell Differentiation/physiology , Chromatin Immunoprecipitation , DNA Methylation , GATA4 Transcription Factor/metabolism , Histone Deacetylase 1/metabolism , Histones/metabolism , Immunoprecipitation , LIM-Homeodomain Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Myocytes, Cardiac/metabolism , Promoter Regions, Genetic , Transcription Factors/metabolism , p300-CBP Transcription Factors/metabolism
5.
Endocr Relat Cancer ; 27(3): 187-198, 2020 03.
Article in English | MEDLINE | ID: mdl-31951590

ABSTRACT

Administration of the microtubule inhibitor docetaxel is a common treatment for metastatic castration-resistant prostate cancer (mCRPC) and results in prolonged patient overall survival. Usually, after a short period of time chemotherapy resistance emerges and there is urgent need to find new therapeutic targets to overcome therapy resistance. The lysine-acetyltransferase p300 has been correlated to prostate cancer (PCa) progression. Here, we aimed to clarify a possible function of p300 in chemotherapy resistance and verify p300 as a target in chemoresistant PCa. Immunohistochemistry staining of tissue samples revealed significantly higher p300 protein expression in patients who received docetaxel as a neoadjuvant therapy compared to control patients. Elevated p300 expression was confirmed by analysis of publicly available patient data, where significantly higher p300 mRNA expression was found in tissue of mCRPC tumors of docetaxel-treated patients. Consistently, docetaxel-resistant PCa cells showed increased p300 protein expression compared to docetaxel-sensitive counterparts. Docetaxel treatment of PCa cells for 72 h resulted in elevated p300 expression. shRNA-mediated p300 knockdown did not alter colony formation efficiency in docetaxel-sensitive cells, but significantly reduced clonogenic potential of docetaxel-resistant cells. Downregulation of p300 in docetaxel-resistant cells also impaired cell migration and invasion. Taken together, we showed that p300 is upregulated by docetaxel, and our findings suggest that p300 is a possible co-target in treatment of chemoresistant PCa.


Subject(s)
Docetaxel/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , p300-CBP Transcription Factors/physiology , Cell Line, Tumor , Cell Movement/drug effects , Drug Resistance, Neoplasm , Humans , Male , Prostatic Neoplasms, Castration-Resistant/pathology , Up-Regulation , p300-CBP Transcription Factors/analysis , p300-CBP Transcription Factors/antagonists & inhibitors , p300-CBP Transcription Factors/genetics
6.
Exp Gerontol ; 126: 110690, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31419472

ABSTRACT

SKN-1/Nrf transcription factors regulate diverse biological processes essentially stress defense, detoxification, and longevity. Studies in model organisms have identified a broad range of regulatory processes and mechanisms that profoundly influence SKN-1/Nrf functions. Defining the mechanisms how SKN-1 is regulated will provide insight how cells defend against diverse stressors contributing to aging and disease. In this study, we demonstrate a crucial role for the acetyltransferase CBP-1, the C. elegans homolog of mammalian CREB-binding protein CBP/p300 in the activation of SKN-1. cbp-1 is essential for tolerance of oxidative stress and normal lifespan. CBP-1 directly interacts with SKN-1 and increases SKN-1 protein abundance. In particular CBP-1 modulates SKN-1 nuclear translocation under basal conditions and in response to stress and promotes SKN-1-dependent transcription of protective genes. Moreover, CBP-1 is required for SKN-1 nuclear recruitment, transcriptional activity, and longevity due to reduced insulin/IGF-1-like signaling, mTOR-, and GSK-3 signaling. Our findings establish the acetyltransferase CBP-1 as a critical activator of SKN-1 that directly modulates SKN-1 protein stability, nuclear localization, and function to ascertain normal stress response and lifespan.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/metabolism , DNA-Binding Proteins/metabolism , Histone Acetyltransferases/physiology , Transcription Factors/metabolism , Transcription Factors/physiology , p300-CBP Transcription Factors/physiology , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Cell Nucleus/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation/physiology , Longevity/physiology , Oxidative Stress/physiology , Transcription Factors/genetics
7.
Int J Biol Sci ; 15(6): 1276-1286, 2019.
Article in English | MEDLINE | ID: mdl-31223286

