Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.895
Filter
1.
Discov Med ; 36(184): 971-980, 2024 May.
Article in English | MEDLINE | ID: mdl-38798256

ABSTRACT

OBJECTIVE: Tissue inhibitors of matrix metalloproteinases (TIMPs) are prognostic markers in cancers. However, the role of TIMPs in DNA methylation during invasive pituitary adenoma (PA) remains unclear. The purpose of this study was to assess the effects of TIMP2 and TIMP3 promoter demethylation on the proliferation, migration, and invasion of invasive PA cells. METHODS: Methylation-specific polymerase chain reaction (PCR), quantitative PCR, and western blots were used to analyze the promoter methylation and expression of TIMP1-3. Cell counting kit-8 (CCK-8), wound healing, and transwell assays were carried out to determine the effects of TIMP2 and TIMP3 demethylation. RESULTS: TIMP1-3 showed downregulated expression in invasive PA tissues and cell lines (p < 0.05). The low expression of TIMP1-3 was due to promoter methylation of these genes (p < 0.05). The results showed that downregulation of TIMP2 and TIMP3 can promote cell proliferation, migration, and invasion (p < 0.05), whereas overexpression of TIMP2 and TIMP3 can inhibit cell proliferation, migration, and invasion (p < 0.05). After treatment with 5-azacytidine (5-AzaC), the cell activity decreased, the proliferation rate decreased, and the invasion ability weakened (p < 0.05). Treatment with 5-AzaC increased TIMP2 and TIMP3 expression and decreased DNA (cytosine-5-)-methyltransferase 1 (DNMT1), DNMT3a, and DNMT3b expression (p < 0.05). CONCLUSIONS: We showed that DNA methylation causes the silencing of TIMP2 and TIMP3 in invasive PA, it can also lead to malignant cell proliferation and cause pathological changes, whereas the use of 5-AzaC can inhibit the methylation process and can inhibit cell proliferation. Our results provide a novel method for clinical diagnosis and prevention of invasive PA.


Subject(s)
Adenoma , Cell Movement , Cell Proliferation , DNA Methylation , Neoplasm Invasiveness , Pituitary Neoplasms , Tissue Inhibitor of Metalloproteinase-2 , Tissue Inhibitor of Metalloproteinase-3 , Humans , Tissue Inhibitor of Metalloproteinase-3/genetics , Tissue Inhibitor of Metalloproteinase-3/metabolism , Cell Proliferation/genetics , Cell Proliferation/drug effects , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Pituitary Neoplasms/metabolism , Cell Movement/genetics , Cell Movement/drug effects , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism , Adenoma/genetics , Adenoma/pathology , Adenoma/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Male , Female , Promoter Regions, Genetic/genetics , Middle Aged , Adult , Azacitidine/pharmacology , DNA Methyltransferase 3A/metabolism
2.
Biomed Res Int ; 2024: 9625043, 2024.
Article in English | MEDLINE | ID: mdl-38807916

ABSTRACT

Epigenetic alterations have been observed in many hematological malignancies, including acute myeloid leukemia (AML). Many of these alterations result from mutations in DNA methyl transferase (DNMT) enzymes, disabling them to methylate target genes in a proper way. In this case-control study, we investigated the association between R882H mutation in DNMT3A gene and DDX43 gene methylation in patients with AML. 47 AML patients and 6 controls were included in this study. After DNA extraction, amplification refractory mutation system (ARMS)-PCR was used to evaluate R882H mutations in DNMT3A gene. The high-resolution melting (HRM) method was used to determine the methylation changes of the DDX43 gene promoter. R882H mutation was only found in 10.6% (5 out of 47) of AML patients. The frequency of DDX43 gene methylation was significantly higher in patients without R882H mutations compared to patients with R882H mutations (P < 0.05). The DNMT3A R882H mutation is typically present in a minority of AML patients. Nevertheless, this mutation is associated with a reduced frequency of methylation in the DDX43 promoter region.


Subject(s)
DEAD-box RNA Helicases , DNA (Cytosine-5-)-Methyltransferases , DNA Methylation , DNA Methyltransferase 3A , Leukemia, Myeloid, Acute , Mutation , Promoter Regions, Genetic , Humans , Leukemia, Myeloid, Acute/genetics , DNA Methyltransferase 3A/genetics , Promoter Regions, Genetic/genetics , DEAD-box RNA Helicases/genetics , DNA Methylation/genetics , Male , DNA (Cytosine-5-)-Methyltransferases/genetics , Female , Middle Aged , Adult , Mutation/genetics , Aged , Case-Control Studies , Neoplasm Proteins
3.
J Am Coll Cardiol ; 83(18): 1717-1727, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38692825

