Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Life Sci ; 292: 119552, 2022 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-33932446

RESUMO

AIMS: Heart failure (HF) is a progressive disease with recurrent hospitalizations and high mortality. However, the mechanisms underlying HF remain unclear. The present study aimed to explore the regulatory mechanism of histone deacetylase 3 (HDAC3) and DNA methyltransferase 1 (DNMT1)/Src homology domain 2-containing tyrosine phosphatase-1 (SHP-1) axis in HF. METHODS: The HF rat models and hypertrophy cell models were established. The characteristic parameters of the heart were detected by echocardiography. A multichannel physiological signal acquisition system was used to detect the hemodynamic parameters. Real-time quantitative polymerase chain reaction (RT-qPCR) was used to detect the expression of HDAC3, DNMT1, and SHP-1 mRNAs, while Western blot was applied to analyze the expression of proteins. Masson staining was used to analyze the degree of collagen fiber infiltration. TdT-mediated DUTP nick end labeling (TUNEL) staining was performed to analyze the apoptosis of myocardial tissue cells. Co-immunoprecipitation (co-IP) was conducted to study the interaction between HDAC3 and DNMT1. Flow cytometry was used to analyze the apoptosis. KEY FINDINGS: HDAC3 and DNMT1 were highly expressed in HF rat and hypertrophy cell models. HDAC3 modified DNMT1 through deacetylation to inhibit ubiquitination-mediated degradation, which promoted the expression of DNMT1. DNMT1 inhibited SHP-1 expression via methylation in the promoter region. In summary, HDAC3 modified DNMT1 by deacetylation to suppress SHP-1 expression, which in turn led to the development of cardiomyocyte hypertrophy-induced HF. SIGNIFICANCE: This study provided potential therapeutic targets for HF treatment.


Assuntos
DNA (Citosina-5-)-Metiltransferase 1/fisiologia , Insuficiência Cardíaca/metabolismo , Histona Desacetilases/fisiologia , Proteína Tirosina Fosfatase não Receptora Tipo 6/fisiologia , Animais , Animais Recém-Nascidos , Metilação de DNA , Masculino , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley
2.
Clin. transl. oncol. (Print) ; 23(7): 1440-1451, jul. 2021. ilus
Artigo em Inglês | IBECS | ID: ibc-221984

RESUMO

Purpose Altered miRNAs play a crucial role in the emergence of the breast cancer stem cell (BCSC) phenotype. The interplay between miRNAs and methylation enzymes has been documented. One of the most aggressive breast cancer cell lines, MDA-MB-231, has expressed much more DNMT3B than DNMT3A. This study aims to evaluate the ability of miR-203 restoration and miR-150 inhibition to regulate DNMT3B and DNMT3A to modify the methylation level of BCSC-associated genes. Methods MDA-MB-231 cells were transfected with miR-203 mimic or miR-150 inhibitor or DNMT3B siRNA, and downstream analysis was performed by flow cytometry, real-time PCR and Western blotting. Results DNMT3A and DNMT3B are regulated both by miR-203a-3p and miR-150-5p. Transfection with miR-203 mimic and miR-150 inhibitor significantly reduced the CD44+CD24− subpopulation and down-regulated the expression of CD44 mRNA by increasing promoter methylation levels. SiRNA knockdown of DNMT3B increased the CD44+CD24− subpopulation and the expression of CD44 and ALDH1A3 by decreasing methylation density. The inhibition of miR-150 down-regulated OCT3/4 and SOX2 expression without affecting methylation levels, while miR-203 restoration and miR-150 inhibition down-regulated NANOG expression by elevating the methylation level. A positive-feedback loop was found between miR-203 and its target DNMT3B, as restoring miR-203 suppressed DNMT3B, while knocking down DNMT3B up-regulated miR-203. The restoration of miR-203 and knockdown of DNMT3B decreased methylation levels and increased the expression of miR-141 and miR-200c. Conclusions The study concluded that miR-203 and miR-150 play a role in the regulation of genes involved in BCSC methylation, including other miRNAs, by targeting DNMT3B and DNMT3A (AU)