ABSTRACT

The transcriptional coactivator CREB-binding protein (CBP) and p300 are adenoviral E1A-binding proteins involved in various cellular processes, including embryonic development, homeostasis, cell differentiation and transcription activation. Previous study suggested that synthetic lethality between CBP and p300 inhibition in lung and hematopoietic cancers. However, the underlying mechanism of CBP and p300 paralog in bladder cancer remains unknown. In this study, we discovered that combined CBP and p300 inhibition impaired cell proliferation and induced apoptosis of bladder cancer cells and normal bladder urothelial cell via decreasing c-Myc expression. Then, we employed the dCas9-KRAB system, hTERT promoter and hUPII promoter to construct an CRISPR interference system which could specifically repress CBP and p300 expression and cause lethality in bladder cancer cells in vitro. The CRISPR interference system we constructed could specifically inhibit the progression of bladder cancer, providing a novel strategy to fight against bladder cancer.


Subject(s)
CREB-Binding Protein/physiology , Clustered Regularly Interspaced Short Palindromic Repeats/physiology , Urinary Bladder Neoplasms/pathology , p300-CBP Transcription Factors/physiology , Apoptosis , CREB-Binding Protein/antagonists & inhibitors , CREB-Binding Protein/metabolism , Cell Line, Tumor , Cell Proliferation , Disease Progression , Humans , Synthetic Lethal Mutations , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/therapy , p300-CBP Transcription Factors/antagonists & inhibitors , p300-CBP Transcription Factors/metabolism
8.
Cancer Res ; 79(7): 1331-1342, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30705122

ABSTRACT

C/EBPß is a key mediator of cancer-induced skeletal muscle wasting. However, the signaling mechanisms that activate C/EBPß in the cancer milieu are poorly defined. Here, we report cancer-induced muscle wasting requires the transcriptional cofactor p300, which is critical for the activation of C/EBPß. Conditioned media from diverse types of tumor cells as well as recombinant HSP70 and HSP90 provoked rapid acetylation of C/EBPß in myotubes, particularly at its Lys39 residue. Overexpression of C/EBPß with mutated Lys39 impaired Lewis lung carcinoma (LLC)-induced activation of the C/EBPß-dependent catabolic response, which included upregulation of E3 ligases UBR2 and atrogin1/MAFbx, increased LC3-II, and loss of muscle proteins both in myotubes and mouse muscle. Silencing p300 in myotubes or overexpressing a dominant negative p300 mutant lacking acetyltransferase activity in mouse muscle attenuated LLC tumor-induced muscle catabolism. Administration of pharmacologic p300 inhibitor C646, but not PCAF/GCN5 inhibitor CPTH6, spared LLC tumor-bearing mice from muscle wasting. Furthermore, mice with muscle-specific p300 knockout were resistant to LLC tumor-induced muscle wasting. These data suggest that p300 is a key mediator of LLC tumor-induced muscle wasting whose acetyltransferase activity may be targeted for therapeutic benefit in this disease. SIGNIFICANCE: These findings demonstrate that tumor-induced muscle wasting in mice is abrogated by knockout, mutation of Lys39 or Asp1399, and pharmacologic inhibition of p300.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/7/1331/F1.large.jpg.


Subject(s)
Cachexia/physiopathology , Carcinoma, Lewis Lung/pathology , p300-CBP Transcription Factors/physiology , Animals , CCAAT-Enhancer-Binding Protein-beta/chemistry , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line , HEK293 Cells , Humans , Lysine/metabolism , Male , Mice , Mice, Knockout , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , p300-CBP Transcription Factors/genetics
9.
Cell ; 175(1): 186-199.e19, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30220457