ABSTRACT

BACKGROUND: The expansion of hematopoietic stem cells caused by acquired somatic mutations (clonal hematopoiesis [CH]) is a novel cardiovascular risk factor. The prognostic value of CH in patients with carotid atherosclerosis remains to be evaluated. OBJECTIVES: This study assessed the prognostic significance of CH in patients with atherosclerosis as detected by ultrasound of the carotid artery. METHODS: We applied deep sequencing of selected genomic regions within the genes DNMT3A, TET2, ASXL1, and JAK2 to screen for CH in 968 prospectively collected patients with asymptomatic carotid atherosclerosis evaluated by duplex sonography. RESULTS: We detected clonal markers at variant allele frequency ≥2% in 133 (13.7%) of 968 patients (median age 69.2 years), with increasing prevalence at advanced age. Multivariate analyses including age and established cardiovascular risk factors revealed overall presence of CH to be significantly associated with increased risk of cardiovascular death (HR: 1.50; 95% CI: 1.12-2.00; P = 0.007), reflected also at the single gene level. The effect of CH was more pronounced in older patients and independent of the patients' inflammatory status as measured by high-sensitivity C-reactive protein. Simultaneous assessment of CH and degree of carotid stenosis revealed combined effects on cardiovascular mortality, depicted by a superior risk for patients with >50% stenosis and concomitant CH (adjusted HR: 1.60; 95% CI: 1.08-2.38; P = 0.020). CONCLUSIONS: CH status in combination with the extent of carotid atherosclerosis jointly predict long-term mortality. Determination of CH can provide additional prognostic information in patients with asymptomatic carotid atherosclerosis.


Subject(s)
Carotid Stenosis , Clonal Hematopoiesis , Janus Kinase 2 , Humans , Male , Female , Aged , Clonal Hematopoiesis/genetics , Carotid Stenosis/genetics , Carotid Stenosis/complications , Carotid Stenosis/diagnostic imaging , Middle Aged , DNA Methyltransferase 3A , Dioxygenases , Prospective Studies , DNA-Binding Proteins/genetics , Repressor Proteins/genetics , Proto-Oncogene Proteins/genetics , Prognosis , Cardiovascular Diseases/genetics , Cardiovascular Diseases/mortality , DNA (Cytosine-5-)-Methyltransferases/genetics
4.
FASEB J ; 38(10): e23690, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38795327

ABSTRACT

Alterations to gene transcription and DNA methylation are a feature of many liver diseases including fatty liver disease and liver cancer. However, it is unclear whether the DNA methylation changes are a cause or a consequence of the transcriptional changes. It is even possible that the methylation changes are not required for the transcriptional changes. If DNA methylation is just a minor player in, or a consequence of liver transcriptional change, then future studies in this area should focus on other systems such as histone tail modifications. To interrogate the importance of de novo DNA methylation, we generated mice that are homozygous mutants for both Dnmt3a and Dnmt3b in post-natal liver. These mice are viable and fertile with normal sized livers. Males, but not females, showed increased adipose depots, yet paradoxically, improved glucose tolerance on both control diet and high-fat diets (HFD). Comparison of the transcriptome and methylome with RNA sequencing and whole-genome bisulfite sequencing in adult hepatocytes revealed that widespread loss of methylation in CpG-rich regions in the mutant did not induce loss of homeostatic transcriptional regulation. Similarly, extensive transcriptional changes induced by HFD did not require de novo DNA methylation. The improved metabolic phenotype of the Dnmt3a/3b mutant mice may be mediated through the dysregulation of a subset of glucose and fat metabolism genes which increase both glucose uptake and lipid export by the liver. However, further work is needed to confirm this.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases , DNA Methylation , DNA Methyltransferase 3A , DNA Methyltransferase 3B , Diet, High-Fat , Glucose Intolerance , Liver , Animals , Male , Diet, High-Fat/adverse effects , Liver/metabolism , Mice , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A/metabolism , Glucose Intolerance/metabolism , Glucose Intolerance/genetics , Female , Mice, Inbred C57BL
5.
Gene ; 920: 148531, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38705424

ABSTRACT

DNA methyltransferases (DNMTs) are important epigenetic modification during spermatogenesis. To further evaluate the pattern of DNMTs in horse testes during development, we investigated the expression and localization of DNMT1, DNMT3a and DNMT3b at different time points. The qRT-PCR results showed that DNMT1 expression was maintained in testes tissue from 6-month-old (0.5y) to 2-year-old (2y) of age and decreased after 3-year-old (3y) (P < 0.01). The expression levels of DNMT3a and DNMT3b peaked in testes tissue at 3y (P < 0.01). At 4-year-old (4y), the expression of DNMT3a and DNMT3b was decreased and became similar to that at 0.5y. Immunofluorescence of DNMT1, DNMT3a and DNMT3b on testis samples confirmed the differential expression and localization of these three DNA methylation transferases during horse development. Further molecular biological studies are needed to understand the implications of the expression patterns of these DNMTs in horse testes.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases , DNA Methyltransferase 3B , Gene Expression Regulation, Developmental , Testis , Animals , Male , Horses/genetics , Testis/metabolism , Testis/growth & development , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , DNA Methylation , Spermatogenesis/genetics , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism
6.
Cancer Immunol Immunother ; 73(7): 127, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38739169

