Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 258
Filter
1.
J Biol Chem ; 297(4): 101147, 2021 10.
Article in English | MEDLINE | ID: mdl-34520756

ABSTRACT

The heterogeneity of respirable particulates and compounds complicates our understanding of transcriptional responses to air pollution. Here, we address this by applying precision nuclear run-on sequencing and the assay for transposase-accessible chromatin sequencing to measure nascent transcription and chromatin accessibility in airway epithelial cells after wood smoke particle (WSP) exposure. We used transcription factor enrichment analysis to identify temporally distinct roles for ternary response factor-serum response factor complexes, the aryl hydrocarbon receptor (AHR), and NFκB in regulating transcriptional changes induced by WSP. Transcription of canonical targets of the AHR, such as CYP1A1 and AHRR, was robustly increased after just 30 min of WSP exposure, and we discovered novel AHR-regulated pathways and targets including the DNA methyltransferase, DNMT3L. Transcription of these genes and associated enhancers rapidly returned to near baseline by 120 min after exposure. The kinetics of AHR- and NFκB-regulated responses to WSP were distinguishable based on the timing of both transcriptional responses and chromatin remodeling, with induction of several cytokines implicated in maintaining NFκB-mediated responses through 120 min of exposure. In aggregate, our data establish a direct and primary role for AHR in mediating airway epithelial responses to WSP and identify crosstalk between AHR and NFκB signaling in controlling proinflammatory gene expression. This work also defines an integrated genomics-based strategy for deconvoluting multiplexed transcriptional responses to heterogeneous environmental exposures.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Repressor Proteins/metabolism , Signal Transduction , Smoke/adverse effects , Transcription, Genetic , Wood , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Transformed , Chromatin Assembly and Disassembly , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A1/genetics , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/genetics , Humans , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , NIH 3T3 Cells , Receptors, Aryl Hydrocarbon/genetics , Repressor Proteins/genetics
2.
Brief Bioinform ; 22(4)2021 07 20.
Article in English | MEDLINE | ID: mdl-33048117

ABSTRACT

The DNA methyltransferases (DNMTs) (DNMT3A, DNMT3B and DNMT3L) are primarily responsible for the establishment of genomic locus-specific DNA methylation patterns, which play an important role in gene regulation and animal development. However, this important protein family's binding mechanism, i.e. how and where the DNMTs bind to genome, is still missing in most tissues and cell lines. This motivates us to explore DNMTs and TF's cooperation and develop a network regularized logistic regression model, GuidingNet, to predict DNMTs' genome-wide binding by integrating gene expression, chromatin accessibility, sequence and protein-protein interaction data. GuidingNet accurately predicted methylation experimental data validated DNMTs' binding, outperformed single data source based and sparsity regularized methods and performed well in within and across tissue prediction for several DNMTs in human and mouse. Importantly, GuidingNet can reveal transcription cofactors assisting DNMTs for methylation establishment. This provides biological understanding in the DNMTs' binding specificity in different tissues and demonstrate the advantage of network regularization. In addition to DNMTs, GuidingNet achieves good performance for other chromatin regulators' binding. GuidingNet is freely available at https://github.com/AMSSwanglab/GuidingNet.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases , DNA Methylation/genetics , Gene Expression Regulation, Enzymologic , Genome, Human , Models, Biological , Protein Interaction Maps , Transcription Factors , Animals , Chromatin/genetics , Chromatin/metabolism , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/genetics , Databases, Genetic , Humans , Mice , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Best Pract Res Clin Haematol ; 33(3): 101194, 2020 09.
Article in English | MEDLINE | ID: mdl-33038988

ABSTRACT

BCR-ABL1 is in the center of chronic myeloid leukemia (CML) pathology, diagnosis and treatment, as confirmed by the success of tyrosine kinase inhibitor (TKI) therapy. However, additional mechanisms and events, many of which function independently of BCR-ABL1, play important roles, particularly in terms of leukemic stem cell (LSC) persistence, primary and secondary resistance, and disease progression. Promising therapeutic approaches aim to disrupt pathways which mediate LSC survival during successful TKI treatment, in the hope of improving long-term treatment-free-remission and perhaps provide a functional cure for some patients. Over the years through advances in sequencing technology frequent molecular aberrations in addition to BCR-ABL1 have been identified not only in advanced disease but also in chronic phase CML, often affecting epigenetic regulators such as ASXL1, DNMT3A and TET2. Analyses of serial samples have revealed various patterns of clonal evolution with some mutations preceding the BCR-ABL1 acquisition. Such mutations can be considered to be important co-factors in the pathogenesis of CML and could potentially influence therapeutic strategies in the future.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases , DNA-Binding Proteins , Epigenesis, Genetic , Fusion Proteins, bcr-abl , Gene Expression Regulation, Leukemic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Proto-Oncogene Proteins , Repressor Proteins , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Dioxygenases , Fusion Proteins, bcr-abl/biosynthesis , Fusion Proteins, bcr-abl/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics
4.
Folia Neuropathol ; 58(2): 133-142, 2020.
Article in English | MEDLINE | ID: mdl-32729292