ABSTRACT

Mutations or aberrant upregulation of EZH2 occur frequently in human cancers, yet clinical benefits of EZH2 inhibitor (EZH2i) remain unsatisfactory and limited to certain hematological malignancies. We profile global posttranslational histone modification changes across a large panel of cancer cell lines with various sensitivities to EZH2i. We report here oncogenic transcriptional reprogramming mediated by MLL1's interaction with the p300/CBP complex, which directs H3K27me loss to reciprocal H3K27ac gain and restricts EZH2i response. Concurrent inhibition of H3K27me and H3K27ac results in transcriptional repression and MAPK pathway dependency in cancer subsets. In preclinical models encompassing a broad spectrum of EZH2-aberrant solid tumors, a combination of EZH2 and BRD4 inhibitors, or a triple-combination including MAPK inhibition display robust efficacy with very tolerable toxicity. Our results suggest an attractive precision treatment strategy for EZH2-aberrant tumors on the basis of tumor-intrinsic MLL1 expression and concurrent inhibition of epigenetic crosstalk and feedback MAPK activation.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/genetics , Histone-Lysine N-Methyltransferase/physiology , Myeloid-Lymphoid Leukemia Protein/physiology , Animals , Carcinogenesis/genetics , Cell Cycle Proteins , Cell Line, Tumor , Epigenesis, Genetic/genetics , Epigenomics/methods , Female , Gene Expression Regulation, Neoplastic/genetics , Histone Code/drug effects , Histone Code/genetics , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Histones/physiology , Humans , MAP Kinase Signaling System , Male , Mice , Mice, Inbred BALB C , Mice, SCID , Mutation , Myeloid-Lymphoid Leukemia Protein/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , Polycomb Repressive Complex 2/metabolism , Transcription Factors/metabolism , Transcription Factors/physiology , Transcriptional Activation , Xenograft Model Antitumor Assays/methods , p300-CBP Transcription Factors/physiology
10.
Birth Defects Res ; 110(14): 1118-1128, 2018 08 15.
Article in English | MEDLINE | ID: mdl-30114346

ABSTRACT

BACKGROUND: Maternal diabetes related neural tube defects (NTDs) are a result of oxidative stress and apoptosis. However, the molecular mechanism behind the pathogenesis is not fully understood. Here, we report that high glucose exposure-induced epigenetic changes influence histone H4 acetylation and neuroepithelial cell proliferation. We also show that the acetyltransferase inhibitor C646 can prevent high glucose induced changes in histone H4 acetylation and neuroepithelial cell proliferation. METHODS: By using LC-MS/MS as an unbiased approach, we screened the histone acetylation profile in an E9 neuroepithelial cell line (NE-4C) under high glucose exposure. We further explored the mechanism in cells in vitro and in maternal diabetes-induced mouse embryos in vivo. RESULTS: We identified 35 core histone acetylation marks in normal E9 neuroepithelial cells, whereas high glucose exposure resulted in novel acetylation sites on H4K31 and H4K44. Acetylation levels of embryonic development associated H4K5/K8/K12/K16 increased in neuroepithelial cells exposed to high glucose in vitro and in brain tissue from maternal diabetes induced exencephalic embryos in vivo. Further, mRNA level of histone acetyltransferase CBP encoded gene Crebbp was significantly increased both in vitro and in vivo. The addition of C646, a selective inhibitor for CBP/p300, significantly rescued increase of H4K5/K8/K12/K16 acetylation levels and H3S10pi-labeled neuroepithelial cell proliferation induced by high glucose exposure. CONCLUSION: Our data provide complementary insights for potential mechanisms of maternal diabetes induced NTDs.


Subject(s)
Benzoates/metabolism , Neuroepithelial Cells/drug effects , Pyrazoles/metabolism , p300-CBP Transcription Factors/antagonists & inhibitors , Acetylation , Animals , Apoptosis/drug effects , Benzoates/pharmacology , Cell Line , Cell Proliferation/drug effects , Chromatography, Liquid/methods , Epigenesis, Genetic , Female , Glucose/adverse effects , Glucose/metabolism , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Male , Mice , Nitrobenzenes , Peptide Fragments/antagonists & inhibitors , Pregnancy , Pyrazoles/pharmacology , Pyrazolones , Sialoglycoproteins/antagonists & inhibitors , Tandem Mass Spectrometry/methods , p300-CBP Transcription Factors/physiology
11.
Nucleic Acids Res ; 45(9): 5086-5099, 2017 May 19.
Article in English | MEDLINE | ID: mdl-28158851