ABSTRACT

Lactate dehydrogenase B (LDHB) reversibly catalyzes the conversion of pyruvate to lactate or lactate to pyruvate and expressed in various malignancies. However, the role of LDHB in modulating immune responses against hepatocellular carcinoma (HCC) remains largely unknown. Here, we found that down-regulation of lactate dehydrogenase B (LDHB) was coupled with the promoter hypermethylation and knocking down the DNA methyltransferase 3A (DNMT 3A) restored LDHB expression levels in HCC cell lines. Bioinformatics analysis of the HCC cohort from The Cancer Genome Atlas revealed a significant positive correlation between LDHB expression and immune regulatory signaling pathways and immune cell infiltrations. Moreover, immune checkpoint inhibitors (ICIs) have shown considerable promise for HCC treatment and patients with higher LDHB expression responded better to ICIs. Finally, we found that overexpression of LDHB suppressed HCC growth in immunocompetent but not in immunodeficient mice, suggesting that the host immune system was involved in the LDHB-medicated tumor suppression. Our findings indicate that DNMT3A-mediated epigenetic silencing of LDHB may contribute to HCC progression through remodeling the tumor immune microenvironment, and LDHB may become a potential prognostic biomarker and therapeutic target for HCC immunotherapy.


Subject(s)
Carcinoma, Hepatocellular , DNA Methyltransferase 3A , Epigenesis, Genetic , L-Lactate Dehydrogenase , Liver Neoplasms , Tumor Microenvironment , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Tumor Microenvironment/immunology , Humans , Animals , Mice , L-Lactate Dehydrogenase/metabolism , L-Lactate Dehydrogenase/genetics , DNA Methyltransferase 3A/metabolism , Gene Expression Regulation, Neoplastic , DNA Methylation , Isoenzymes/genetics , Isoenzymes/metabolism , Cell Line, Tumor , Gene Silencing , Prognosis
7.
Nat Commun ; 15(1): 3266, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38627502

ABSTRACT

DNA methyltransferase 3A (DNMT3A) and its catalytically inactive cofactor DNA methyltransferase 3-Like (DNMT3L) proteins form functional heterotetramers to deposit DNA methylation in mammalian germ cells. While both proteins have an ATRX-DNMT3-DNMT3L (ADD) domain that recognizes histone H3 tail unmethylated at lysine-4 (H3K4me0), the combined and differential roles of the domains in the two proteins have not been fully defined in vivo. Here we investigate DNA methylation landscapes in female and male germ cells derived from mice with loss-of-function amino acid substitutions in the ADD domains of DNMT3A and/or DNMT3L. Mutations in either the DNMT3A-ADD or the DNMT3L-ADD domain moderately decrease global CG methylation levels, but to different degrees, in both germ cells. Furthermore, when the ADD domains of both DNMT3A and DNMT3L lose their functions, the CG methylation levels are much more reduced, especially in oocytes, comparable to the impact of the Dnmt3a/3L knockout. In contrast, aberrant accumulation of non-CG methylation occurs at thousands of genomic regions in the double mutant oocytes and spermatozoa. These results highlight the critical role of the ADD-H3K4me0 binding in proper CG and non-CG methylation in germ cells and the various impacts of the ADD domains of the two proteins.


Subject(s)
DNA Methylation , DNA Methyltransferase 3A , Animals , Female , Male , Mice , DNA (Cytosine-5-)-Methyltransferases/metabolism , Germ Cells/metabolism , Histones/metabolism , Transcription Factors/metabolism
8.
Biomed Pharmacother ; 174: 116572, 2024 May.
Article in English | MEDLINE | ID: mdl-38626519

ABSTRACT

Epigenetic regulation and mitochondrial dysfunction are essential to the progression of idiopathic pulmonary fibrosis (IPF). Curcumin (CCM) in inhibits the progression of pulmonary fibrosis by regulating the expression of specific miRNAs and pulmonary fibroblast mitochondrial function; however, the underlying mechanism is unclear. C57BL/6 mice were intratracheally injected with bleomycin (5 mg/kg) and treated with CCM (25 mg/kg body weight/3 times per week, intraperitoneal injection) for 28 days. Verhoeff-Van Gieson, Picro sirius red, and Masson's trichrome staining were used to examine the expression and distribution of collagen and elastic fibers in the lung tissue. Pulmonary fibrosis was determined using micro-computed tomography and transmission electron microscopy. Human pulmonary fibroblasts were transfected with miR-29a-3p, and RT-qPCR, immunostaining, and western blotting were performed to determine the expression of DNMT3A and extracellular matrix collagen-1 (COL1A1) and fibronectin-1 (FN1) levels. The expression of mitochondrial electron transport chain complex (MRC) and mitochondrial function were detected using western blotting and Seahorse XFp Technology. CCM in increased the expression of miR-29a-3p in the lung tissue and inhibited the DNMT3A to reduce the COL1A1 and FN1 levels leading to pulmonary extracellular matrix remodeling. In addition, CCM inhibited pulmonary fibroblasts MRC and mitochondrial function via the miR-29a-3p/DNMT3A pathway. CCM attenuates pulmonary fibrosis via the miR-29a-3p/DNMT3A axis to regulate extracellular matrix remodeling and mitochondrial function and may provide a new therapeutic intervention for preventing pulmonary fibrosis.