Assuntos
Humanos , Feminino , Neoplasias da Mama/genética , DNA (Citosina-5-)-Metiltransferase 1/fisiologia , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Metilação , Células Tumorais Cultivadas
3.
Am J Physiol Endocrinol Metab ; 320(3): E598-E608, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33284093

RESUMO

Diabetic retinopathy (DR) is one of the serious complications that occurs in diabetic patients that frequently causes blindness. Long noncoding RNAs (lncRNAs) have been associated with DR pathology. This study aimed to determine the underlying mechanism of lncRNA maternally expressed gene 3 (MEG3) in association with DNA methyltransferase 1 (DNMT1) in the endothelial-mesenchymal transition (endMT) that occurs in DR. A rat model of DR was induced by streptozotocin (STZ) injection, and a high-glucose (HG)-induced cell model was established by exposing microvascular endothelial cells obtained from retina of rats to HG. Subsequently, MEG3 was overexpressed in rat and cell models to characterize its impact on endMT in DR and the involvement of the phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling pathway. Furthermore, the methylation level of MEG3 promoter region was determined with the application of methylation-specific polymerase chain reaction, followed by chromatin immunoprecipitation assay for methyltransferase enrichment. Finally, we examined the regulation of DNMT1 on MEG3 methylation and endMT in the HG-induced cell model. The results obtained revealed downregulated MEG3 expression in DR rat and cell models. Overexpressed MEG3 was shown to suppress endMT in DR rat and cell models through the inhibition of the PI3K/Akt/mTOR signaling pathway. Notably, DNMT1 could promote MEG3 promoter methylation to inhibit MEG3 expression by recruiting methyltransferase, which activated the PI3K/Akt/mTOR signaling pathway to accelerate endMT in DR. These findings further highlighted the inhibitory effect of MEG3 on endMT in DR, thus presenting a novel therapeutic target candidate for DR treatment.


Assuntos
DNA (Citosina-5-)-Metiltransferase 1/fisiologia , Retinopatia Diabética/genética , Retinopatia Diabética/patologia , Células Endoteliais/fisiologia , RNA Longo não Codificante/genética , Animais , Glicemia/metabolismo , Transdiferenciação Celular/genética , Células Cultivadas , Metilação de DNA/genética , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Retinopatia Diabética/metabolismo , Retinopatia Diabética/fisiopatologia , Modelos Animais de Doenças , Masculino , Células-Tronco Mesenquimais/fisiologia , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Longo não Codificante/metabolismo , Ratos , Ratos Sprague-Dawley , Retina/metabolismo , Retina/patologia , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/metabolismo
4.
Sci Rep ; 10(1): 11293, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32647199

RESUMO

The ciliary marginal zone (CMZ) of the zebrafish retina contains a population of actively proliferating resident stem cells, which generate retinal neurons throughout life. The maintenance methyltransferase, dnmt1, is expressed within the CMZ. Loss of dnmt1 function results in gene misregulation and cell death in a variety of developmental contexts, however, its role in retinal stem cell (RSC) maintenance is currently unknown. Here, we demonstrate that zebrafish dnmt1s872 mutants possess severe defects in RSC maintenance within the CMZ. Using a combination of immunohistochemistry, in situ hybridization, and a transgenic reporter assay, our results demonstrate a requirement for dnmt1 activity in the regulation of RSC proliferation, gene expression and in the repression of endogenous retroelements (REs). Ultimately, cell death is elevated in the dnmt1-/- CMZ, but in a p53-independent manner. Using a transgenic reporter for RE transposition activity, we demonstrate increased transposition in the dnmt1-/- CMZ. Taken together our data identify a critical role for dnmt1 function in RSC maintenance in the vertebrate eye.