ABSTRACT

INTRODUCTION: Meningiomas comprises of a wide variety of histological entities with heterogeneous biological behaviour and prognosis. The plethora of genetic data are yet to produce relevant biomarkers for routine use. In contrast, epigenetic alterations are less elucidated. MATERIAL AND METHODS: The expression of the key molecules involved in the two principal epigenetic systems, i.e. DNA methylation (DNA methyltransferases [DNMT-1, -3A and -3B]) and histone modification (Enhancer of Zeste homolog-2 [EZH2] and trimethyl histone-3 [H3K27me3]) were assessed in 149 cases of meningiomas (grade I - 102, grade II - 47) by immunohistochemistry. RESULTS: Immunopositivity for EZH2 (38.3% vs. 6.0%) and negativity for H3K27me3 (10.6% vs. 1.0%) were significantly more common in grade II tumours. Both were associated with significantly higher proliferative activity. The majority of the cases of both grades showed expression of all three DNMTs. However, high expression of DNMT-1 was significantly more common in grade II tumours (87.8% vs. 66.2%). Expression of EZH2 and loss of H3K27me3 were associated with significantly shorter progression-free survival (hazard ratio [HR] = 4.07 and 0.24, respectively). CONCLUSIONS: The key epigenetic regulators play important roles in the pathobiology of meningiomas. EZH2 positivity and H3K27me3 negativity are associated with aggressive tumour-biology and poor prognosis. Both these markers can easily be assessed by immunohistochemistry and can be incorporated in routine practice.


Subject(s)
Biomarkers, Tumor/genetics , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , Enhancer of Zeste Homolog 2 Protein/biosynthesis , Histones/biosynthesis , Meningeal Neoplasms/genetics , Meningioma/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Child , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Meningeal Neoplasms/mortality , Meningioma/mortality , Middle Aged , Prognosis , Progression-Free Survival , Young Adult
5.
Histochem Cell Biol ; 154(3): 301-314, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32514790

ABSTRACT

Ovarian aging is one of the main causes of female infertility, and its molecular background is still largely unknown. As DNA methylation regulates many oogenesis/folliculogenesis-related genes, the expression levels and cellular localizations of DNA methyltransferases (DNMTs) playing key roles in this process is important in the ovaries from early to aged terms. In the present study, we aimed to evaluate the spatial and temporal expression of the Dnmt1, Dnmt3a, Dnmt3b, and Dnmt3l genes as well as global DNA methylation levels in the mouse ovaries during aging. For this purpose, the following groups were created: young (1- and 2-week old; n = 3 from each week), prepubertal (3- and 4-week-old; n = 3 from each week), pubertal (5- and 6-week-old; n = 3 from each week), postpubertal (16- and 18-week-old; n = 3 from each week), and aged (52-, 60- and 72-week-old; n = 3 from each week). We found here that Dnmt1, Dnmt3a, and Dnmt3l genes' expression at mRNA and protein levels as well as global DNA methylation profiles were gradually and significantly decreased in the postnatal ovaries from young to aged groups (P < 0.05). In contrast, there was a remarkable increase of Dnmt3b expression in the pubertal, postpubertal and aged groups (P < 0.05). Our findings suggest that the significantly altered DNMT expression and global DNA methylation levels during ovarian aging may contribute to female infertility development at the later terms of lifespan. Also, new researches are required to determine the molecular biological mechanism(s) that how altered DNMT expression and decreased DNA methylation lead to ovarian aging.


Subject(s)
Aging/genetics , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , Infertility/genetics , Ovary/metabolism , Aging/metabolism , Animals , DNA (Cytosine-5-)-Methyltransferase 1/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA Methylation/genetics , DNA Methyltransferase 3A , Female , Immunohistochemistry , Infertility/metabolism , Mice , Mice, Inbred BALB C , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , DNA Methyltransferase 3B
6.
Blood Cells Mol Dis ; 82: 102417, 2020 05.
Article in English | MEDLINE | ID: mdl-32179410

ABSTRACT

OBJECTIVE: miR-194-5p and NEAT1 have been reported to be associated with multiple malignancies, but their roles in acute myeloid leukemia (AML) remains not fully understood. METHODS: Bone marrow samples were collected for monocyte separation. qRT-PCR assay was performed to investigate the expression patterns of NEAT1 and miR-194-5p in AML. CCK-8, soft agar colony formation, flow cytometry and transwell assays were employed to explore the biological functions of NEAT1 or miR-194-5p. Methylation PCR was performed to monitor the methylation of NEAT1. Luciferase reporter assay was subjected to verify the relationship between miR-194-5p and DNMT3A. Immunofluorescence and western blotting were performed to detect the alterations of protein expression. RESULTS: NEAT1 and miR-194-5p were both down-regulated in AML. Overexpression of either NEAT1 or miR-194-5p repressed proliferation, induced apoptosis and restrained migration and invasion of AML cells. There was a negative correlation between NEAT1 and DNMT3A in AML. Knockdown of DNMT3A dramatically decreased the methylation of NEAT1. Moreover, DNMT3A was identified as a downstream target of miR-194-5p. Furthermore, down-regulation of DNMT3A rescued the impacts on the malignant phenotypes of NEAT1 inhibition by miR-194-5p inhibitor. CONCLUSION: Altogether, down-regulation of NEAT1 mediated by miR-194-5p/DNMT3A axis promotes AML progression, which might provide therapeutic targets in AML treatment.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/biosynthesis , Down-Regulation , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/metabolism , MicroRNAs/biosynthesis , Neoplasm Proteins/biosynthesis , RNA, Long Noncoding/biosynthesis , RNA, Neoplasm/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Female , HEK293 Cells , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , MicroRNAs/genetics , Neoplasm Proteins/genetics , RNA, Long Noncoding/genetics , RNA, Neoplasm/genetics , THP-1 Cells
7.
Mol Microbiol ; 114(1): 127-139, 2020 07.
Article in English | MEDLINE | ID: mdl-32187735