ABSTRACT

The cyclin-dependent kinase inhibitor p27Kip1 (p27) also behaves as a transcriptional repressor. Data showing that the p300/CBP-associated factor (PCAF) acetylates p27 inducing its degradation suggested that PCAF and p27 could collaborate in the regulation of transcription. However, this possibility remained to be explored. We analyzed here the transcriptional programs regulated by PCAF and p27 in the colon cancer cell line HCT116 by chromatin immunoprecipitation sequencing (ChIP-seq). We identified 269 protein-encoding genes that contain both p27 and PCAF binding sites being the majority of these sites different for PCAF and p27. PCAF or p27 knock down revealed that both regulate the expression of these genes, PCAF as an activator and p27 as a repressor. The double knock down of PCAF and p27 strongly reduced their expression indicating that the activating role of PCAF overrides the repressive effect of p27. We also observed that the transcription factor Pax5 interacts with both p27 and PCAF and that the knock down of Pax5 induces the expression of p27/PCAF target genes indicating that it also participates in the transcriptional regulation mediated by p27/PCAF. In summary, we report here a previously unknown mechanism of transcriptional regulation mediated by p27, Pax5 and PCAF.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/physiology , Gene Expression Regulation , PAX5 Transcription Factor/physiology , p300-CBP Transcription Factors/physiology , Animals , Binding Sites , Cell Line, Tumor , Cells, Cultured , Chromatin/metabolism , HCT116 Cells , Humans , MCF-7 Cells , Mice , Protein Binding , Proteins/genetics , Tissue Array Analysis , Transcription, Genetic
12.
J Biol Chem ; 292(7): 2815-2829, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28053092

ABSTRACT

Differentiation therapy with all-trans-retinoic acid (ATRA) improves the treatment outcome of acute promyelocytic leukemia (APL); however, the molecular mechanism by which ATRA induces granulocytic differentiation remains unclear. We previously reported that the inhibition of the NAD-dependent histone deacetylase (HDAC) SIRT2 induces granulocytic differentiation in leukemia cells, suggesting the involvement of protein acetylation in ATRA-induced leukemia cell differentiation. Herein, we show that p300/CREB-binding protein-associated factor (PCAF), a histone acetyltransferase (HAT), is a prerequisite for ATRA-induced granulocytic differentiation in leukemia cells. We found that PCAF expression was markedly increased in leukemia cell lines (NB4 and HL-60) and primary APL cells during ATRA-induced granulocytic differentiation. Consistent with these results, the expression of PCAF was markedly up-regulated in the bone marrow cells of APL patients who received ATRA-containing chemotherapy. The knockdown of PCAF inhibited ATRA-induced granulocytic differentiation in leukemia cell lines and primary APL cells. Conversely, the overexpression of PCAF induced the expression of the granulocytic differentiation marker CD11b at the mRNA level. Acetylome analysis identified the acetylated proteins after ATRA treatment, and we found that histone H3, a known PCAF acetylation substrate, was preferentially acetylated by the ATRA treatment. Furthermore, we have demonstrated that PCAF is required for the acetylation of histone H3 on the promoter of ATRA target genes, such as CCL2 and FGR, and for the expression of these genes in ATRA-treated leukemia cells. These results strongly support our hypothesis that PCAF is induced and activated by ATRA, and the subsequent acetylation of PCAF substrates promotes granulocytic differentiation in leukemia cells. Targeting PCAF and its downstream acetylation targets could serve as a novel therapeutic strategy to overcome all subtypes of AML.


Subject(s)
Cell Differentiation/physiology , Granulocytes/drug effects , Leukemia, Myeloid, Acute/pathology , Tretinoin/pharmacology , p300-CBP Transcription Factors/physiology , Acetylation , CD11b Antigen/genetics , Cell Differentiation/drug effects , Gene Knockdown Techniques , Granulocytes/pathology , HL-60 Cells , Histones/metabolism , Humans , p300-CBP Transcription Factors/genetics
13.
Nucleic Acids Res ; 44(22): 10662-10675, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27613418