Assuntos
DNA (Citosina-5-)-Metiltransferase 1/fisiologia , Neurônios Retinianos/fisiologia , Células-Tronco/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Neurônios Retinianos/citologia , Células-Tronco/citologia
5.
Aging (Albany NY) ; 12(8): 6558-6569, 2020 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-32310825

RESUMO

Inflammation plays an essential role in the development of lumbar disc degeneration (LDD), although the exact effects of macrophage subtypes on LDD remain unclear. Based on previous studies, we hypothesized that M2-polarization of local macrophages and simultaneous suppression of their production of fibrotic transforming growth factor beta 1 (TGFß1) could inhibit progression of LDD. Thus, we applied an orthotopic injection of adeno-associated virus (AAV) carrying shRNA for DNA Methyltransferase 1 (DNMT1) and/or shRNA for TGFß1 under a macrophage-specific CD68 promoter to specifically target local macrophages in a mouse model for LDD. We found that shDNMT1 significantly reduced levels of the pro-inflammatory cytokines TNFα, IL-1ß and IL-6, significantly increased levels of the anti-inflammatory cytokines IL-4 and IL-10, significantly increased M2 macrophage polarization, significantly reduced cell apoptosis in the disc degeneration zone and significantly reduced LDD-associated pain. The anti-apoptotic and anti-pain effects were further strengthened by co-application of shTGFß1. Together, these data suggest that M2 polarization of macrophages induced by both epigenetic modulation and suppressed production and release of TGFß1 from polarized M2 macrophages, may have a demonstrable therapeutic effect on LDD.


Assuntos
Epigênese Genética , Degeneração do Disco Intervertebral/prevenção & controle , Vértebras Lombares , Macrófagos/fisiologia , Animais , Antígenos CD/genética , Antígenos de Diferenciação Mielomonocítica/genética , Apoptose , Polaridade Celular , Colágeno Tipo II/análise , Citocinas/biossíntese , DNA (Citosina-5-)-Metiltransferase 1/genética , DNA (Citosina-5-)-Metiltransferase 1/fisiologia , Dependovirus/genética , Terapia Genética , Degeneração do Disco Intervertebral/imunologia , Degeneração do Disco Intervertebral/patologia , Camundongos , Camundongos Endogâmicos BALB C , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/fisiologia
6.
Dev Biol ; 455(2): 473-484, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31394080

RESUMO

Intestinal tract development is a coordinated process involving signaling among the progenitors and developing cells from all three germ layers. Development of endoderm-derived intestinal epithelium has been shown to depend on epigenetic modifications, but whether that is also the case for intestinal tract cell types from other germ layers remains unclear. We found that functional loss of a DNA methylation machinery component, ubiquitin-like protein containing PHD and RING finger domains 1 (uhrf1), leads to reduced numbers of ectoderm-derived enteric neurons and severe disruption of mesoderm-derived intestinal smooth muscle. Genetic chimeras revealed that Uhrf1 functions both cell-autonomously in enteric neuron precursors and cell-non-autonomously in surrounding intestinal cells, consistent with what is known about signaling interactions between these cell types that promote one another's development. Uhrf1 recruits the DNA methyltransferase Dnmt1 to unmethylated DNA during replication. Dnmt1 is also expressed in enteric neurons and smooth muscle progenitors. dnmt1 mutants have fewer enteric neurons and disrupted intestinal smooth muscle compared to wildtypes. Because dnmt1;uhrf1 double mutants have a similar phenotype to dnmt1 and uhrf1 single mutants, Dnmt1 and Uhrf1 must function together during enteric neuron and intestinal muscle development. This work shows that genes controlling epigenetic modifications are important to coordinate intestinal tract development, provides the first demonstration that these genes influence development of the ENS, and advances uhrf1 and dnmt1 as potential new Hirschsprung disease candidates.


Assuntos
DNA (Citosina-5-)-Metiltransferase 1/fisiologia , Sistema Nervoso Entérico/embriologia , Epigênese Genética , Intestinos/embriologia , Transativadores/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Animais , Quimera , DNA (Citosina-5-)-Metiltransferase 1/genética , Células-Tronco Embrionárias/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Intestinos/citologia , Intestinos/inervação , Masculino , Músculo Liso/embriologia , Mutação , Neurônios , Transativadores/genética , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
7.
Dev Biol ; 454(2): 108-117, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31242446

RESUMO

Lung endoderm development occurs through a series of finely coordinated transcriptional processes that are regulated by epigenetic mechanisms. However, the role of DNA methylation in regulating lung endoderm development remains poorly understood. We demonstrate that DNA methyltransferase 1 (Dnmt1) is required for early branching morphogenesis of the lungs and for restraining epithelial fate specification. Loss of Dnmt1 leads to an early branching defect, a loss of epithelial polarity and proximal endodermal cell differentiation, and an expansion of the distal endoderm compartment. Dnmt1 deficiency also disrupts epithelial-mesenchymal crosstalk and leads to precocious distal endodermal cell differentiation with premature expression of alveolar type 2 cell restricted genes. These data reveal an important requirement for Dnmt1 mediated DNA methylation in early lung development to promote proper branching morphogenesis, maintain proximal endodermal cell fate, and suppress premature activation of the distal epithelial fate.


