Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Sci Rep ; 14(1): 7726, 2024 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-38565619

RESUMO

Decidualization can be induced by culturing human endometrial stromal cells (ESCs) with several decidualization stimuli, such as cAMP, medroxyprogesterone acetate (MPA) or Estradiol (E2). However, it has been unclear how decidualized cells induced by different stimuli are different. We compared transcriptomes and cellular functions of decidualized ESCs induced by different stimuli (MPA, E2 + MPA, cAMP, and cAMP + MPA). We also investigated which decidualization stimulus induces a closer in vivo decidualization. Differentially expressed genes (DEGs) and altered cellular functions by each decidualization stimuli were identified by RNA-sequence and gene-ontology analysis. DEGs was about two times higher for stimuli that use cAMP (cAMP and cAMP + MPA) than for stimuli that did not use cAMP (MPA and E2 + MPA). cAMP-using stimuli altered the cellular functions including angiogenesis, inflammation, immune system, and embryo implantation whereas MPA-using stimuli (MPA, E2 + MPA, and cAMP + MPA) altered the cellular functions associated with insulin signaling. A public single-cell RNA-sequence data of the human endometrium was utilized to analyze in vivo decidualization. The altered cellular functions by in vivo decidualization were close to those observed by cAMP + MPA-induced decidualization. In conclusion, decidualized cells induced by different stimuli have different transcriptome and cellular functions. cAMP + MPA may induce a decidualization most closely to in vivo decidualization.


Assuntos
Endométrio , Acetato de Medroxiprogesterona , Feminino , Humanos , Células Cultivadas , Endométrio/metabolismo , Acetato de Medroxiprogesterona/farmacologia , Células Estromais/metabolismo , Expressão Gênica , RNA/metabolismo , Decídua/metabolismo
2.
Obstet Gynecol ; 143(3): 358-365, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38061038

RESUMO

OBJECTIVE: To establish prediction models for the diagnosis of the subtypes of uterine leiomyomas by machine learning using magnetic resonance imaging (MRI) data. METHODS: This is a prospective observational study. Ninety uterine leiomyoma samples were obtained from 51 patients who underwent surgery for uterine leiomyomas. Seventy-one samples (49 mediator complex subunit 12 [ MED12 ] mutation-positive and 22 MED12 mutation-negative leiomyomas) were assigned to the primary data set to establish prediction models. Nineteen samples (13 MED12 mutation-positive and 6 MED12 mutation-negative leiomyomas) were assigned to the unknown testing data set to validate the prediction model utility. The tumor signal intensity was quantified by seven MRI sequences (T2-weighted imaging, apparent diffusion coefficient, magnetic resonance elastography, T1 mapping, magnetization transfer contrast, T2* blood oxygenation level dependent, and arterial spin labeling) that can estimate the collagen and water contents of uterine leiomyomas. After surgery, the MED12 mutations were genotyped. These results were used to establish prediction models based on machine learning by applying support vector classification and logistic regression for the diagnosis of uterine leiomyoma subtypes. The performance of the prediction models was evaluated by cross-validation within the primary data set and then finally evaluated by external validation using the unknown testing data set. RESULTS: The signal intensities of five MRI sequences (T2-weighted imaging, apparent diffusion coefficient, T1 mapping, magnetization transfer contrast, and T2* blood oxygenation level dependent) differed significantly between the subtypes. In cross-validation within the primary data set, both machine learning models (support vector classification and logistic regression) based on the five MRI sequences were highly predictive of the subtypes (area under the curve [AUC] 0.974 and 0.988, respectively). External validation with the unknown testing data set confirmed that both models were able to predict the subtypes for all samples (AUC 1.000, 100.0% accuracy). Our prediction models with T2-weighted imaging alone also showed high accuracy to discriminate the uterine leiomyoma subtypes. CONCLUSION: We established noninvasive prediction models for the diagnosis of the subtypes of uterine leiomyomas by machine learning using MRI data.


Assuntos
Leiomioma , Neoplasias Uterinas , Feminino , Humanos , Neoplasias Uterinas/diagnóstico por imagem , Neoplasias Uterinas/genética , Leiomioma/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos , Imagem de Difusão por Ressonância Magnética/métodos , Mutação
3.
Reprod Med Biol ; 22(1): e12548, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38107653