ABSTRACT

HOXB9 is a homeobox domain-containing transcription factor, playing an important role in embryonic development and cancer progression. However, the precise post-translational modifications (PTMs) of HOXB9 and the corresponding roles are unclear. Here, we report that acetyltransferase p300/CBP-associated factor (PCAF) interacts with and acetylates HOXB9 both in vivo and in vitro Conversely, the acetylation of HOXB9 can be reversed by deacetylase SIRT1. Furthermore, we found that HOXB9 is acetylated at lysine 27 (AcK27). Functionally, in contrast to the wild type HOXB9, AcK27-HOXB9 decreased its capacity in promoting lung cancer cell migration and tumor growth in mice. Mechanistically, AcK27-HOXB9 suppresses the transcription of its target gene Jumonji domain-containing protein 6 (JMJD6) by direct occupying the promoter of JMJD6 gene. For clinical relevance, elevated HOXB9 acetylation at K27 predicts a better prognosis in lung adenocarcinoma patients. Taken together, we identified the first PTM of HOXB9 by demonstrating that HOXB9 can be acetylated and AcK27-HOXB9 counteracts the role of the wild-type HOXB9 in regulating lung adenocarcinoma progression.


Subject(s)
Adenocarcinoma/metabolism , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Lung Neoplasms/metabolism , p300-CBP Transcription Factors/physiology , A549 Cells , Acetylation , Adenocarcinoma/genetics , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Amino Acid Sequence , Animals , Cell Movement , Disease Progression , HEK293 Cells , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Kaplan-Meier Estimate , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Protein Processing, Post-Translational , Sirtuin 1/metabolism , Transcription, Genetic
14.
Oncotarget ; 7(34): 55789-55810, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27322556

ABSTRACT

Lysine acetylation is a post-translational modification that regulates gene transcription by targeting histones as well as a variety of transcription factors in the nucleus. Recently, several reports have demonstrated that numerous cytosolic proteins are also acetylated and that this modification, affecting protein activity, localization and stability has profound consequences on their cellular functions. Interestingly, most non-histone proteins targeted by acetylation are relevant for tumorigenesis. In this review, we will analyze the functional implications of lysine acetylation in different cellular compartments, and will examine our current understanding of lysine acetyltransferases family, highlighting the biological role and prognostic value of these enzymes and their substrates in cancer. The latter part of the article will address challenges and current status of molecules targeting lysine acetyltransferase enzymes in cancer therapy.


Subject(s)
Lysine Acetyltransferases/physiology , Lysine/metabolism , Neoplasms/drug therapy , Acetylation , Acetyltransferases/physiology , Animals , Biomarkers , Humans , Lysine Acetyltransferases/antagonists & inhibitors , Mice , Microtubule Proteins , Neoplasms/metabolism , Prognosis , Protein Processing, Post-Translational , p300-CBP Transcription Factors/physiology
15.
Mol Biol Cell ; 27(15): 2479-92, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27251061

ABSTRACT

Developmental morphogenesis and tumor progression require a transient or stable breakdown of epithelial junctional complexes to permit programmed migration, invasion, and anoikis resistance, characteristics endowed by the epithelial-mesenchymal transition (EMT). The epithelial master-regulatory transcription factor Grainyhead-like 2 (GRHL2) suppresses and reverses EMT, causing a mesenchymal-epithelial transition to the default epithelial phenotype. Here we investigated the role of GRHL2 in tubulogenesis of Madin-Darby canine kidney cells, a process requiring transient, partial EMT. GRHL2 was required for cystogenesis, but it suppressed tubulogenesis in response to hepatocyte growth factor. Surprisingly, GRHL2 suppressed this process by inhibiting the histone acetyltransferase coactivator p300, preventing the induction of matrix metalloproteases and other p300-dependent genes required for tubulogenesis. A 13-amino acid region of GRHL2 was necessary for inhibition of p300, suppression of tubulogenesis, and interference with EMT. The results demonstrate that p300 is required for partial or complete EMT occurring in tubulogenesis or tumor progression and that GRHL2 suppresses EMT in both contexts through inhibition of p300.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , p300-CBP Transcription Factors/metabolism , Animals , Cadherins/metabolism , Cell Line, Tumor , Dogs , Epithelial-Mesenchymal Transition/physiology , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Humans , Madin Darby Canine Kidney Cells , Morphogenesis , Transcriptional Activation , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/physiology
16.
Oncotarget ; 7(12): 15135-49, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26934656