Assuntos
Células Epiteliais Alveolares/metabolismo , DNA (Citosina-5-)-Metiltransferase 1/genética , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Animais , Diferenciação Celular/genética , Linhagem da Célula/fisiologia , Polaridade Celular , Proliferação de Células/genética , DNA (Citosina-5-)-Metiltransferase 1/fisiologia , Metilação de DNA/genética , Endoderma/metabolismo , Epigênese Genética/genética , Células Epiteliais/citologia , Transição Epitelial-Mesenquimal , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Pulmão/citologia , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos , Morfogênese , Organogênese/fisiologia , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo
8.
Exp Cell Res ; 382(1): 111461, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31194977

RESUMO

Emerging evidence indicates that long noncoding RNAs (LncRNAs) are new players in gene regulation but their mechanisms of action are mainly undocumented. In this study, we investigated LncRNA alterations that contribute to lung cancer by analyzing published microarray data in Gene Expression Obminus (GEO) and The Cancer Genome Atlas RNA (TCGA) sequencing data. Here, we reported that HAGLR (also called HOXD-AS1) was frequently down-regulated in lung adenocarcinoma (LUAD) tissues, and decreased HAGLR expression was clinically associated with shorter survival of LUAD patients. Preclinical studies using multiple LUAD cells and in vivo mouse model indicated that HAGLR could attenuate LUAD cell growth in vitro and in vivo. Mechanistically, HAGLR could physically interact with DNMT1, and recruit DNMT1 on E2F1 promoter to increase local DNA methylation. Overall, our study demonstrated that HAGLR promoted LUAD progression by recruiting DNMT1 to modulate the promoter methylation and expression of E2F1, which expanded potential therapeutic strategies for LUAD treatment.


Assuntos
Adenocarcinoma/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Fator de Transcrição E2F1/genética , Genes Supressores de Tumor , Proteínas de Neoplasias/genética , RNA Longo não Codificante/fisiologia , RNA Neoplásico/fisiologia , Adenocarcinoma/mortalidade , Adenocarcinoma/patologia , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Carcinoma Pulmonar de Células não Pequenas/patologia , DNA (Citosina-5-)-Metiltransferase 1/fisiologia , Metilação de DNA , DNA de Neoplasias/genética , Fator de Transcrição E2F1/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , Estimativa de Kaplan-Meier , Proteínas de Neoplasias/antagonistas & inibidores , Prognóstico , Regiões Promotoras Genéticas/genética , Mapeamento de Interação de Proteínas , Interferência de RNA , RNA Longo não Codificante/genética , RNA Neoplásico/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia
9.
Birth Defects Res ; 110(20): 1551-1555, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30576090

RESUMO

BACKGROUND: Cardiac fibrosis is a cardinal feature of multiple types of cardiovascular disease, which lead to heart failure. Multiple studies connect adverse maternal environment (AME) with cardiac fibrosis. AME does not always result in fibrosis, though. An additional "insult", such as an adult Western diet (WD), is frequently necessary. The additive effects of AME and adult WD on cardiac fibrosis is not well-understood. AME can also alter DNA methylation. DNA methyltransferase (DNMT) and ten-eleven translocation (TET) are methylation modifying genes that regulate DNA methylation, but it is unknown if AME changes cardiac gene expression of DNMT and TET. We sought to use a model of AME and adult WD to investigate the development of cardiac fibrosis and cardiac mRNA expression of DNMT and TET genes. METHODS: We exposed dams to WD or control diet (CD) 5 weeks before pregnancy and through lactation. We added environmental stressors during the last third of pregnancy to dams on WD to create AME. Dams on CD experienced no added stressors to create control maternal environment (CME). Male offspring were weaned at Postnatal Week 3 (W3) and placed on WD or CD to create four groups: CME-CD, CME-WD, AME-CD, and AME-WD. RESULTS: AME-WD increased cardiac fibrosis in adulthood (p < .05), whereas AME-CD and CME-WD did not. TET1-3 and DNMT3a mRNA levels decreased in AME versus CME offspring (p < .01). CONCLUSION: AME increases susceptibility to cardiac fibrosis in adult male mice. Early-life changes to TET expression may mediate susceptibility to fibrosis, but further testing is needed.


