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.
Comput Biol Chem ; 102: 107796, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36495748

ABSTRACT

Epigenetic mechanisms leading to transcriptional regulation, including DNA methylation, are frequently dysregulated in diverse cancers. Interfering with aberrant DNA methylation performed by DNA cytosine methyltransferases (DNMTs) is a clinically validated approach. In particular, the selective inhibition of the de novo DNMT3A and DNMT3B enzymes, whose expression is limited to early embryogenesis, adult stem cells, and in cancers, is particularly attractive; such selectivity is likely to attenuate the dose limiting toxicity shown by current, non-selective DNMT inhibitors. We use molecular dynamics (MD) based computational analysis to study known small molecule binders of DNMT3A, then propose reversible, tight binding, and selective inhibitors that exploit the Asn1192/Arg688 difference between the maintenance DNMT1 and DNMT3A near the active site. A similar strategy exploiting the presence of a unique active site cysteine Cys666 is used to propose DNMT3A-selective irreversible inhibitors. We report our results of relative binding energies of the known and proposed compounds estimated using MM/GBSA and umbrella sampling (US) techniques, and our evaluation of other end-point binding free energy calculation methods for these receptors. These calculations offer insight into the potential for small molecules to selectively target the active site of DNMT3A.


Subject(s)
DNA Methyltransferase 3A , Neoplasms , Adult , Humans , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA Methyltransferase 3A/antagonists & inhibitors , Methionine/genetics , Methionine/metabolism , Neoplasms/genetics , Racemethionine/metabolism , S-Adenosylmethionine/metabolism
2.
Biochim Biophys Acta Mol Basis Dis ; 1868(4): 166353, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35063646

ABSTRACT

The inactivation of tumor suppressor DOC-2/DAB2 interactive protein (DAB2IP) by epigenetic and post-transcriptional modification has been reported in multiple human malignancies. DNA methyltransferase 3A (DNMT3A) is involved in de novo establishment of DNA methylation and plays a vital role in tumorigenesis. However, whether DNMT3A can regulate colorectal cancer (CRC) progression via modulation of DAB2IP remains unclear. In this study, we revealed that DNMT3A was significantly increased in CRC, predicting a poor overall survival. Functionally, ectopic expression of DNMT3A in CRC cells enhanced cell proliferation, whereas DNMT3A knockdown had the opposite effect by inducing cell cycle arrest. Mechanistically, methylation-specific PCR (MSP) and bisulfite sequencing PCR (BSP) proved that the expression of DAB2IP was epigenetically suppressed by DNMT3A-mediated promoter methylation in CRC cells. Using dual-luciferase reporter assay and ChIP-PCR assay, we further confirmed that DNMT3A restrained the transcriptional activity of DAB2IP promoter through directly binging to it. In addition, DNMT3A could activate the MEK/ERK signaling pathway via efficiently downregulating DAB2IP. Inhibition of the MEK/ERK cascade abrogated the oncogenic effects of DNMT3A on CRC cells. In conclusion, our study demonstrates that DNMT3A facilitates CRC progression by regulating DAB2IP mediated MEK/ERK activation, providing promising targets for CRC treatment.


Subject(s)
Colorectal Neoplasms/pathology , DNA Methyltransferase 3A/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , ras GTPase-Activating Proteins/metabolism , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , DNA Methylation , DNA Methyltransferase 3A/antagonists & inhibitors , DNA Methyltransferase 3A/genetics , Humans , Prognosis , Pyridones/pharmacology , Pyrimidinones/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Survival Analysis , ras GTPase-Activating Proteins/antagonists & inhibitors , ras GTPase-Activating Proteins/genetics
3.
Immunol Lett ; 237: 17-26, 2021 09.
Article in English | MEDLINE | ID: mdl-34192561

