Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cancer Ther ; 20(8): 1422-1430, 2021 08.
Article in English | MEDLINE | ID: mdl-34108263

ABSTRACT

The peripheral T-cell lymphomas (PTCL) could be considered the prototypical epigenetic disease. As a disease, they are uniquely sensitive to histone deacetylase (HDAC) and DNA methyltransferase (DNMT) inhibitors, both alone and in combination, are characterized by a host of mutations in epigenetic genes, and can develop spontaneously in genetically engineered murine models predicated on established recurring mutations in (RHOAG17V) and TET2, an epigenetic gene governing DNA methylation. Given the clinical benefit of HDAC inhibitors (HDACi) and hypomethlyation agents alone and in combination in PTCL, we sought to explore a mechanistic basis for these agents in PTCL. Herein, we reveal profound class synergy between HDAC and DNMT inhibitors in PTCL, and that the combination induces degrees of gene expression that are substantially different and more extensive than that observed for the single agents. A prominent signature of the combination relates to the transcriptional induction of cancer testis antigens and genes involved in the immune response. Interestingly, TBX21 and STAT4, master regulators of TH1 differentiation, were among the genes upregulated by the combination, suggesting the induction of a TH1-like phenotype. Moreover, suppression of genes involved in cholesterol metabolism and the matrisome were also identified. We believe that these data provide a strong rationale for clinical studies, and future combinations leveraging an immunoepigenetic platform.


Subject(s)
Antigens, Neoplasm/genetics , Biomarkers, Tumor/metabolism , Epigenome , Gene Expression Regulation, Neoplastic/drug effects , Immunity/genetics , Lymphoma, T-Cell/pathology , Testis/metabolism , Antimetabolites, Antineoplastic/pharmacology , Apoptosis , Azacitidine/pharmacology , Biomarkers, Tumor/genetics , Cell Proliferation , DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors , DNA Methylation , Epigenesis, Genetic , Gene Expression Profiling , Histone Deacetylase Inhibitors/pharmacology , Humans , Lymphoma, T-Cell/drug therapy , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/immunology , Male , Tumor Cells, Cultured
2.
Clin Cancer Res ; 25(17): 5271-5283, 2019 09 01.
Article in English | MEDLINE | ID: mdl-30979734

ABSTRACT

PURPOSE: Both gain-of-function enhancer of zeste homolog 2 (EZH2) mutations and inactivating histone acetyltransferases mutations, such as CREBBP and EP300, have been implicated in the pathogenesis of germinal center (GC)-derived lymphomas. We hypothesized that direct inhibition of EZH2 and histone deacetyltransferase (HDAC) would be synergistic in GC-derived lymphomas. EXPERIMENTAL DESIGN: Lymphoma cell lines (n = 21) were exposed to GSK126, an EZH2 inhibitor, and romidepsin, a pan-HDAC inhibitor. Synergy was assessed by excess over bliss. Western blot, mass spectrometry, and coimmunoprecipitation were performed. A SU-DHL-10 xenograft model was utilized to validate in vitro findings. Pretreatment RNA-sequencing of cell lines was performed. MetaVIPER analysis was used to infer protein activity. RESULTS: Exposure to GSK126 and romidepsin demonstrated potent synergy in lymphoma cell lines with EZH2 dysregulation. Combination of romidepsin with other EZH2 inhibitors also demonstrated synergy suggesting a class effect of EZH2 inhibition with romidepsin. Dual inhibition of EZH2 and HDAC led to modulation of acetylation and methylation of H3K27. The synergistic effects of the combination were due to disruption of the PRC2 complex secondary to acetylation of RbAP 46/48. A common basal gene signature was shared among synergistic lymphoma cell lines and was characterized by upregulation in chromatin remodeling genes and transcriptional regulators. This finding was supported by metaVIPER analysis which also revealed that HDAC 1/2 and DNA methyltransferase were associated with EZH2 activation. CONCLUSIONS: Inhibition of EZH2 and HDAC is synergistic and leads to the dissociation of PRC2 complex. Our findings support the clinical translation of the combination of EZH2 and HDAC inhibition in EZH2 dysregulated lymphomas.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 2/antagonists & inhibitors , Lymphoma/drug therapy , Lymphoma/genetics , Acetylation , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Methylation , Depsipeptides/administration & dosage , Drug Synergism , Enhancer of Zeste Homolog 2 Protein/genetics , Epigenesis, Genetic , Female , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/pharmacology , Humans , Indoles/administration & dosage , Lymphoma/pathology , Mice , Mice, SCID , Molecular Targeted Therapy , Pyridones/administration & dosage , Random Allocation , Xenograft Model Antitumor Assays
3.
Biochim Biophys Acta Mol Basis Dis ; 1864(11): 3708-3722, 2018 11.
Article in English | MEDLINE | ID: mdl-30251690

ABSTRACT

Nephrotic syndrome (NS), a frequent chronic kidney disease in children and young adults, is the most common phenotype associated with primary coenzyme Q10 (CoQ10) deficiency and is very responsive to CoQ10 supplementation, although the pathomechanism is not clear. Here, using a mouse model of CoQ deficiency-associated NS, we show that long-term oral CoQ10 supplementation prevents kidney failure by rescuing defects of sulfides oxidation and ameliorating oxidative stress, despite only incomplete normalization of kidney CoQ levels and lack of rescue of CoQ-dependent respiratory enzymes activities. Liver and kidney lipidomics, and urine metabolomics analyses, did not show CoQ metabolites. To further demonstrate that sulfides metabolism defects cause oxidative stress in CoQ deficiency, we show that silencing of sulfide quinone oxido-reductase (SQOR) in wild-type HeLa cells leads to similar increases of reactive oxygen species (ROS) observed in HeLa cells depleted of the CoQ biosynthesis regulatory protein COQ8A. While CoQ10 supplementation of COQ8A depleted cells decreases ROS and increases SQOR protein levels, knock-down of SQOR prevents CoQ10 antioxidant effects. We conclude that kidney failure in CoQ deficiency-associated NS is caused by oxidative stress mediated by impaired sulfides oxidation and propose that CoQ supplementation does not significantly increase the kidney pool of CoQ bound to the respiratory supercomplexes, but rather enhances the free pool of CoQ, which stabilizes SQOR protein levels rescuing oxidative stress.