Assuntos
Fibrose/etiologia , Exposição Materna/efeitos adversos , Miocárdio/patologia , Animais , Doenças Cardiovasculares/etiologia , DNA (Citosina-5-)-Metiltransferase 1/genética , DNA (Citosina-5-)-Metiltransferase 1/fisiologia , Metilação de DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Dieta Ocidental/efeitos adversos , Feminino , Fibrose/genética , Coração/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/fisiologia
10.
Shanghai Kou Qiang Yi Xue ; 27(3): 275-279, 2018 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-30411123

RESUMO

PURPOSE: To investigate the effects of DNA methyltransferase in mucoepidermoid carcinoma of human salivary glands tissues. METHODS: Forty-three samples from mucoepidermoid carcinoma of salivary glands and 17 normal salivary gland tissues were collected from January 2010 to September 2013. Immunohistochemistry and Western blot were used to detect the expression of Dnmt1 and Dnmt3b in normal tissues and specimen of mucoepidermoid carcinoma of salivary glands. The data were analysed with SPSS 22.0 software package. RESULTS: The positive expression rate of Dnmt1 in mucoepidermoid carcinoma tissue was 37.21%, and that in normal salivary gland tissues was 17.65%, there was no significant difference between them; the positive expression rate of Dnmt3b in mucoepidermoid carcinoma tissue was 83.72%, which was significantly higher than that in normal salivary gland tissue (11.76%, P<0.01). However, there was no significant correlation between high expression of Dnmt1 and Dnmt3b and clinicopathological parameters. CONCLUSIONS: Dnmt3b may play a role in the tumorigenesis of mucoepidermoid carcinoma in salivary glands.


Assuntos
Carcinoma Mucoepidermoide , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases , Neoplasias das Glândulas Salivares , Carcinoma Mucoepidermoide/metabolismo , Transformação Celular Neoplásica , DNA (Citosina-5-)-Metiltransferase 1/fisiologia , DNA (Citosina-5-)-Metiltransferases/fisiologia , Humanos , Imuno-Histoquímica , Neoplasias das Glândulas Salivares/metabolismo , Glândulas Salivares
11.
Proc Natl Acad Sci U S A ; 115(33): 8394-8399, 2018 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-30061411

RESUMO

The bladder's remarkable regenerative capacity had been thought to derive exclusively from its own progenitors. While examining consequences of DNA methyltransferase 1 (Dnmt1) inactivation in mouse embryonic bladder epithelium, we made the surprising discovery that Wolffian duct epithelial cells can support bladder regeneration. Conditional Dnmt1 inactivation in mouse urethral and bladder epithelium triggers widespread apoptosis, depletes basal and intermediate bladder cells, and disrupts uroplakin protein expression. These events coincide with Wolffian duct epithelial cell recruitment into Dnmt1 mutant urethra and bladder where they are reprogrammed to express bladder markers, including FOXA1, keratin 5, P63, and uroplakin. This is evidence that Wolffian duct epithelial cells are summoned in vivo to replace damaged bladder epithelium and function as a reservoir of cells for bladder regeneration.