ABSTRACT

In Caulobacter crescentus the combined action of chromosome replication and the expression of DNA methyl-transferase CcrM at the end of S-phase maintains a cyclic alternation between a full- to hemi-methylated chromosome. This transition of the chromosomal methylation pattern affects the DNA-binding properties of the transcription factor GcrA that controls the several key cell cycle functions. However, the molecular mechanism by which GcrA and methylation are linked to transcription is not fully elucidated yet. Using a combination of cell biology, genetics, and in vitro analysis, we deciphered how GcrA integrates the methylation pattern of several S-phase expressed genes to their transcriptional output. We demonstrated in vitro that transcription of ctrA from the P1 promoter in its hemi-methylated state is activated by GcrA, while in its fully methylated state GcrA had no effect. Further, GcrA and methylation together influence a peculiar distribution of creS transcripts, encoding for crescentin, the protein responsible for the characteristic shape of Caulobacter cells. This gene is duplicated at the onset of chromosome replication and the two hemi-methylated copies are spatially segregated. Our results indicated that GcrA transcribed only the copy where coding strand is methylated. In vitro transcription assay further substantiated this finding. As several of the cell cycle-regulated genes are also under the influence of methylation and GcrA-dependent transcriptional regulation, this could be a mechanism responsible for maintaining the gene transcription dosage during the S-phase.


Subject(s)
Caulobacter crescentus/genetics , DNA Methylation/genetics , Gene Expression Regulation, Bacterial/genetics , Transcription, Genetic/genetics , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA-Binding Proteins/genetics , DNA-Directed RNA Polymerases/genetics , Promoter Regions, Genetic/genetics , Sigma Factor/genetics
8.
Pathol Res Pract ; 216(4): 152848, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32051106

ABSTRACT

Piwi-interacting RNAs (piRNAs) dysregulation occurs frequently in extensive cancers. However, there was no report about piRNA expression in esophageal cancer (EC). In this study, the expression levels of piR-823 and DNMT1, DNMT3A, DNMT3B were detected in 54 pairs of ESCC tissues and adjacent normal tissues using the quantitative real-time polymerase chain reaction method. Pearson's chi-squared test and receiver operating characteristic curves were established to evaluate the diagnostic and prognostic value of piR-823 in ESCC. Spearman's correlation analysis was used to evaluate the association between piR-823 and DNMTs. We found that piR-823 was significantly upregulated in ESCC tissues compared with matched normal tissues (P = 0.0213), the level of piR-823 was significantly associated with lymph node metastasis (P = 0.042). The ROC curve analysis of piR-823 expression level yielded an area under the ROC curve value of 0.713 (P = 0.0001). DNMT3B was upregulated in ESCC tissues compared with matched normal tissues (P = 0.0286). There was an obvious positive correlation between piR-823 and DNMT3B expression (r = 0.6420, P < 0.0001). In conclusion, for the first time, we provided evidence about piRNA expression in EC. piRNA-823 and DNMT3B were both upregulated in ESCC and positively correlated with each other, suggesting the tumor oncogenic role of piR-823 in ESCC to epigenetically induce aberrant DNA methylation through DNMT3B. In addition, piRNA-823 showed high specificity in detecting ESCC and higher piRNA-823 level indicated higher risk of lymph node metastasis, suggesting its diagnostic and prognostic biomarker potential.


Subject(s)
Biomarkers, Tumor/genetics , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , Gene Expression Regulation, Neoplastic/genetics , RNA, Small Interfering/metabolism , Adult , Aged , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/metabolism , Female , Humans , Male , Middle Aged , RNA, Small Interfering/genetics , DNA Methyltransferase 3B
9.
J Clin Invest ; 130(5): 2347-2363, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31990678