RESUMO

Purpose: To test the theory that invaginated ovarian surface epithelium and endometrial implants on the ovary form ovarian endometriomas. Methods: Adhesion sites of ovarian endometrioma on the peritoneum and consecutive ovarian endometrioma cyst wall, called non-adhesion sites, were histologically examined. DNA methylomes of the adhesion sites, non-adhesion sites, and blueberry spots were compared with those of ovary, endometrium, and peritoneum. Results: The non-adhesion sites showed an ovarian surface epithelium-like structure near the adhesion site, which continued to a columnar epithelium-like structure. Calretinin staining was strong in the ovarian surface epithelium-like structure but weak in the columnar epithelium-like structure. Estrogen receptors were absent in the ovarian surface epithelium-like structure, but present in the columnar epithelium-like structure. The adhesion sites had endometrial gland-like structures that expressed estrogen receptors. Analyses of DNA methylomes classified the non-adhesion sites and ovaries into the same group, suggesting that ovarian endometriomas originate from the ovarian surface epithelium. The adhesion sites, blueberry spots and peritoneum were classified in the same group, suggesting that the adhesion sites and blueberry spots originate from the peritoneum. Conclusions: The present results support the invagination theory. Ovarian endometriomas consist of invaginated ovarian surface epithelium with celomic metaplasia and endometrium implants on the peritoneum.

4.
Mol Hum Reprod ; 29(7)2023 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-37310913

RESUMO

Human endometrial stromal cells (hESCs) undergo a differentiation process with dramatic changes in cell functions during the menstrual cycle, which is called decidualization. This is an important event for implantation of the embryo and successful pregnancy. Defective decidualization can cause implantation failure, miscarriage, and unexplained infertility. A number of genes are upregulated or downregulated during decidualization. Recent studies have shown that epigenetic mechanisms are involved in the regulation of decidualization-related genes and that histone modifications occur throughout the genome during decidualization. The present review focuses on the involvement of genome-wide histone modifications in dramatic changes in gene expression during decidualization. The main histone modifications are the increases of H3K27ac and H3K4me3, which activate transcription. C/EBPß works as a pioneer factor throughout the genome by recruiting p300. This is the main cause of the genome-wide acetylation of H3K27 during decidualization. Histone modifications were observed in both the proximal promoter and distal enhancer regions. Genome editing experiments show that the distal regions have transcriptional activities, which suggests that decidualization induces the interactions between proximal promoter and distal enhancer regions. Taken together, these findings show that gene regulation during decidualization is closely associated with genome-wide changes of histone modifications. This review provides new insights regarding the cases of implantation failure in terms of decidualization insufficiency owing to epigenetic dysregulation, and may lead to novel treatment options for women with implantation failure.


Assuntos
Decídua , Endométrio , Gravidez , Humanos , Feminino , Endométrio/metabolismo , Decídua/metabolismo , Código das Histonas/genética , Expressão Gênica , Células Estromais/metabolismo
5.
J Obstet Gynaecol Res ; 49(7): 1827-1837, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37170675

RESUMO

AIM: Abnormal uterine bleeding, as proposed in 2007, is defined as unexpected uterine bleeding in women of reproductive age; the cause of the bleeding is categorized using the PALM-COEIN system. Identifying the diagnostic and treatment modalities for each cause might be intriguing. To summarize the options for abnormal uterine bleeding assessment, we employed text-mining analysis for each of its causes. METHODS: We analyzed abstracts based on PALM-COEIN from PubMed and Web of Science in March 2022. The literature was divided into categories; topics about the disorders were retrieved, and covalent network analysis was conducted to find information for evaluating abnormal uterine bleeding. RESULTS: Diagnostic approaches for PALM included histological and image analysis, including computerized tomography, magnetic resonance imaging, sonography, and hysteroscopy. The therapeutic approaches varied according to the cause. Diagnostic approaches for COEIN were mostly medical history interviews and blood sampling, and the therapeutic approaches for COEIN were ablation, hysteroscopy, and hormonal treatment. The PALM-COEIN classification co-occurrence search revealed each cause's diagnostic procedures, symptoms, and treatment procedures. CONCLUSION: Our text-mining methodology revealed comprehensive insights, important study themes, and clinical trends for abnormal uterine bleeding. A tailored approach to medical realities is required for treating abnormal uterine bleeding properly.


Assuntos
Doenças Uterinas , Feminino , Humanos , Gravidez , Doenças Uterinas/complicações , Hemorragia Uterina/diagnóstico , Hemorragia Uterina/etiologia , Hemorragia Uterina/terapia , Histeroscopia/efeitos adversos , Imageamento por Ressonância Magnética , Ultrassonografia
6.
Echocardiography ; 39(11): 1457-1461, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36258638

RESUMO

We present the case of a fetus with cardiac capillary hemangioma in the right atrial cavity. The tumor showed dramatic growth between the 28th and 32nd week of gestation and resulted in tachyarrhythmia. The patient was born at the 33 weeks of gestation weighing 2430 g via urgent cesarean section because the rapidly growing cardiac tumor caused incessant tachyarrhythmia, pericardial effusion, and fetal circulatory incompetence. Coronary angiography revealed that the right coronary artery drained into the tumor. Due to hemodynamic deterioration, the patient underwent subtotal resection of the tumor on the 2nd day after birth. Histopathological examination revealed an undifferentiated capillary hemangioma. The patient was discharged at the age of 86 days, as the tachyarrhythmia and hemodynamic incompetence had subsided; however, bradycardia and intermittent atrioventricular conduction disturbance gradually developed. Capillary hemangioma, a rare primary cardiac space-occupying tumor in children, can invade the conduction system.