Assuntos
Bexiga Urinária/fisiologia , Urotélio/fisiologia , Ductos Mesonéfricos/fisiologia , Animais , Animais Recém-Nascidos , Apoptose , Linhagem da Célula , DNA (Citosina-5-)-Metiltransferase 1/fisiologia , Dano ao DNA , Metilação de DNA , Células Epiteliais/fisiologia , Camundongos , Regeneração
12.
Cell Signal ; 51: 243-256, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30121333

RESUMO

It is well known that epithelial-mesenchymal transition (EMT) can confer cancer cells with invasive and migratory capabilities associated with distant metastasis. As a key upstream factor in the Hippo pathway, Kibra (wwc1 gene) has been shown to suppress EMT in breast cancer cells, and we have found that its expression is reduced or lost completely in both human breast cancer cell lines and clinical tissue samples, particularly in triple negative breast cancer (TNBC). Unfortunately, the molecular mechanisms underlying this progression-associated event remain to be elucidated. Epigenetic gene silencing is one of the most common causes of suppressed expression of tumor suppressor genes. Furthermore, recent studies have demonstrated that EZH2 can recruit DNA methyltransferases, resulting in DNA methylation and subsequent gene silencing in certain circumstances. Thus, we hypothesized that there may exist a link between EZH2 and DNA methylation in association with wwc1 silencing in breast cancer. To test this hypothesis, we performed bisulfite sequencing, shRNA, co-IP, ChIP, MeDIP and ChIP-qPCR. As expected, RG108 or 5-Aza treatment improved the wwc1 gene transcription and Kibra protein expression. Both bisulfite sequencing and MeDIP demonstrated higher CpG methylation of the wwc1 promoter the TNBC cells (MDA-MB-231) than in luminal breast cancer cells (MCF7). It is noteworthy that ChIP and co-IP assays showed that EZH2, H3K27me3 and DNMT1 are enriched at the wwc1 promoter, and there exist physiologically relevant protein-protein interactions between them. We also found that EZH2 knockdown leads to a partial increase in Kibra expression and a considerable reduction in H3K27 and DNMT1 trimethylation. Moreover, ChIP-qPCR revealed more DNA fragments containing the wwc1 promoter in MDA-MB-231 than in MCF7 cells after immunoprecipitation with EZH2, DNMT1 and H3K27me3 antibodies. Collectively, our results reveal crosstalk between H3K27me3 inhibition catalyzed by EZH2 and CpG island methylation mediated by DNMT1 within the wwc1 promoter, which synergistically silence wwc1 gene expression in TNBC. Based on these results, we conclude that EZH2 shows promise as a potential anti-tumor target.


Assuntos
DNA (Citosina-5-)-Metiltransferase 1/fisiologia , Proteína Potenciadora do Homólogo 2 de Zeste/fisiologia , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Histona Desmetilases com o Domínio Jumonji/fisiologia , Fosfoproteínas/genética , Neoplasias de Mama Triplo Negativas/genética , Movimento Celular , Ilhas de CpG , Metilação de DNA , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Transição Epitelial-Mesenquimal , Feminino , Via de Sinalização Hippo , Humanos , Células MCF-7 , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais
13.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 43(4): 415-420, 2018 Apr 28.
Artigo em Chinês | MEDLINE | ID: mdl-29774879

RESUMO

OBJECTIVE: To explore the role ofDNA methyltransferase 1 (DNMT1) in mouse skin aging.
 Methods: Epidermal conditional K14 Cre-mediated DNA methyltransferase 1 (DNMT1) knockout mice (Mut group, n=4) and the littermate normal mice with the same age (WT group) n=4) were used in this study. HE staining was used to detect the pathological changes of skin; the changes of number in the dermal elastic fibers were detected by Gomori aldehyde fuchsin staining, the number of 5-bromo-2-deoxyuridine (BrdU)-labeled transit amplifying cells (TAC) in epidermis were detected by immunohistochemical staining; the number of chlorodeoxyuridine (CldU)-label-retaining cells (LRC) in epidermis were detected by immunofluorescent staining.
 Results: Compared with the WT group, the skin showed premature aging symptoms in the Mut group concomitant with the decreased epidermal thickness as well as the number of dermal collagen fibers, while the increased dermal elastic fiber fracture. Compared with the WT group, the number of TAC in the epidermis was significantly increased (P<0.05), and the number of LRC was significantly decreased (P<0.05) in the Mut group.
 Conclusion: The phenotype of skin premature aging in epidermal stem cell conditional DNMT1-knockout mice suggests an important role of DNMT1 in skin aging.