ABSTRACT

Fibroblasts are key effector cells in tissue remodeling. They remain persistently activated in fibrotic diseases, resulting in progressive deposition of extracellular matrix. Although fibroblast activation may be initiated by external factors, prolonged activation can induce an "autonomous," self-maintaining profibrotic phenotype in fibroblasts. Accumulating evidence suggests that epigenetic alterations play a central role in establishing this persistently activated pathologic phenotype of fibroblasts. We demonstrated that in fibrotic skin of patients with systemic sclerosis (SSc), a prototypical idiopathic fibrotic disease, TGF-ß induced the expression of DNA methyltransferase 3A (DNMT3A) and DNMT1 in fibroblasts in a SMAD-dependent manner to silence the expression of suppressor of cytokine signaling 3 (SOCS3) by promoter hypermethylation. Downregulation of SOCS3 facilitated activation of STAT3 to promote fibroblast-to-myofibroblast transition, collagen release, and fibrosis in vitro and in vivo. Reestablishment of the epigenetic control of STAT3 signaling by genetic or pharmacological inactivation of DNMT3A reversed the activated phenotype of SSc fibroblasts in tissue culture, inhibited TGF-ß-dependent fibroblast activation, and ameliorated experimental fibrosis in murine models. These findings identify a pathway of epigenetic imprinting of fibroblasts in fibrotic disease with translational implications for the development of targeted therapies in fibrotic diseases.


Subject(s)
Epigenesis, Genetic , Myofibroblasts/metabolism , STAT3 Transcription Factor/metabolism , Scleroderma, Systemic/metabolism , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Animals , DNA (Cytosine-5-)-Methyltransferase 1/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA Methyltransferase 3A , Female , Fibrosis , Gene Expression Regulation, Enzymologic , Humans , Male , Mice , Myofibroblasts/pathology , Scleroderma, Systemic/pathology
10.
Toxicol Lett ; 322: 12-19, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-31899212

ABSTRACT

Benzene exposure is a risk factor of acute myeloid leukemia (AML), during such carcinogenesis long non-coding RNAs (lncRNAs) are important epigenetic regulators. HOTAIRM1 (HOXA transcript antisense RNA, myeloid-specific 1) plays an indispensable role in the development of AML. Hydroquinone (HQ) is one major metabolite of benzene and its ideal replacement in toxicology research. But the influence of benzene or HQ on HOTAIRM1 expression in AML associated pathway is still unclear. In the TK6 cells with short-term exposure to HQ (HQ-ST cells) or long term HQ exposure induced malignant transformed TK6 cells (HQ-MT cells), the relationship between DNMT3b and HOTAIRM1 was explored. Comparing to counterparts, HOTAIRM1 expression was increased firstly and then decreased in HQ-ST cells, and definitely decreased in HQ-MT cells; while the expression change tendency of DNMT3b was in contrast to that of HOTAIRM1. Moreover, the average HOTAIRM1 expression of 17 paired workers being exposed to benzene within 1.5 years was increased, but that of the remaining 92 paired workers with longer exposure time was decreased. Furthermore, in 5-AzaC (DNA methyltransferase inhibitor) or TSA (histone deacetylation inhibitor) treated HQ-MT cells, the expression of HOTAIRM1 was restored by reduced DNA promoter methylation levels. HQ-MT cells with DNMT3b knockout by CRISPR/Cas9 displayed the promoter hypomethylation and the increase of HOTAIRM1, also confirmed in benzene exposure workers. These suggest that long term exposure to HQ or benzene might induce the increase of DNMT3b expression and the promoter hypermethylation to silence the expression of HOTAIRM1, a possible tumor-suppressor in the AML associated carcinogenesis pathway.


Subject(s)
Benzene/adverse effects , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA Methylation/drug effects , Gene Silencing/drug effects , Hydroquinones/toxicity , Leukemia, Myeloid, Acute/chemically induced , MicroRNAs/metabolism , Occupational Diseases/chemically induced , Occupational Exposure/adverse effects , Case-Control Studies , Cell Line, Transformed , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferases/genetics , Enzyme Induction , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , MicroRNAs/genetics , Occupational Diseases/enzymology , Occupational Diseases/genetics , Promoter Regions, Genetic , Risk Assessment , DNA Methyltransferase 3B
11.
Toxicol Lett ; 319: 175-186, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31733319

ABSTRACT

Information on the effects of gibberellic acid (gibberellin A3, GA3) on ovarian follicle development is limited. In our present study, 21-day-old female Wistar rats were exposed to GA3 by gavage (25, 50, and 100 mg/kg body weight, once per day) for eight weeks to evaluate the influence of GA3 on ovarian follicle development. After treatment, significant (P < 0.05) increases (to 40.17 % and 44.5 %, respectively) in atretic follicle proportions and significant decreases (to 19.49 % and 17.86 %, respectively) in corpus luteum proportions were observed in the 50 and 100 mg/kg treatment groups compared to the control group. Significant (P < 0.05) increases (to 31.3 % and 42.0 %, respectively) in follicle apoptosis were observed in the 50 and 100 mg/kg treatment groups by transmission electron microscopy and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assays. Significantly increased expression of caspase-3, caspase-8, caspase-9 and Fas was observed by real-time PCR and Western blotting. Bisulfite sequencing PCR (BSP) revealed obviously decreased total methylation percentages of the caspase-3 promoter region in the two treatment groups. Real-time quantitative PCR also showed significantly decreased mRNA expression of DNA methyltransferase (Dnmt) 3a and Dnmt3b. Further in vitro studies showed that a DNA methylation inhibitor could enhance the GA3-induced increase in the mRNA expression of caspase-3. Overall, our present study indicates that GA3 administration from weaning until sexual maturity can affect ovarian follicle development by inducing apoptosis and suggests that signaling through the Fas-mediated apoptotic pathway may be an important underlying mechanism of this apoptosis. In addition, GA3-induced aberrant DNA methylation patterns might be partly responsible for upregulation of caspase-3 gene expression.