Subject(s)
Curcumin , DNA Methyltransferase 3A , Extracellular Matrix , Fibroblasts , Mice, Inbred C57BL , MicroRNAs , Mitochondria , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Curcumin/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , DNA Methyltransferase 3A/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , Humans , Mice , Male , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Bleomycin , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/drug therapy , Lung/drug effects , Lung/pathology , Lung/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/metabolism , Disease Models, Animal
9.
Drug Metab Dispos ; 52(6): 555-564, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38565301

ABSTRACT

Cytochrome P450 1A2 (CYP1A2) is a known tumor suppressor in hepatocellular carcinoma (HCC), but its expression is repressed in HCC and the underlying mechanism is unclear. In this study, we investigated the epigenetic mechanisms of CYP1A2 repression and potential therapeutic implications. In HCC tumor tissues, the methylation rates of CYP1A2 CpG island (CGI) and DNA methyltransferase (DNMT) 3A protein levels were significantly higher, and there was a clear negative correlation between DNMT3A and CYP1A2 protein expression. Knockdown of DNMT3A by siRNA significantly increased CYP1A2 expression in HCC cells. Additionally, treating HCC cells with decitabine (DAC) resulted in a dose-dependent upregulation of CYP1A2 expression by reducing the methylation level of CYP1A2 CGI. Furthermore, we observed a decreased enrichment of H3K27Ac in the promoter region of CYP1A2 in HCC tissues. Treatment with the trichostatin A (TSA) restored CYP1A2 expression in HCC cells by increasing H3K27Ac levels in the CYP1A2 promoter region. Importantly, combination treatment of sorafenib with DAC or TSA resulted in a leftward shift of the dose-response curve, lower IC50 values, and reduced colony numbers in HCC cells. Our findings suggest that hypermethylation of the CGI at the promoter, mediated by the high expression of DNMT3A, and hypoacetylation of H3K27 in the CYP1A2 promoter region, leads to CYP1A2 repression in HCC. Epigenetic drugs DAC and TSA increase HCC cell sensitivity to sorafenib by restoring CYP1A2 expression. Our study provides new insights into the epigenetic regulation of CYP1A2 in HCC and highlights the potential of epigenetic drugs as a therapeutic approach for HCC. SIGNIFICANCE STATEMENT: This study marks the first exploration of the epigenetic mechanisms underlying cytochrome P450 (CYP) 1A2 suppression in hepatocellular carcinoma (HCC). Our findings reveal that heightened DNA methyltransferase expression induces hypermethylation of the CpG island at the promoter, coupled with diminished H3K27Ac levels, resulting in the repression of CYP1A2 in HCC. The use of epigenetic drugs such as decitabine and trichostatin A emerges as a novel therapeutic avenue, demonstrating their potential to restore CYP1A2 expression and enhance sorafenib sensitivity in HCC cells.


Subject(s)
Carcinoma, Hepatocellular , Cytochrome P-450 CYP1A2 , DNA Methylation , Epigenesis, Genetic , Liver Neoplasms , Sorafenib , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Humans , Sorafenib/pharmacology , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/genetics , DNA Methylation/drug effects , Cell Line, Tumor , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2/metabolism , DNA Methyltransferase 3A , Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Decitabine/pharmacology , CpG Islands/genetics , Hydroxamic Acids/pharmacology , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/drug effects
10.
Neurosci Lett ; 830: 137770, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38616004

ABSTRACT

Women are disproportionately affected by stress-related disorders like depression. In our prior research, we discovered that females exhibit lower basal hypothalamic reelin levels, and these levels are differentially influenced by chronic stress induced through repeated corticosterone (CORT) injections. Although epigenetic mechanisms involving DNA methylation and the formation of repressor complexes by DNA methyl-transferases (DNMTs) and Methyl-CpG binding protein 2 (MeCP2) have been recognized as regulators of reelin expression in vitro, there is limited understanding of the impact of stress on the epigenetic regulation of reelin in vivo and whether sex differences exist in these mechanisms. To address these questions, we conducted various biochemical analyses on hypothalamic brain samples obtained from male and female rats previously treated with either 21 days of CORT (40 mg/kg) or vehicle (0.9 % saline) subcutaneous injections. Upon chronic CORT treatment, a reduction in reelin fragment NR2 was noted in males, while the full-length molecule remained unaffected. This decrease paralleled with an elevation in MeCP2 and a reduction in DNMT3a protein levels only in males. Importantly, sex differences in baseline and CORT-induced reelin protein levels were not associated with changes in the methylation status of the Reln promoter. These findings suggest that CORT-induced reelin decreases in the hypothalamus may be a combination of alterations in downstream processes beyond gene transcription. This research brings novel insights into the sexually distinct consequences of chronic stress, an essential aspect to understand, particularly concerning its role in the development of depression.