Assuntos
Senilidade Prematura/patologia , DNA (Citosina-5-)-Metiltransferase 1/fisiologia , Envelhecimento da Pele/patologia , Senilidade Prematura/enzimologia , Animais , DNA (Citosina-5-)-Metiltransferase 1/genética , Células Epidérmicas , Camundongos , Camundongos Knockout , Envelhecimento da Pele/fisiologia
14.
Bone ; 111: 82-91, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29555308

RESUMO

Disuse osteoporosis (DOP) is a common complication of the lack of mechanical loading. The precise mechanism underlying DOP remains unknown, although epigenetic modifications may be a major cause. Recently, cumulative research has revealed that DNA methyltransferase (DNMT) proteins can catalyze the conversion of cytosine to 5-methylcytosine (5mC), altering the epigenetic state of DNA. Here, we report that DNMT1 expression and lncRNA-H19 methylation are upregulated in the femoral tissues of DOP rats, accompanied with inhibited Erk signaling pathway. Overexpression of DNMT1 in UMR-106 cells mimics 5mC enrichment in the H19 promoter, inhibition of Erk signaling and impairment of osteogenesis, which can be rescued by 5'-aza-deoxycytidine (5'-Aza) treatment. Moreover, local intramedullary injection of Dnmt1 siRNA (siDNMT1) in Sprague-Dawley (SD) rats abrogated disuse lncRNA-H19 (H19) downregulation, Erk signaling inhibition, histopathological changes, and bone microstructure declines in the distal femur in vivo. Therefore, our data identify for the first time a new signaling cascade in DOP: mechanical unloading causes upregulation of DNMT1 and hypermethylation of H19 promoter, which subsequently leads to downregulation of lncRNA-H19 and inhibition of the ERK signaling, suggesting a new potential therapeutic target.


Assuntos
DNA (Citosina-5-)-Metiltransferase 1/fisiologia , Epigênese Genética , Osteoporose/etiologia , Osteoporose/genética , RNA Longo não Codificante/genética , Animais , Linhagem Celular Tumoral , DNA (Citosina-5-)-Metiltransferase 1/genética , Metilação de DNA , Regulação para Baixo , Fêmur/ultraestrutura , Elevação dos Membros Posteriores , Sistema de Sinalização das MAP Quinases , Osteoporose/patologia , Osteoporose/terapia , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/uso terapêutico , Ratos , Ratos Sprague-Dawley , Regulação para Cima
15.
Epigenetics ; 12(9): 811-824, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-29099280

RESUMO

The DNA methylome is re-patterned during discrete phases of vertebrate development. In zebrafish, there are 2 waves of global DNA demethylation and re-methylation: the first occurs before gastrulation when the parental methylome is changed to the zygotic pattern and the second occurs after formation of the embryonic body axis, during organ specification. The occupancy of the histone variant H2A.Z and regions of DNA methylation are generally anti-correlated, and it has been proposed that H2A.Z restricts the boundaries of highly methylated regions. While many studies have described the dynamics of methylome changes during early zebrafish development, the factors involved in establishing the DNA methylation landscape in zebrafish embryos have not been identified. We test the hypothesis that the zebrafish ortholog of H2A.Z (H2afv) restricts DNA methylation during development. We find that, in control embryos, bulk genome methylation decreases after gastrulation, with a nadir at the bud stage, and peaks during mid-somitogenesis; by 24 hours post -fertilization, total DNA methylation levels return to those detected in gastrula. Early zebrafish embryos depleted of H2afv have significantly more bulk DNA methylation during somitogenesis, suggesting that H2afv limits methylation during this stage of development. H2afv deficient embryos are small, with multisystemic abnormalities. Genetic interaction experiments demonstrate that these phenotypes are suppressed by depletion of DNA methyltransferase 1 (Dnmt1). This work demonstrates that H2afv is essential for global DNA methylation reprogramming during early vertebrate development and that embryonic development requires crosstalk between H2afv and Dnmt1.