Subject(s)
Apoptosis/drug effects , Caspase 3/drug effects , DNA Methylation/drug effects , Fas-Associated Death Domain Protein/biosynthesis , Gibberellins/toxicity , Granulosa Cells/drug effects , Ovary/cytology , Signal Transduction/drug effects , Animals , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Fas-Associated Death Domain Protein/drug effects , Female , Ovarian Follicle/drug effects , Ovary/drug effects , Promoter Regions, Genetic/drug effects , Rats , Rats, Wistar , Superovulation/drug effects , DNA Methyltransferase 3B
12.
Pathology ; 52(2): 218-227, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31864524

ABSTRACT

DNA methyltransferases (DNMTs) regulate gene expression by methylating cytosine residues within CpG dinucleotides. Aberrant methylation patterns have been shown in a variety of human tumours including prostate cancer. However, the expression of DNMTs in clinical samples across the spectrum of prostate cancer progression has not been studied before. Tissue microarrays were constructed from the prostatectomy specimens of 309 patients across the spectrum of prostate cancer progression: hormone-naïve low-grade prostate cancer (n=49), hormone-naïve high-grade prostate cancer (n=151), hormonally treated high-grade prostate cancer (n=65), and castrate-resistant prostate cancer (CRPC) including neuroendocrine carcinoma (n=44). Adjacent non-neoplastic parenchyma was also available in 100 patients. In 71 patients with high-grade carcinoma and lymph node metastasis, tissue from the metastasis was also available for analysis. Immunohistochemical staining was performed with antibodies against DNMT1, DNMT2, DNMT3A, DNMT3B, and DNMT3L. Our results showed that DNMT1 and DNMT3L were upregulated early in prostate cancer progression, whereas DNMT2 was upregulated as a response to androgen ablation. DNMT1, DNMT3A, and DNMT3B were higher in the late stages of prostate cancer progression, i.e., the emergence of castrate resistance and androgen-independent growth. Lastly, DNMT1, DNMT2, and DNMT3L were upregulated in lymph node metastases compared to primary carcinomas. Our results highlight a cascade of epigenetic events in prostate cancer progression.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA Methylation/physiology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Aged , DNA (Cytosine-5-)-Methyltransferases/analysis , Disease Progression , Humans , Male , Middle Aged
13.
Int J Mol Sci ; 20(24)2019 Dec 13.
Article in English | MEDLINE | ID: mdl-31847128

ABSTRACT

Anoctamin1 (ANO1), a calcium activated chloride channel, is known to play a critical role in salivary secretion. In the salivary gland, ANO1 is expressed exclusively in the acinar cells, with no expression in the ductal cells. However, the mechanisms that determine this distinctive cell type-dependent expression pattern of ANO1 remain unknown. In this study, we discovered that the cell-dependent expression of ANO1 during salivary gland organogenesis is regulated by DNA methylation of ANO1 CpG islands. ANO1 CpG islands in e12 embryonic submandibular glands (eSMG) are highly methylated, but those in e14 eSMG or adult SMG are significantly unmethylated. The differential expression pattern of ANO1 in duct and acini is defined at e14. Artificial demethylation by treatment with the demethylating agent 5-aza-2'-deoxycytidine (5-Aza-CdR), induced the expression of ANO1 in both the ductal cell line Human Submandibular Gland (HSG) and in the duct cells of adult mouse SMG. During the trans-differentiation in Matrigel of duct-origin HSG cells into acinar-like phenotype, significant demethylation of ANO1 CpG islands is observed. This may be due to the reduced expression of DNA methyltransferase (DNMT) 3a and 3b. These results suggest that the differential expression of ANO1 in salivary glands during organogenesis and differentiation is mainly regulated by epigenetic demethylation of the ANO1 gene.


Subject(s)
Anoctamin-1/biosynthesis , CpG Islands , DNA Methylation , Epigenesis, Genetic , Neoplasm Proteins/biosynthesis , Salivary Glands/metabolism , Animals , Cell Line , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA Methyltransferase 3A , Decitabine/pharmacology , Humans , Mice , Salivary Glands/cytology , DNA Methyltransferase 3B
14.
J Exp Clin Cancer Res ; 38(1): 474, 2019 Nov 26.
Article in English | MEDLINE | ID: mdl-31771617