Subject(s)
Cell Adhesion Molecules, Neuronal , Corticosterone , DNA Methyltransferase 3A , Extracellular Matrix Proteins , Hypothalamus , Methyl-CpG-Binding Protein 2 , Nerve Tissue Proteins , Reelin Protein , Serine Endopeptidases , Animals , Female , Male , Rats , Cell Adhesion Molecules, Neuronal/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A/metabolism , Extracellular Matrix Proteins/metabolism , Hypothalamus/metabolism , Hypothalamus/drug effects , Methyl-CpG-Binding Protein 2/metabolism , Methyl-CpG-Binding Protein 2/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Serine Endopeptidases/metabolism , Sex Characteristics , Rats, Long-Evans
11.
Nat Commun ; 15(1): 3111, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38600075

ABSTRACT

DNA methyltransferases DNMT3A- and DNMT3B-mediated DNA methylation critically regulate epigenomic and transcriptomic patterning during development. The hotspot DNMT3A mutations at the site of Arg822 (R882) promote polymerization, leading to aberrant DNA methylation that may contribute to the pathogenesis of acute myeloid leukemia (AML). However, the molecular basis underlying the mutation-induced functional misregulation of DNMT3A remains unclear. Here, we report the crystal structures of the DNMT3A methyltransferase domain, revealing a molecular basis for its oligomerization behavior distinct to DNMT3B, and the enhanced intermolecular contacts caused by the R882H or R882C mutation. Our biochemical, cellular, and genomic DNA methylation analyses demonstrate that introducing the DNMT3B-converting mutations inhibits the R882H-/R882C-triggered DNMT3A polymerization and enhances substrate access, thereby eliminating the dominant-negative effect of the DNMT3A R882 mutations in cells. Together, this study provides mechanistic insights into DNMT3A R882 mutations-triggered aberrant oligomerization and DNA hypomethylation in AML, with important implications in cancer therapy.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases , Leukemia, Myeloid, Acute , Humans , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Mutation , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , DNA Methylation/genetics , DNA/metabolism
12.
Diabetologia ; 67(6): 1079-1094, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38512414

ABSTRACT

AIMS/HYPOTHESIS: Beta cells within the pancreatic islet represent a heterogenous population wherein individual sub-groups of cells make distinct contributions to the overall control of insulin secretion. These include a subpopulation of highly connected 'hub' cells, important for the propagation of intercellular Ca2+ waves. Functional subpopulations have also been demonstrated in human beta cells, with an altered subtype distribution apparent in type 2 diabetes. At present, the molecular mechanisms through which beta cell hierarchy is established are poorly understood. Changes at the level of the epigenome provide one such possibility, which we explore here by focusing on the imprinted gene Nnat (encoding neuronatin [NNAT]), which is required for normal insulin synthesis and secretion. METHODS: Single-cell RNA-seq datasets were examined using Seurat 4.0 and ClusterProfiler running under R. Transgenic mice expressing enhanced GFP under the control of the Nnat enhancer/promoter regions were generated for FACS of beta cells and downstream analysis of CpG methylation by bisulphite sequencing and RNA-seq, respectively. Animals deleted for the de novo methyltransferase DNA methyltransferase 3 alpha (DNMT3A) from the pancreatic progenitor stage were used to explore control of promoter methylation. Proteomics was performed using affinity purification mass spectrometry and Ca2+ dynamics explored by rapid confocal imaging of Cal-520 AM and Cal-590 AM. Insulin secretion was measured using homogeneous time-resolved fluorescence imaging. RESULTS: Nnat mRNA was differentially expressed in a discrete beta cell population in a developmental stage- and DNA methylation (DNMT3A)-dependent manner. Thus, pseudo-time analysis of embryonic datasets demonstrated the early establishment of Nnat-positive and -negative subpopulations during embryogenesis. NNAT expression is also restricted to a subset of beta cells across the human islet that is maintained throughout adult life. NNAT+ beta cells also displayed a discrete transcriptome at adult stages, representing a subpopulation specialised for insulin production, and were diminished in db/db mice. 'Hub' cells were less abundant in the NNAT+ population, consistent with epigenetic control of this functional specialisation. CONCLUSIONS/INTERPRETATION: These findings demonstrate that differential DNA methylation at Nnat represents a novel means through which beta cell heterogeneity is established during development. We therefore hypothesise that changes in methylation at this locus may contribute to a loss of beta cell hierarchy and connectivity, potentially contributing to defective insulin secretion in some forms of diabetes. DATA AVAILABILITY: The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the dataset identifier PXD048465.