ABSTRACT

OBJECTIVE: To understand the characteristics of DNA methyltransferase 3a (DNMT3a) in thymoma associated Myasthenia Gravis reveal its transcriptional regulator network as while as analyze the effect of DNMT3a on Rel/ nuclear factor-kappaB family (RelA/RelB) and its downstream autoimmune regulatory factor (Aire). METHODS: Tissues of 30 patients with thymoma, with or without myasthenia gravis (MG), were collected and the DNMT3a protein expression were evaluated through immunohistochemistry. We performed mRNA expression profiling microarray detection and analysis, and integrated the analysis by constructing protein-protein interaction networks and the integration with other database. We identified molecular difference between low and high DNMT3a in the thymoma by heatmap. We also performed PCR validation in thymoma tissues. The DNMT3a-shRNA plasmid was transfected into TEC cells, and these cells were treated with 5-aza-2-deoxycytidine, a blocker of DNMT3a. After the down-regulation of DNMT3a in TEC cells, the transcript and protein levels of RelA, RelB, Aire, and CHRNA3 were evaluated by western blotting. In addition, changes in gene expression profiles were screened through microarray technology. We performed differential gene analysis in the thymoma cohort by heatmap with R (v.4.3.0) software. RESULTS: In 30 matched tissue specimens, the expression of DNMT3a protein in thymoma with MG was lower than that in thymoma. Through mRNA expression profiling analysis, we constructed a co-expression network of DNMT3a and found direct interaction between IKZF1 and DNMT3a, and this co-expression relationship was overlappted with Cistrome DB database. We found up-regulation of 149 mRNAs and repression of 177 mRNAs in thymoma with MG compared with thymoma. Gene ontology and pathway analysis show the involvement of a multitude of genes in the mis-regulation of MG-related pathways. RNA interference significantly reduced the level of mRNA of DNMT3a, which proved that plasmid DNMT3a was effective. In comparison to the control group, the levels of DNMT3a, Aire, and CHRNA3 mRNA and protein in TEC cells transfected with DNMT3a-shRNA interference plasmid were significantly decreased, while the expression level of RelA and RelA/RelB was significantly increased. CONCLUSIONS: Our study reveals the DNMT3a-NF-κB pathway has a major effect on MG, and can be used as a marker for diagnosis as well as a target for MG treatment.


Subject(s)
DNA Methyltransferase 3A/biosynthesis , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Myasthenia Gravis/metabolism , NF-kappa B/biosynthesis , Neoplasm Proteins/biosynthesis , RNA Interference , Thymoma/metabolism , Thymus Gland/metabolism , Thymus Neoplasms/metabolism , Adolescent , Adult , DNA Methyltransferase 3A/antagonists & inhibitors , DNA Methyltransferase 3A/genetics , Decitabine/pharmacology , Gene Ontology , Humans , Male , Middle Aged , Myasthenia Gravis/etiology , Myasthenia Gravis/genetics , NF-kappa B/genetics , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Protein Interaction Maps , RNA, Neoplasm/genetics , RNA, Small Interfering/genetics , Receptors, Nicotinic/biosynthesis , Receptors, Nicotinic/genetics , Thymoma/complications , Thymoma/genetics , Thymus Neoplasms/complications , Thymus Neoplasms/genetics , Tissue Array Analysis , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcriptome , AIRE Protein
4.
Dev Comp Immunol ; 123: 104092, 2021 10.
Article in English | MEDLINE | ID: mdl-33819545