ABSTRACT

BACKGROUND: The inflammatory cytokine interleukin-6 (IL-6) is critical for the expression of octamer-binding transcription factor 4 (OCT4), which is highly associated with early tumor recurrence and poor prognosis of hepatocellular carcinomas (HCC). DNA methyltransferase (DNMT) family is closely linked with OCT4 expression and drug resistance. However, the underlying mechanism regarding the interplay between DNMTs and IL-6-induced OCT4 expression and the sorafenib resistance of HCC remains largely unclear. METHODS: HCC tissue samples were used to examine the association between DNMTs/OCT4 expression levels and clinical prognosis. Serum levels of IL-6 were detected using ELISA assays (n = 144). Gain- and loss-of-function experiments were performed in cell lines and mouse xenograft models to determine the underlying mechanism in vitro and in vivo. RESULTS: We demonstrate that levels of DNA methyltransferase 3 beta (DNMT3b) are significantly correlated with the OCT4 levels in HCC tissues (n = 144), and the OCT4 expression levels are positively associated with the serum IL-6 levels. Higher levels of IL-6, DNMT3b, or OCT4 predicted early HCC recurrence and poor prognosis. We show that IL-6/STAT3 activation increases DNMT3b/1 and OCT4 in HCC. Activated phospho-STAT3 (STAT-Y640F) significantly increased DNMT3b/OCT4, while dominant negative phospho-STAT3 (STAT-Y705F) was suppressive. Inhibiting DNMT3b with RNA interference or nanaomycin A (a selective DNMT3b inhibitor) effectively suppressed the IL-6 or STAT-Y640F-induced increase of DNMT3b-OCT4 and ALDH activity in vitro and in vivo. The fact that OCT4 regulates the DNMT1 expressions were further demonstrated either by OCT4 forced expression or DNMT1 silence. Additionally, the DNMT3b silencing reduced the OCT4 expression in sorafenib-resistant Hep3B cells with or without IL-6 treatment. Notably, targeting DNMT3b with nanaomycin A significantly increased the cell sensitivity to sorafenib, with a synergistic combination index (CI) in sorafenib-resistant Hep3B cells. CONCLUSIONS: The DNMT3b plays a critical role in the IL-6-mediated OCT4 expression and the drug sensitivity of sorafenib-resistant HCC. The p-STAT3 activation increases the DNMT3b/OCT4 which confers the tumor early recurrence and poor prognosis of HCC patients. Findings from this study highlight the significance of IL-6-DNMT3b-mediated OCT4 expressions in future therapeutic target for patients expressing cancer stemness-related properties or sorafenib resistance in HCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , Interleukin-6/metabolism , Liver Neoplasms/drug therapy , Octamer Transcription Factor-3/biosynthesis , STAT3 Transcription Factor/metabolism , Sorafenib/pharmacology , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Disease Models, Animal , Drug Resistance, Neoplasm , Female , Hep G2 Cells , Heterografts , Humans , Interleukin-6/blood , Interleukin-6/genetics , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Middle Aged , Octamer Transcription Factor-3/genetics , Prognosis , DNA Methyltransferase 3B
15.
Sci Rep ; 9(1): 17602, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31772242

ABSTRACT

DNA methylation, a central component of the epigenetic network is altered in response to nutritional influences. In one-carbon cycle, folate acts as a one-carbon carrier and vitamin B12 acts as co-factor for the enzyme methionine synthase. Both folate and vitamin B12 are the important regulators of DNA methylation which play an important role in development in early life. Previous studies carried out in this regard have shown the individual effects of these vitamins but recently the focus has been to study the combined effects of both the vitamins during pregnancy. Therefore, this study was planned to elucidate the effect of the altered dietary ratio of folate and B12 on the expression of transporters, related miRNAs and DNA methylation in C57BL/6 mice. Female mice were fed diets with 9 combinations of folate and B12 for 4 weeks. They were mated and off-springs born (F1) were continued on the same diet for 6 weeks post-weaning. Maternal and fetal (F2) tissues were collected at day 20 of gestation. Deficient state of folate led to an increase in the expression of folate transporters in both F1 and F2 generations, however, B12 deficiency (BDFN) also led to an increase in the expression in both the generations. B12 transporters/proteins were found to be increased with B12 deficiency in F1 and F2 generations except for TC-II in the kidney which was found to be decreased in the F1 generation. miR-483 was found to be increased with all conditions of folate and B12 in both F1 and F2 generations, however, deficient conditions of B12 led to an increase in the expression of miR-221 in both F1 and F2 generations. The level of miR-133 was found to be increased in BDFN group in F1 generation however; in F2 generation the change in expression was tissue and sex-specific. Global DNA methylation was decreased with deficiency of both folate and B12 in maternal tissues (F1) but increased with folate deficiency in placenta (F1) and under all conditions in fetal tissues (F2). DNA methyltransferases were overall found to be increased with deficiency of folate and B12 in both F1 and F2 generations. Results suggest that the dietary ratio of folate and B12 resulted in altered expression of transporters, miRNAs, and genomic DNA methylation in association with DNMTs.