Subject(s)
CpG Islands , DNA Methylation , Insulin-Secreting Cells , Insulin-Secreting Cells/metabolism , Animals , Mice , CpG Islands/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice, Transgenic , DNA Methyltransferase 3A/metabolism , Humans , Insulin/metabolism , Insulin Secretion/physiology
13.
Bone ; 183: 117085, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38522809

ABSTRACT

Overgrowth and intellectual disability disorders in humans are typified by length/height and/or head circumference ≥ 2 standard deviations above the mean as well as intellectual disability and behavioral comorbidities, including autism and anxiety. Tatton-Brown-Rahman Syndrome is one type of overgrowth and intellectual disability disorder caused by heterozygous missense mutations in the DNA methyltransferase 3A (DNMT3A) gene. Numerous DNMT3A mutations have been identified in Tatton-Brown-Rahman Syndrome patients and may be associated with varying phenotype severities of clinical presentation. Two such mutations are the R882H and P904L mutations which result in severe and mild phenotypes, respectively. Mice with paralogous mutations (Dnmt3aP900L/+ and Dnmt3aR878H/+) exhibit overgrowth in their long bones (e.g., femur, humerus), but the mechanisms responsible for their skeletal overgrowth remain unknown. The goal of this study is to characterize skeletal phenotypes in mouse models of Tatton-Brown-Rahman Syndrome and identify potential cellular mechanisms involved in the skeletal overgrowth phenotype. We report that mature mice with the Dnmt3aP900L/+ or Dnmt3aR878H/+ mutation exhibit tibial overgrowth, cortical bone thinning, and weakened bone mechanical properties. Dnmt3aR878H/+ mutants also contain larger bone marrow adipocytes while Dnmt3aP900L/+ mutants show no adipocyte phenotype compared to control animals. To understand the potential cellular mechanisms regulating these phenotypes, growth plate chondrocytes, osteoblasts, and osteoclasts were assessed in juvenile mutant mice using quantitative static histomorphometry and dynamic histomorphometry. Tibial growth plates appeared thicker in mutant juvenile mice, but no changes were observed in osteoblast activity or osteoclast number in the femoral mid-diaphysis. These studies reveal new skeletal phenotypes associated with Tatton-Brown-Rahman Syndrome in mice and provide a rationale to extend clinical assessments of patients with this condition to include bone density and quality testing. These findings may be also informative for skeletal characterization of other mouse models presenting with overgrowth and intellectual disability phenotypes.


Subject(s)
Abnormalities, Multiple , Intellectual Disability , Musculoskeletal Abnormalities , Humans , Animals , Mice , DNA (Cytosine-5-)-Methyltransferases/genetics , Intellectual Disability/genetics , Mutation, Missense , DNA Methyltransferase 3A , Abnormalities, Multiple/genetics , Mutation
14.
Blood Adv ; 8(9): 2138-2147, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38429084

ABSTRACT

ABSTRACT: Epstein-Barr virus (EBV)-positive (EBV+) nodal T- and natural killer (NK)-cell lymphoma is a peripheral T-cell lymphoma (EBV+ nPTCL) that presents as a primary nodal disease with T-cell phenotype and EBV-harboring tumor cells. To date, the genetic aspect of EBV+ nPTCL has not been fully investigated. In this study, whole-exome and/or whole-genome sequencing was performed on 22 cases of EBV+ nPTCL. TET2 (68%) and DNMT3A (32%) were observed to be the most frequently mutated genes whose presence was associated with poor overall survival (P = .004). The RHOA p.Gly17Val mutation was identified in 2 patients who had TET2 and/or DNMT3A mutations. In 4 patients with TET2/DNMT3A alterations, blood cell-rich tissues (the bone marrow [BM] or spleen) were available as paired normal samples. Of 4 cases, 3 had at least 1 identical TET2/DNMT3A mutation in the BM or spleen. Additionally, the whole part of the EBV genome was sequenced and structural variations (SVs) were found frequent among the EBV genomes (63%). The most frequently identified type of SV was deletion. In 1 patient, 4 pieces of human chromosome 9, including programmed death-ligand 1 gene (PD-L1) were identified to be tandemly incorporated into the EBV genome. The 3' untranslated region of PD-L1 was truncated, causing a high-level of PD-L1 protein expression. Overall, the frequent TET2 and DNMT3A mutations in EBV+ nPTCL seem to be closely associated with clonal hematopoiesis and, together with the EBV genome deletions, may contribute to the pathogenesis of this intractable lymphoma.


Subject(s)
Epstein-Barr Virus Infections , Genome, Viral , Mutation , Humans , Male , Female , Middle Aged , Aged , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/genetics , Adult , Herpesvirus 4, Human/genetics , DNA Methyltransferase 3A , Lymphoma, Extranodal NK-T-Cell/genetics , Lymphoma, Extranodal NK-T-Cell/virology , Genomic Structural Variation , Lymphoma, T-Cell, Peripheral/genetics , Lymphoma, T-Cell, Peripheral/virology , Dioxygenases
15.
Oncogene ; 43(18): 1386-1396, 2024 May.
Article in English | MEDLINE | ID: mdl-38467852

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) presents a unique profile characterized by high levels of angiogenesis and robust vascularization. Understanding the underlying mechanisms driving this heterogeneity is essential for developing effective therapeutic strategies. This study revealed that ubiquitin B (UBB) is downregulated in ccRCC, which adversely affects the survival of ccRCC patients. UBB exerts regulatory control over vascular endothelial growth factor A (VEGFA) by directly interacting with specificity protein 1 (SP1), consequently exerting significant influence on angiogenic processes. Subsequently, we validated that DNA methyltransferase 3 alpha (DNMT3A) is located in the promoter of UBB to epigenetically inhibit UBB transcription. Additionally, we found that an unharmonious UBB/VEGFA ratio mediates pazopanib resistance in ccRCC. These findings underscore the critical involvement of UBB in antiangiogenic therapy and unveil a novel therapeutic strategy for ccRCC.