ABSTRACT

De novo fatty acid (FA) synthesis is required for prostate cancer (PCa) survival and progression. As a key enzyme for FA synthesis fatty acid synthase (FASN) is often overexpressed in human prostate cancers and its expression correlates with worse prognosis and poor survival. P300 is an acetyltransferase that acts as a transcription co-activator. Increasing evidence suggests that P300 is a major PCa promoter, although the underlying mechanism remains poorly understood. Here, we demonstrated that P300 binds to and increases histone H3 lysine 27 acetylation (H3K27Ac) in the FASN gene promoter. We provided evidence that P300 transcriptionally upregulates FASN expression and promotes lipid accumulation in human PCa cells in culture and Pten knockout prostate tumors in mice. Pharmacological inhibition of P300 decreased FASN expression and lipid droplet accumulation in PCa cells. Immunohistochemistry analysis revealed that expression of P300 protein positively correlates with FASN protein levels in a cohort of human PCa specimens. We further showed that FASN is a key mediator of P300-induced growth of PCa cells in culture and in mice. Together, our findings demonstrate P300 as a key factor that regulates FASN expression, lipid accumulation and cell growth in PCa. They also suggest that this regulatory pathway can serve as a new therapeutic target for PCa treatment.


Subject(s)
Biomarkers, Tumor/metabolism , Fatty Acid Synthases/metabolism , Gene Expression Regulation, Enzymologic , Lipid Metabolism , PTEN Phosphohydrolase/physiology , Prostatic Neoplasms/pathology , p300-CBP Transcription Factors/physiology , Aged , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Fatty Acid Synthases/genetics , Female , Follow-Up Studies , Humans , Male , Mice , Mice, Knockout , Mice, Nude , Middle Aged , Prognosis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
Cancer Lett ; 375(1): 190-198, 2016 May 28.
Article in English | MEDLINE | ID: mdl-26945969

ABSTRACT

UNLABELLED: The p300-CBP-associated factor (PCAF), other than its histone acetyltransferase (HAT) activity, possesses an intrinsic ubiquitination activity that is involved in various transcriptional regulators, including the transcription factor glioma-associated oncogene 1 (Gli1), a well-known regulator of epithelial-mesenchymal transition (EMT) in cancer. In present research, we detected that PCAF was down-regulated in hepatocellular carcinoma (HCC) tissues compared with the adjacent non-tumor tissues and significantly associated with malignant portal vein invasion (p < 0.05) and poor survival (p < 0.05) of HCC patients. Moreover, functional study demonstrated that downregulation of PCAF facilitated tumor cell migration, invasion via EMT. Further study found that Gli1 as a direct target of PCAF induced EMT and promoted tumor metastasis and invasion. CONCLUSION: PCAF is an anti-oncogene that plays an important role in the development of HCC by suppressing HCC cell metastasis and EMT by targeting Gli1, which indicates the potential therapeutic value of PCAF for suppression of metastasis of HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Epithelial-Mesenchymal Transition , Liver Neoplasms/metabolism , Lung Neoplasms/metabolism , Transcription Factors/metabolism , p300-CBP Transcription Factors/physiology , Animals , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/secondary , Cell Line, Tumor , Female , Humans , Kaplan-Meier Estimate , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Male , Mice, Nude , Middle Aged , Neoplasm Transplantation , Zinc Finger Protein GLI1
18.
J Mol Biol ; 427(7): 1644-54, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25688804

ABSTRACT

Heat shock factor 1 (HSF1) is critical for defending cells from both acute and chronic stresses. In aging cells, the DNA binding activity of HSF1 deteriorates correlating with the onset of pathological events including neurodegeneration and heart disease. We find that DNA binding by HSF1 is controlled by lysine deacetylases with HDAC7, HDAC9, and SIRT1 distinctly increasing the magnitude and length of a heat shock response (HSR). In contrast, HDAC1 inhibits HSF1 in a deacetylase-independent manner. In aging cells, the levels of HDAC1 are elevated and the HSR is impaired, yet reduction of HDAC1 in aged cells restores the HSR. Our results provide a mechanistic basis for the age-associated regulation of the HSR. Besides HSF1, the deacetylases differentially modulate the activities of unrelated DNA binding proteins. Taken together, our data further support the model that lysine deacetylases are selective regulators of DNA binding proteins.