Subject(s)
DNA Methylation , Diet , Folic Acid Deficiency/metabolism , Folic Acid/administration & dosage , Maternal Exposure , MicroRNAs/genetics , Paternal Exposure , Pregnancy Complications/metabolism , Vitamin B 12 Deficiency/metabolism , Vitamin B 12/administration & dosage , Animals , Brain/embryology , Brain/metabolism , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/genetics , Female , Fetal Growth Retardation/etiology , Fetal Growth Retardation/genetics , Fetus/metabolism , Folic Acid/blood , Folic Acid Deficiency/genetics , Gene Expression Regulation, Developmental , Homocysteine/blood , Kidney/embryology , Kidney/metabolism , Liver/embryology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , MicroRNAs/biosynthesis , Placenta/metabolism , Pregnancy , Pregnancy Complications/genetics , Reverse Transcriptase Polymerase Chain Reaction , Vitamin B 12/blood , Vitamin B 12 Deficiency/genetics
16.
Sci Rep ; 9(1): 13343, 2019 09 16.
Article in English | MEDLINE | ID: mdl-31527824

ABSTRACT

Our objectives were to determine whether clinic-pathological markers and immune-related gene signatures in breast cancer exhibit any change upon brain metastasis and whether previously reported genes significantly associated with brain metastases and the epithelial-mesenchymal transition (EMT) were reproducible and consistent in our dataset. Sixteen pair-matched samples from primary breast cancers and brain metastases diagnosed were collected from the Japan Clinical Oncology Group Breast Cancer Study Group. Gene expression profiles for immune-, brain metastases-, and EMT-related genes were compared between primary breast cancers and brain metastases. Potential therapeutic target genes of 41 FDA-approved or under-investigation agents for brain metastases were explored. Immune-related signatures exhibited significantly lower gene expression in brain metastases than in primary breast cancers. No significant differences were detected for the majority of genes associated with brain metastases and EMT in the two groups. Among 41 therapeutic target candidates, VEGFA and DNMT3A demonstrated significantly higher gene expression in brain metastases. We found that distinct patterns of gene expression exist between primary breast cancers and brain metastases. Further studies are needed to explore whether these distinct expression profiles derive from or underlie disease status and compare these features between metastases to the brain and other sites.


Subject(s)
Brain Neoplasms/genetics , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Transcriptome/genetics , Adult , Aged , Biomarkers, Tumor/genetics , Blood-Brain Barrier/physiology , Brain Neoplasms/secondary , Breast Neoplasms/pathology , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Middle Aged , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
17.
Mitochondrion ; 49: 56-65, 2019 11.
Article in English | MEDLINE | ID: mdl-31299394

ABSTRACT

Somatic mutations within mitochondrial DNA (mtDNA) encoded cytochrome c oxidase subunit I (MT-CO1 or MT-COI) are frequent in various cancer types. In addition, perturbation from orchestrated expression of mitochondrial DNA encoded genes is also associated with complex disorders, including cancer. Since codon bias and the mitochondrial translation system restricts functional characterization of over-expressed wild type or mutant mitochondrial DNA encoded genes, the codon optimization and artificial synthesis of entire MT-CO1 allowed us to over-express the wild type and one of its deleterious mutants into the mitochondria of the transfected cells. Ectopically expressed MT-CO1 was observed to efficiently express and localized to mitochondria but showed high level of aggregation under denaturing condition. Over-expression of wild type or mutant variant of MT-CO1 promoted anchorage dependent and independent proliferation potential in in-vitro experiments and introduced the cancer cell metabolic phenotype of high glucose uptake and lactate release. Reactive oxygen species generated in cells over-expressing MT-CO1 variants acted as key effectors mediating differential expression of apoptosis and DNA damage pathway related genes. High ROS generated also down-regulated the expression of global regulators of gene expression, DNMT3A and DNMT3B. The down-regulated expression of DNMTs co-related with differential methylation of the CpG islands in the promoter region of a select set of studied genes, in a manner to promote pro-cancerous phenotype. Apart from assigning the mechanistic role to the MT-CO1 variants and their perturbed expression in cancer development, the present study provides novel insights into the functional role of somatic mutations within MT-CO1 promoting cancer phenotype.


Subject(s)
Carcinogenesis/metabolism , DNA, Mitochondrial/metabolism , DNA, Neoplasm/metabolism , Ectopic Gene Expression , Electron Transport Complex IV/biosynthesis , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Mutation , Neoplasm Proteins/biosynthesis , Carcinogenesis/genetics , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , DNA, Mitochondrial/genetics , DNA, Neoplasm/genetics , Electron Transport Complex IV/genetics , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Neoplasm Proteins/genetics , DNA Methyltransferase 3B
18.
Neuropharmacology ; 157: 107679, 2019 10.
Article in English | MEDLINE | ID: mdl-31229451