Subject(s)
Carcinoma, Renal Cell , Down-Regulation , Gene Expression Regulation, Neoplastic , Kidney Neoplasms , Neovascularization, Pathologic , Sp1 Transcription Factor , Vascular Endothelial Growth Factor A , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/drug therapy , Humans , Kidney Neoplasms/pathology , Kidney Neoplasms/genetics , Kidney Neoplasms/blood supply , Kidney Neoplasms/metabolism , Kidney Neoplasms/drug therapy , Sp1 Transcription Factor/metabolism , Sp1 Transcription Factor/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Cell Line, Tumor , Animals , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Indazoles/pharmacology , Indazoles/therapeutic use , DNA Methyltransferase 3A/metabolism , Sulfonamides/pharmacology , Mice , Ubiquitin/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , Drug Resistance, Neoplasm/genetics , Promoter Regions, Genetic , Female , Male , Angiogenesis
16.
Br J Haematol ; 204(5): 1844-1855, 2024 May.
Article in English | MEDLINE | ID: mdl-38522849

ABSTRACT

Recursive partitioning of healthy consortia led to the development of the Clonal Hematopoiesis Risk Score (CHRS) for clonal haematopoiesis (CH); however, in the practical setting, most cases of CH are diagnosed after patients present with cytopenias or related symptoms. To address this real-world population, we characterize the clinical trajectories of 94 patients with CH and distinguish CH harbouring canonical DNMT3A/TET2/ASXL1 mutations alone ('sole DTA') versus all other groups ('non-sole DTA'). TET2, rather than DNMT3A, was the most prevalent mutation in the real-world setting. Sole DTA patients did not progress to myeloid neoplasm (MN) in the absence of acquisition of other mutations. Contrastingly, 14 (20.1%) of 67 non-sole DTA patients progressed to MN. CHRS assessment showed a higher frequency of high-risk CH in non-sole DTA (vs. sole DTA) patients and in progressors (vs. non-progressors). RUNX1 mutation conferred the strongest risk for progression to MN (odds ratio [OR] 10.27, 95% CI 2.00-52.69, p = 0.0053). The mean variant allele frequency across all genes was higher in progressors than in non-progressors (36.9% ± 4.62% vs. 24.1% ± 1.67%, p = 0.0064). This analysis in the post-CHRS era underscores the natural history of CH, providing insight into patterns of progression to MN.


Subject(s)
Clonal Hematopoiesis , DNA-Binding Proteins , Dioxygenases , Mutation , Humans , Clonal Hematopoiesis/genetics , Male , Female , Middle Aged , Aged , DNA-Binding Proteins/genetics , DNA Methyltransferase 3A , Adult , Aged, 80 and over , Disease Progression , Core Binding Factor Alpha 2 Subunit/genetics , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics
17.
Cell Rep Med ; 5(4): 101484, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38554704

ABSTRACT

The use of Bruton tyrosine kinase (BTK) inhibitors such as ibrutinib achieves a remarkable clinical response in mantle cell lymphoma (MCL). Acquired drug resistance, however, is significant and affects long-term survival of MCL patients. Here, we demonstrate that DNA methyltransferase 3A (DNMT3A) is involved in ibrutinib resistance. We find that DNMT3A expression is upregulated upon ibrutinib treatment in ibrutinib-resistant MCL cells. Genetic and pharmacological analyses reveal that DNMT3A mediates ibrutinib resistance independent of its DNA-methylation function. Mechanistically, DNMT3A induces the expression of MYC target genes through interaction with the transcription factors MEF2B and MYC, thus mediating metabolic reprogramming to oxidative phosphorylation (OXPHOS). Targeting DNMT3A with low-dose decitabine inhibits the growth of ibrutinib-resistant lymphoma cells both in vitro and in a patient-derived xenograft mouse model. These findings suggest that targeting DNMT3A-mediated metabolic reprogramming to OXPHOS with decitabine provides a potential therapeutic strategy to overcome ibrutinib resistance in relapsed/refractory MCL.