Assuntos
Metilação de DNA , Histonas/metabolismo , Peixe-Zebra/genética , Animais , DNA (Citosina-5-)-Metiltransferase 1/genética , DNA (Citosina-5-)-Metiltransferase 1/fisiologia , Variação Genética , Fenótipo , Peixe-Zebra/embriologia , Peixe-Zebra/crescimento & desenvolvimento , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/fisiologia
16.
Oncogene ; 36(50): 6919-6928, 2017 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-28869603

RESUMO

Overexpression of DNMT1 and KIT is prevalent in lung cancer, yet the underlying molecular mechanisms are poorly understood. While the deregulated activation of DNMT1 or KIT has been implicated in lung cancer pathogenesis, whether and how DNMT1 and KIT orchestrate lung tumorigenesis are unclear. Here, using human lung cancer tissue microarrays and fresh frozen tissues, we found that the overexpression of DNMT1 is positively correlated with the upregulation of KIT in tumor tissues. We demonstrated that DNMT1 and KIT form a positive regulatory loop, in which ectopic DNMT1 expression increases, whereas targeted DNMT1 depletion abrogates KIT signaling cascade through Sp1/miR-29b network. Conversely, an increase of KIT levels augments, but a reduction of KIT expression ablates DNMT1 transcription by STAT3 pathway leading to in-parallel modification of the DNA methylation profiles. We provided evidence that KIT inactivation induces global DNA hypomethylation, restores the expression of tumor suppressor p15INK4B through promoter demethylation; in turn, DNMT1 dysfunction impairs KIT kinase signaling. Functionally, KIT and DNMT1 co-expression promotes, whereas dual inactivation of them suppresses, lung cancer cell proliferation and metastatic growth in vitro and in vivo, in a synergistic manner. These findings demonstrate the regulatory and functional interplay between DNA methylation and tyrosine kinase signaling in propelling tumorigenesis, providing a widely applicable approach for targeting lung cancer.


Assuntos
DNA (Citosina-5-)-Metiltransferase 1/fisiologia , Neoplasias Pulmonares/patologia , Proteínas Proto-Oncogênicas c-kit/fisiologia , Animais , Linhagem Celular Tumoral , Metilação de DNA , Humanos , Neoplasias Pulmonares/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/fisiologia , Fator de Transcrição Sp1/fisiologia , Proteína Supressora de Tumor p53/fisiologia
17.
Neurobiol Learn Mem ; 135: 139-145, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27545441

RESUMO

DNA methylation has been shown to impact certain forms of synaptic and behavioral plasticity that have been implicated in the development in psychiatric disorders. DNA methylation is catalyzed by DNA methyltransferase (DNMT) enzymes that continue to be expressed in postmitotic neurons in the forebrain. Using a conditional forebrain knockout of DNMT1 or DNMT3a we assessed the role of these DNMTs in anxiety and depressive-like behavior in mice using an array of behavioral testing paradigms. Forebrain deletion of DNMT1 had anxiolytic and antidepressant-like properties as assessed by elevated plus maze, novelty suppressed feeding, forced swim, and social interaction tests. DNMT3a knockout mice, by contrast, did not exhibit significant behavioral alterations in these tests. Given the putative role of altered DNA methylation patterns in the development of schizophrenia, we also assessed DNMT1 and DNMT3a knockout mice in a prepulse inhibition task and found an enhanced prepulse inhibition of startle in DNMT1 knockouts relative to wild type mice, with no change evident in DNMT3a knockout mice. Our data suggest that DNMT1 and DNMT3a are distinctly involved in affective behavior and that DNMT1 may ultimately represent a potential target for treatment of certain affective behavioral disorders.


Assuntos
Ansiedade , Comportamento Animal/fisiologia , DNA (Citosina-5-)-Metiltransferase 1/fisiologia , DNA (Citosina-5-)-Metiltransferases/fisiologia , Depressão , Inibição Pré-Pulso , Prosencéfalo/metabolismo , Animais , Ansiedade/genética , Ansiedade/fisiopatologia , DNA (Citosina-5-)-Metiltransferase 1/genética , DNA (Citosina-5-)-Metiltransferases/genética , DNA Metiltransferase 3A , Depressão/genética , Depressão/fisiopatologia , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inibição Pré-Pulso/genética , Inibição Pré-Pulso/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...