Subject(s)
Cellular Senescence/physiology , DNA-Binding Proteins/metabolism , Heat-Shock Response , Histone Deacetylase 1/physiology , Transcription Factors/metabolism , Acetylation , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , HEK293 Cells , HeLa Cells , Heat Shock Transcription Factors , Heat-Shock Response/genetics , Humans , Lysine/metabolism , Mice , Transcription Factors/genetics , p300-CBP Transcription Factors/physiology
19.
Cancer Res ; 74(6): 1870-1880, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24480624

ABSTRACT

Overexpression of the histone acetyltransferase p300 is implicated in the proliferation and progression of prostate cancer, but evidence of a causal role is lacking. In this study, we provide genetic evidence that this generic transcriptional coactivator functions as a positive modifier of prostate tumorigenesis. In a mouse model of PTEN deletion-induced prostate cancer, genetic ablation of p300 attenuated expression of the androgen receptor (AR). This finding was confirmed in human prostate cancer cells in which PTEN expression was abolished by RNA interference-mediated attenuation. These results were consistent with clinical evidence that the expression of p300 and AR correlates positively in human prostate cancer specimens. Mechanistically, PTEN inactivation increased AR phosphorylation at serine 81 (Ser81) to promote p300 binding and acetylation of AR, thereby precluding its polyubiquitination and degradation. In support of these findings, in PTEN-deficient prostate cancer in the mouse, we found that p300 was crucial for AR target gene expression. Taken together, our work identifies p300 as a molecular determinant of AR degradation and highlights p300 as a candidate target to manage prostate cancer, especially in cases marked by PTEN loss.


Subject(s)
Carcinogenesis/metabolism , PTEN Phosphohydrolase/genetics , Prostatic Intraepithelial Neoplasia/enzymology , Prostatic Neoplasms/enzymology , Receptors, Androgen/metabolism , p300-CBP Transcription Factors/physiology , Aged , Animals , Cell Line, Tumor , Cell Proliferation , Female , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Middle Aged , PTEN Phosphohydrolase/deficiency , Phosphorylation , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/pathology , Proteolysis , Transcription, Genetic , Ubiquitination
20.
Dev Biol ; 385(2): 230-41, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24247009

ABSTRACT

The mechanisms that regulate appropriate genesis and differentiation of interneurons in the developing mammalian brain are of significant interest not only because interneurons play key roles in the establishment of neural circuitry, but also because when they are deficient, this can cause epilepsy. In this regard, one genetic syndrome that is associated with deficits in neural development and epilepsy is Rubinstein-Taybi Syndrome (RTS), where the transcriptional activator and histone acetyltransferase CBP is mutated and haploinsufficient. Here, we have asked whether CBP is necessary for the appropriate genesis and differentiation of interneurons in the murine forebrain, since this could provide an explanation for the epilepsy that is associated with RTS. We show that CBP is expressed in neural precursors within the embryonic medial ganglionic eminence (MGE), an area that generates the vast majority of interneurons for the cortex. Using primary cultures of MGE precursors, we show that knockdown of CBP causes deficits in differentiation of these precursors into interneurons and oligodendrocytes, and that overexpression of CBP is by itself sufficient to enhance interneuron genesis. Moreover, we show that levels of the neurotransmitter synthesis enzyme GAD67, which is expressed in inhibitory interneurons, are decreased in the dorsal and ventral forebrain of neonatal CBP(+/-) mice, indicating that CBP plays a role in regulating interneuron development in vivo. Thus, CBP normally acts to ensure the differentiation of appropriate numbers of forebrain interneurons, and when its levels are decreased, this causes deficits in interneuron development, providing a potential explanation for the epilepsy seen in individuals with RTS.


Subject(s)
Cell Differentiation/physiology , Interneurons/cytology , Prosencephalon/cytology , p300-CBP Transcription Factors/physiology , Animals , Haploinsufficiency , Mice , Polymerase Chain Reaction , Prosencephalon/embryology , p300-CBP Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...