Subject(s)
Adenine/analogs & derivatives , Lymphoma, Mantle-Cell , Piperidines , Protein-Tyrosine Kinases , Humans , Animals , Mice , Adult , Agammaglobulinaemia Tyrosine Kinase/metabolism , Drug Resistance, Neoplasm/genetics , DNA Methyltransferase 3A , Oxidative Phosphorylation , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/pathology , Decitabine/metabolism , Decitabine/therapeutic use
18.
J Cell Biol ; 223(4)2024 04 01.
Article in English | MEDLINE | ID: mdl-38376465

ABSTRACT

DNA methylation (DNAme) is a key epigenetic mark that regulates critical biological processes maintaining overall genome stability. Given its pleiotropic function, studies of DNAme dynamics are crucial, but currently available tools to interfere with DNAme have limitations and major cytotoxic side effects. Here, we present cell models that allow inducible and reversible DNAme modulation through DNMT1 depletion. By dynamically assessing whole genome and locus-specific effects of induced passive demethylation through cell divisions, we reveal a cooperative activity between DNMT1 and DNMT3B, but not of DNMT3A, to maintain and control DNAme. We show that gradual loss of DNAme is accompanied by progressive and reversible changes in heterochromatin, compartmentalization, and peripheral localization. DNA methylation loss coincides with a gradual reduction of cell fitness due to G1 arrest, with minor levels of mitotic failure. Altogether, this system allows DNMTs and DNA methylation studies with fine temporal resolution, which may help to reveal the etiologic link between DNAme dysfunction and human disease.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methylation , DNA Methyltransferase 3A , Epigenomics , Humans , Cell Division , Heterochromatin/genetics , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA Methyltransferase 3A/genetics , Cell Line
19.
Adv Sci (Weinh) ; 11(16): e2308531, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38380551

ABSTRACT

Gallbladder cancer (GBC) is an extremely lethal malignancy with aggressive behaviors, including liver or distant metastasis; however, the underlying mechanisms driving the metastasis of GBC remain poorly understood. In this study, it is found that DNA methyltransferase DNMT3A is highly expressed in GBC tumor tissues compared to matched adjacent normal tissues. Clinicopathological analysis shows that DNMT3A is positively correlated with liver metastasis and poor overall survival outcomes in patients with GBC. Functional analysis confirms that DNMT3A promotes the metastasis of GBC cells in a manner dependent on its DNA methyltransferase activity. Mechanistically, DNMT3A interacts with and is recruited by YAP/TAZ to recognize and access the CpG island within the CDH1 promoter and generates hypermethylation of the CDH1 promoter, which leads to transcriptional silencing of CDH1 and accelerated epithelial-to-mesenchymal transition. Using tissue microarrays, the association between the expression of DNMT3A, YAP/TAZ, and CDH1 is confirmed, which affects the metastatic ability of GBC. These results reveal a novel mechanism through which DNMT3A recruitment by YAP/TAZ guides DNA methylation to drive GBC metastasis and provide insights into the treatment of GBC metastasis by targeting the functional connection between DNMT3A and YAP/TAZ.


Subject(s)
DNA Methyltransferase 3A , Gallbladder Neoplasms , Animals , Female , Humans , Male , Mice , Middle Aged , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Antigens, CD , Cadherins , Cell Line, Tumor , Disease Models, Animal , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation/genetics , DNA Methyltransferase 3A/metabolism , DNA Methyltransferase 3A/genetics , Epithelial-Mesenchymal Transition/genetics , Gallbladder Neoplasms/genetics , Gallbladder Neoplasms/metabolism , Gallbladder Neoplasms/pathology , Gene Expression Regulation, Neoplastic/genetics , Neoplasm Metastasis/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , YAP-Signaling Proteins/metabolism , YAP-Signaling Proteins/genetics
20.
Mol Biol Rep ; 51(1): 78, 2024 Jan 06.
Article in English | MEDLINE | ID: mdl-38183507

ABSTRACT

BACKGROUND: Aberrant DNA methylation has been implicated in the development of gastric cancer (GC). In our previous study, we demonstrated that fructose-1,6-bisphosphatase-2 (FBP2), an enzyme that suppresses cell glycolysis and growth, is downregulated in GC due to promoter methylation. However, the precise mechanism underlying this process remains unknown. Thus, this study aimed to elucidate the mechanisms involved in FBP2 promoter hypermethylation. METHODS AND RESULTS: The methylation levels in GC and normal adjacent tissues were quantified using methylation-specific polymerase chain reaction. FBP2 promoter was frequently hypermethylated in primary GC tissues compared to adjacent normal tissues. To explore the functional consequences of this hypermethylation, we employed small interfering RNA-mediated knockdown of DNA methyltransferase 3a (DNMT3a) in GC cells. FBP2 expression increased following DNMT3a knockdown, suggesting that reduced methylation of the FBP2 promoter contributed to this upregulation. To further investigate this interaction, chromatin immunoprecipitation assays were conducted. The results confirmed an interaction between DNMT3a and the FBP2 promoter region, providing evidence that DNMT3a-mediated hypermethylation of the FBP2 promoter promotes GC progression. CONCLUSIONS: This study provides evidence that DNMT3a is involved in the hypermethylation of the FBP2 promoter and regulation of GC cell metabolism. Hypermethylation of the FBP2 promoter may be a promising prognostic biomarker in GC.


Subject(s)
DNA Methylation , Stomach Neoplasms , Humans , Carcinogenesis , DNA Methylation/genetics , DNA Methyltransferase 3A , DNA Modification Methylases , Fructose , Fructose-Bisphosphatase/genetics , Promoter Regions, Genetic/genetics , Stomach Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...