ABSTRACT

DNA methyltransferase, a key enzyme mediating DNA methylation, is involved in numerous processes including genomic imprinting, X chromosome inactivation, transposable element suppression, and immune defense in vertebrates. In the present study, a DNA cytosine-5-methyltransferase 3 was identified from oyster Crassostrea gigas (designed as CgDNMT3). There were a PWWP domain, a PHD domain and a DNA-methylase domain in the deduced amino acid sequences of CgDNMT3, and the conserved motifs I, IV, VI, Ⅷ, IX and X were identified in its C-terminal catalytic DNA-methylase domain. The mRNA transcripts of CgDNMT3 were detected in haemocytes, mantle, gill, adductor muscle, digestive gland and labial palp, with higher expression level in haemocytes (6.54 folds of those in gill, p < 0.01). The expression level of CgDNMT3 mRNA in haemocytes increased significantly after LPS primed (2.87 folds of that in control group, p < 0.05) in vitro or Vibrio splendidus challenging (1.94 folds of that in control group, p < 0.05) in vivo. Immunocytochemical analysis revealed that CgDNMT3 protein was distributed mainly in cytoplasm and partial in nucleus of oyster haemocytes. After CgDNMT3 was transfected and expressed in HEK293T cells, the DNA 5-methylcytosine (5-mc) level in the transfected group was significantly increased, which was 1.22 folds (p < 0.05) of the pcDNA-3.1 group. The expressions of oyster CgIL17-1, CgIL17-2 and CgIL17-5 in haemocytes increased (13.05 folds, 4.78 folds and 9.41 folds of that in control group, respectively) at 12 h after V. splendidus challenging, but the increase were significantly inhibited when the oysters were pre-treated with DNA methyltransferase inhibitor 5-Azacytidine, which were 9 folds, 1.93 folds and 3.22 folds of that in control group, respectively. These results collectively suggested that CgDNMT3 was a conserved member of DNA methyltransferase 3 family in oyster, and participated in regulating the expression of cytokines during immune response.


Subject(s)
Crassostrea/immunology , DNA Methyltransferase 3A/genetics , Vibrio Infections/immunology , Vibrio/physiology , Amino Acid Sequence , Animals , Azacitidine/pharmacology , Cloning, Molecular , DNA Methyltransferase 3A/antagonists & inhibitors , DNA Methyltransferase 3A/metabolism , HEK293 Cells , Humans , Immunity, Innate , Interleukin-17/metabolism , Organ Specificity , Protein Domains/genetics , Transcriptome , Up-Regulation
5.
Cereb Cortex ; 31(4): 1998-2012, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33230530

ABSTRACT

Emerging evidence suggests that epigenetic mechanisms regulate aberrant gene transcription in stress-associated mental disorders. However, it remains to be elucidated about the role of DNA methylation and its catalyzing enzymes, DNA methyltransferases (DNMTs), in this process. Here, we found that male rats exposed to chronic (2-week) unpredictable stress exhibited a substantial reduction of Dnmt3a after stress cessation in the prefrontal cortex (PFC), a key target region of stress. Treatment of unstressed control rats with DNMT inhibitors recapitulated the effect of chronic unpredictable stress on decreased AMPAR expression and function in PFC. In contrast, overexpression of Dnmt3a in PFC of stressed animals prevented the loss of glutamatergic responses. Moreover, the stress-induced behavioral abnormalities, including the impaired recognition memory, heightened aggression, and hyperlocomotion, were partially attenuated by Dnmt3a expression in PFC of stressed animals. Finally, we found that there were genome-wide DNA methylation changes and transcriptome alterations in PFC of stressed rats, both of which were enriched at several neural pathways, including glutamatergic synapse and microtubule-associated protein kinase signaling. These results have therefore recognized the potential role of DNA epigenetic modification in stress-induced disturbance of synaptic functions and cognitive and emotional processes.


Subject(s)
DNA Methyltransferase 3A/metabolism , Locomotion/physiology , Prefrontal Cortex/enzymology , Stress, Psychological/enzymology , Stress, Psychological/psychology , Synapses/enzymology , Animals , Chronic Disease , DNA Methyltransferase 3A/antagonists & inhibitors , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Locomotion/drug effects , Male , Mice , Phthalimides/pharmacology , Prefrontal Cortex/drug effects , Rats , Rats, Sprague-Dawley , Tryptophan/analogs & derivatives , Tryptophan/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...