Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Mol Cell ; 49(2): 310-21, 2013 Jan 24.
Article in English | MEDLINE | ID: mdl-23201122

ABSTRACT

Differences in global levels of histone acetylation occur in normal and cancer cells, although the reason why cells regulate these levels has been unclear. Here we demonstrate a role for histone acetylation in regulating intracellular pH (pH(i)). As pH(i) decreases, histones are globally deacetylated by histone deacetylases (HDACs), and the released acetate anions are coexported with protons out of the cell by monocarboxylate transporters (MCTs), preventing further reductions in pH(i). Conversely, global histone acetylation increases as pH(i) rises, such as when resting cells are induced to proliferate. Inhibition of HDACs or MCTs decreases acetate export and lowers pH(i), particularly compromising pH(i) maintenance in acidic environments. Global deacetylation at low pH is reflected at a genomic level by decreased abundance and extensive redistribution of acetylation throughout the genome. Thus, acetylation of chromatin functions as a rheostat to regulate pH(i) with important implications for mechanism of action and therapeutic use of HDAC inhibitors.


Subject(s)
Histones/metabolism , Intracellular Fluid/metabolism , Protein Processing, Post-Translational , Acetates , Acetylation , Carbohydrate Metabolism , Chromatin , Gene Expression Regulation , Glucose/physiology , Glutamine/physiology , HeLa Cells , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Histones/genetics , Humans , Hydrogen-Ion Concentration , Hydroxamic Acids/pharmacology , Monocarboxylic Acid Transporters/metabolism , Niacinamide/pharmacology , Pyruvic Acid/metabolism , Sequence Analysis, RNA , Transcriptome
2.
J Biol Chem ; 287(38): 32006-16, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22822071

ABSTRACT

Histone deacetylases (HDACs) function in a wide range of molecular processes, including gene expression, and are of significant interest as therapeutic targets. Although their native complexes, subcellular localization, and recruitment mechanisms to chromatin have been extensively studied, much less is known about whether the enzymatic activity of non-sirtuin HDACs can be regulated by natural metabolites. Here, we show that several coenzyme A (CoA) derivatives, such as acetyl-CoA, butyryl-CoA, HMG-CoA, and malonyl-CoA, as well as NADPH but not NADP(+), NADH, or NAD(+), act as allosteric activators of recombinant HDAC1 and HDAC2 in vitro following a mixed activation kinetic. In contrast, free CoA, like unconjugated butyrate, inhibits HDAC activity in vitro. Analysis of a large number of engineered HDAC1 mutants suggests that the HDAC activity can potentially be decoupled from "activatability" by the CoA derivatives. In vivo, pharmacological inhibition of glucose-6-phosphate dehydrogenase (G6PD) to decrease NADPH levels led to significant increases in global levels of histone H3 and H4 acetylation. The similarity in structures of the identified metabolites and the exquisite selectivity of NADPH over NADP(+), NADH, and NAD(+) as an HDAC activator reveal a previously unrecognized biochemical feature of the HDAC proteins with important consequences for regulation of histone acetylation as well as the development of more specific and potent HDAC inhibitors.


Subject(s)
Gene Expression Regulation, Enzymologic , Histone Deacetylase 1/metabolism , Histone Deacetylases/metabolism , Sirtuins/chemistry , Allosteric Site , Animals , Cell Nucleus/metabolism , Chromatin/chemistry , Coenzyme A/chemistry , Epigenesis, Genetic , HeLa Cells , Histone Deacetylase 1/antagonists & inhibitors , Histones/metabolism , Humans , Insecta , Kinetics , Mutation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Subcellular Fractions/metabolism
3.
Am J Pathol ; 174(5): 1619-28, 2009 May.
Article in English | MEDLINE | ID: mdl-19349354

ABSTRACT

Cancer cells exhibit alterations in histone modification patterns at individual genes and globally at the level of single nuclei in individual cells. We demonstrated previously that lower global/cellular levels of histone H3 lysine 4 dimethylation (H3K4me2) and H3K18 acetylation (ac) predict a higher risk of prostate cancer recurrence. Here we show that the cellular levels of both H3K4me2 and H3K18ac also predict clinical outcome in both lung and kidney cancer patients, with lower levels predicting significantly poorer survival probabilities in both cancer groups. We also show that lower cellular levels of H3K9me2, a modification associated with both gene activity and repression, is also prognostic of poorer outcome for individuals with either prostate or kidney cancers. The predictive power of these histone modifications was independent of tissue-specific clinicopathological variables, the proliferation marker Ki-67, or a p53 tumor suppressor mutation. Chromatin immunoprecipitation experiments indicated that the lower cellular levels of histone modifications in more aggressive cancer cell lines correlated with lower levels of modifications at DNA repetitive elements but not with gene promoters across the genome. Our results suggest that lower global levels of histone modifications are predictive of a more aggressive cancer phenotype, revealing a surprising commonality in prognostic epigenetic patterns of adenocarcinomas of different tissue origins.


Subject(s)
Adenocarcinoma/diagnosis , Histones/metabolism , Kidney Neoplasms/diagnosis , Lung Neoplasms/diagnosis , Methylation , Prostatic Neoplasms/diagnosis , Acetylation , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adult , Aged , Aged, 80 and over , Blotting, Western , Cell Proliferation , Chromatin Immunoprecipitation , Cohort Studies , Female , Histones/genetics , Humans , Immunoenzyme Techniques , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Middle Aged , Prognosis , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Protein Processing, Post-Translational , Repetitive Sequences, Nucleic Acid , Tissue Array Analysis
4.
Science ; 321(5892): 1084-5, 2008 Aug 22.
Article in English | MEDLINE | ID: mdl-18719283

ABSTRACT

Adenovirus small early region 1a (e1a) protein drives cells into S phase by binding RB family proteins and the closely related histone acetyl transferases p300 and CBP. The interaction with RB proteins displaces them from DNA-bound E2F transcription factors, reversing their repression of cell cycle genes. However, it has been unclear how the e1a interaction with p300 and CBP promotes passage through the cell cycle. We show that this interaction causes a threefold reduction in total cellular histone H3 lysine 18 acetylation (H3K18ac). CBP and p300 are required for acetylation at this site because their knockdown causes specific hypoacetylation at H3K18. SV40 T antigen also induces H3K18 hypoacetylation. Because global hypoacetylation at this site is observed in prostate carcinomas with poor prognosis, this suggests that processes resulting in global H3K18 hypoacetylation may be linked to oncogenic transformation.


Subject(s)
Adenovirus E1A Proteins/metabolism , Adenoviruses, Human/metabolism , Cell Cycle , Histones/metabolism , Acetylation , Adenovirus E1A Proteins/genetics , Antigens, Polyomavirus Transforming/metabolism , CREB-Binding Protein/metabolism , Cell Line , Cell Transformation, Viral , Cells, Cultured , HeLa Cells , Humans , Lysine/metabolism , Mutation , p300-CBP Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...