ABSTRACT

Binge drinking during adolescence increases the risk for neuropsychiatric disorders including alcoholism in adulthood. DNA methylation in post-mitotic neurons is an important epigenetic modification that plays a crucial role in neurodevelopment. We examined the effects of intermittent ethanol exposure during adolescence on adult behavior and whether DNA methylation changes provide a plausible explanation for the lasting effects of this developmental insult. One hour after last adolescent intermittent ethanol (AIE), growth arrest and DNA damage inducible protein 45 (Gadd45a, Gadd45b, and Gadd45g) mRNA expression was increased and DNA methyltransferase (DNMT) activity and Dnmt3b expression was decreased in the amygdala as compared to adolescent intermittent saline (AIS) rats. However, AIE rats 24 h after last exposure displayed increased DNMT activity but normalized Gadd45 and Dnmt3b mRNA expression compared to AIS rats. In adulthood, rats exposed to AIE show increased Dnmt3b mRNA expression and DNMT activity, along with decreased Gadd45g mRNA expression in the amygdala. DNA methylation of neuropeptide Y (Npy) and brain-derived neurotrophic factor (Bdnf) exon IV is increased in the AIE adult amygdala compared to AIS adult rats. Treatment with the DNMT inhibitor 5-azacytidine (5-azaC) at adulthood normalizes the AIE-induced DNA hypermethylation of Npy and Bdnf exon IV with concomitant reversal of AIE-induced anxiety-like and alcohol-drinking behaviors. These results suggest that binge-like ethanol exposure during adolescence leads to dysregulation in DNA methylation mechanisms in the amygdala which may contribute to behavioral phenotypes of anxiety and alcohol use in adulthood.


Subject(s)
Alcohol Drinking/physiopathology , Amygdala/metabolism , Anxiety/physiopathology , DNA Methylation/physiology , Ethanol/pharmacology , Age Factors , Animals , Antigens, Differentiation/biosynthesis , Anxiety/chemically induced , Azacitidine/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Cell Cycle Proteins/biosynthesis , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , Ethanol/antagonists & inhibitors , Exons/drug effects , Intracellular Signaling Peptides and Proteins/biosynthesis , Male , Neuropeptide Y/metabolism , Rats , DNA Methyltransferase 3B , GADD45 Proteins
19.
Mol Reprod Dev ; 86(6): 614-623, 2019 06.
Article in English | MEDLINE | ID: mdl-30834655

ABSTRACT

DNA methylation is generally known to inactivate gene expression. The DNA methyltransferases (DNMTs), DNMT3A and DNMT3B, catalyze somatic cell lineage-specific DNA methylation, while DNMT3A and DNMT3L catalyze germ cell lineage-specific DNA methylation. How such lineage- and gene-specific DNA methylation patterns are created remains to be elucidated. To better understand the regulatory mechanisms underlying DNA methylation, we generated transgenic mice that constitutively expressed DNMT3A and DNMT3L, and analyzed DNA methylation, gene expression, and their subsequent impact on ontogeny. All transgenic mice were born normally but died within 20 weeks accompanied with cardiac hypertrophy. Several genes were repressed in the hearts of transgenic mice compared with those in wild-type mice. CpG islands of these downregulated genes were highly methylated in the transgenic mice. This abnormal methylation occurred in the perinatal stage. Conversely, monoallelic DNA methylation at imprinted loci was faithfully maintained in all transgenic mice, except H19. Thus, the loci preferred by DNMT3A and DNMT3L differ between somatic and germ cell lineages.


Subject(s)
Cardiomegaly/enzymology , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA Methylation , Ectopic Gene Expression , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , CpG Islands , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Female , Germ Cells/enzymology , Germ Cells/pathology , Male , Mice , Mice, Transgenic
20.
Br J Haematol ; 186(1): 91-100, 2019 07.
Article in English | MEDLINE | ID: mdl-30891745

ABSTRACT

Sickle cell disease (SCD) affects over 2 million people worldwide with high morbidity and mortality in underdeveloped countries. Therapeutic interventions aimed at reactivating fetal haemoglobin (HbF) is an effective approach for improving survival and ameliorating the clinical severity of SCD. A class of agents that inhibit DNA methyltransferase (DNMT) activity show promise as HbF inducers because off-target effects are not observed at low concentrations. However, these compounds are rapidly degraded by cytidine deaminase when taken by oral administration, creating a critical barrier to clinical development for SCD. We previously demonstrated that microRNA29B (MIR29B) inhibits de novo DNMT synthesis, therefore, the goal of our study was to determine if MIR29 mediates HbF induction. Overexpression of MIR29B in human KU812 cells and primary erythroid progenitors significantly increased the percentage of HbF positive cells, while decreasing the expression of DNMT3A and the HBG repressor MYB. Furthermore, HBG promoter methylation levels decreased significantly following MIR29B overexpression in human erythroid progenitors. We subsequently, observed higher MIR29B expression in SCD patients with higher HbF levels compared to those with low HbF. Our findings provide evidence for the ability of MIR29B to induce HbF and supports further investigation to expand treatment options for SCD.


Subject(s)
Anemia, Sickle Cell/genetics , Epigenesis, Genetic/drug effects , Fetal Hemoglobin/genetics , MicroRNAs/physiology , Transcriptional Activation/drug effects , gamma-Globins/genetics , Cell Line , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/drug effects , DNA Methyltransferase 3A , DNA Modification Methylases/biosynthesis , DNA Modification Methylases/drug effects , Erythroid Precursor Cells/metabolism , Fetal Hemoglobin/metabolism , Humans , MicroRNAs/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...