Subject(s)
Antioxidants/pharmacology , Ataxia/drug therapy , Hydrogen Sulfide/metabolism , Mitochondrial Diseases/drug therapy , Muscle Weakness/drug therapy , Nephrotic Syndrome/drug therapy , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Alkyl and Aryl Transferases/genetics , Animals , Antioxidants/therapeutic use , Ataxia/complications , Ataxia/metabolism , Disease Models, Animal , HeLa Cells , Humans , Kidney/metabolism , Kidney/pathology , Metabolic Networks and Pathways/drug effects , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Diseases/complications , Mitochondrial Diseases/metabolism , Muscle Weakness/complications , Muscle Weakness/metabolism , Nephrotic Syndrome/etiology , Nephrotic Syndrome/metabolism , Nephrotic Syndrome/pathology , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Oxidoreductases Acting on Sulfur Group Donors/genetics , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Reactive Oxygen Species/metabolism , Ubiquinone/metabolism , Ubiquinone/pharmacology , Ubiquinone/therapeutic use
4.
EMBO Mol Med ; 9(1): 96-111, 2017 01.
Article in English | MEDLINE | ID: mdl-27856618

ABSTRACT

Coenzyme Q (CoQ) is an electron acceptor for sulfide-quinone reductase (SQR), the first enzyme of the hydrogen sulfide oxidation pathway. Here, we show that lack of CoQ in human skin fibroblasts causes impairment of hydrogen sulfide oxidation, proportional to the residual levels of CoQ. Biochemical and molecular abnormalities are rescued by CoQ supplementation in vitro and recapitulated by pharmacological inhibition of CoQ biosynthesis in skin fibroblasts and ADCK3 depletion in HeLa cells. Kidneys of Pdss2kd/kd mice, which only have ~15% residual CoQ concentrations and are clinically affected, showed (i) reduced protein levels of SQR and downstream enzymes, (ii) accumulation of hydrogen sulfides, and (iii) glutathione depletion. These abnormalities were not present in brain, which maintains ~30% residual CoQ and is clinically unaffected. In Pdss2kd/kd mice, we also observed low levels of plasma and urine thiosulfate and increased blood C4-C6 acylcarnitines. We propose that impairment of the sulfide oxidation pathway induced by decreased levels of CoQ causes accumulation of sulfides and consequent inhibition of short-chain acyl-CoA dehydrogenase and glutathione depletion, which contributes to increased oxidative stress and kidney failure.


Subject(s)
Ataxia/physiopathology , Mitochondrial Diseases/physiopathology , Muscle Weakness/physiopathology , Sulfides/metabolism , Ubiquinone/deficiency , Alkyl and Aryl Transferases/deficiency , Animals , Cells, Cultured , Fibroblasts/metabolism , Humans , Mice , Mice, Knockout , Oxidation-Reduction , Quinone Reductases/analysis
5.
Mol Cancer Res ; 4(10): 769-78, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17050670

ABSTRACT

Transcription factor p53 regulates its target genes through binding to DNA consensus sequence and activating the promoters of its downstream genes. The conventional p53 consensus binding sequence was defined as two copies of the 10-bp motif 5'-PuPuPuC(A/T)(T/A)GPyPyPy-3' with a spacer of 0 to 13 bp, which exists in the regulatory regions of some p53 target genes. However, there is no such p53 consensus sequence in the promoters of a number of p53-responsive genes, suggesting that there might be other mechanisms whereby p53 transactivates the promoters of its target genes. We report here that p53 uses a novel binding mechanism to regulate the transcription of epithelial cell kinase (ECK), a receptor protein-tyrosine kinase implicated in signal transduction. We show that p53 binds to a 10-bp perfect palindromic decanucleotide (GTGACGTCAC) in the ECK promoter, activates the ECK promoter, and increases the transcription of ECK. This palindrome is required for p53-mediated transactivation of the ECK promoter. ECK is highly responsive to oxidative damage that leads to cell death. Ectopic expression of ECK causes spontaneous apoptosis in breast cancer cells. We found that ectopic expression of a mutant ECK fails to induce apoptosis in cancer cells. Our findings show that p53 is a transcriptional regulator of ECK in mediating apoptosis. The discovery of the novel p53-binding motif in the promoter may lead to the identification of a new class of p53 target genes.


Subject(s)
Apoptosis , Receptor, EphA2/metabolism , Signal Transduction , Transcription, Genetic , Tumor Suppressor Protein p53/physiology , Amino Acid Motifs , Base Sequence , Binding Sites , Cell Line, Tumor , Cell Survival , Gene Expression Regulation , Humans , Molecular Sequence Data , Oxidative Stress , Promoter Regions, Genetic , Receptor, EphA2/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...