Assuntos
Neoplasias Cardíacas , Hemangioma Capilar , Criança , Humanos , Gravidez , Feminino , Lactente , Cesárea , Neoplasias Cardíacas/complicações , Neoplasias Cardíacas/diagnóstico por imagem , Neoplasias Cardíacas/cirurgia , Hemangioma Capilar/complicações , Hemangioma Capilar/diagnóstico por imagem , Hemangioma Capilar/cirurgia , Taquicardia , Feto/patologia
7.
Sci Rep ; 12(1): 8912, 2022 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-35618793

RESUMO

Somatic mutations in Mediator complex subunit 12 (MED12m) have been reported as a biomarker of uterine fibroids (UFs). However, the role of MED12m is still unclear in the pathogenesis of UFs. Therefore, we investigated the differences in DNA methylome, transcriptome, and histological features between MED12m-positive and -negative UFs. DNA methylomes and transcriptomes were obtained from MED12m-positive and -negative UFs and myometrium, and hierarchically clustered. Differentially expressed genes in comparison with the myometrium and co-expressed genes detected by weighted gene co-expression network analysis were subjected to gene ontology enrichment analyses. The amounts of collagen fibers and the number of blood vessels and smooth muscle cells were histologically evaluated. Hierarchical clustering based on DNA methylation clearly separated the myometrium, MED12m-positive, and MED12m-negative UFs. MED12m-positive UFs had the increased activities of extracellular matrix formation, whereas MED12m-negative UFs had the increased angiogenic activities and smooth muscle cell proliferation. The MED12m-positive and -negative UFs had different DNA methylation, gene expression, and histological features. The MED12m-positive UFs form the tumor with a rich extracellular matrix and poor blood vessels and smooth muscle cells compared to the MED12m-negative UFs, suggesting MED12 mutations affect the tissue composition of UFs.


Assuntos
Epigenoma , Leiomioma , Feminino , Humanos , Leiomioma/patologia , Complexo Mediador/genética , Complexo Mediador/metabolismo , Mutação , Miométrio/metabolismo , Fatores de Transcrição/metabolismo , Transcriptoma
8.
J Biol Chem ; 298(5): 101874, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35358514

RESUMO

We previously reported that CCAAT/enhancer-binding protein beta (C/EBPß) is the pioneer factor inducing transcription enhancer mark H3K27 acetylation (H3K27ac) in the promoter and enhancer regions of genes encoding insulin-like growth factor-binding protein-1 (IGFBP-1) and prolactin (PRL) and that this contributes to decidualization of human endometrial stromal cells (ESCs). Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α; PPARGC1A) is a transcriptional coactivator known to regulate H3K27ac. However, although PGC-1α is expressed in ESCs, the potential role of PGC-1α in mediating decidualization is unclear. Here, we investigated the involvement of PGC-1α in the regulation of decidualization. We incubated ESCs with cAMP to induce decidualization and knocked down PPARGC1A to inhibit cAMP-induced expression of IGFBP-1 and PRL. We found cAMP increased the recruitment of PGC-1α and p300 to C/EBPß-binding sites in the promoter and enhancer regions of IGFBP-1 and PRL, corresponding with increases in H3K27ac. Moreover, PGC-1α knockdown inhibited these increases, suggesting PGC-1α forms a histone-modifying complex with C/EBPß and p300 at these regions. To further investigate the regulation of PGC-1α, we focused on C/EBPß upstream of PGC-1α. We found cAMP increased C/EBPß recruitment to the novel enhancer regions of PPARGC1A. Deletion of these enhancers decreased PGC-1α expression, indicating that C/EBPß upregulates PGC-1α expression by binding to novel enhancer regions. In conclusion, PGC-1α is upregulated by C/EBPß recruitment to novel enhancers and contributes to decidualization by forming a histone-modifying complex with C/EBPß and p300, thereby inducing epigenomic changes in the promoters and enhancers of IGFBP-1 and PRL.


Assuntos
Histonas , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , AMP Cíclico/metabolismo , Regulação da Expressão Gênica , Histonas/genética , Histonas/metabolismo , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Prolactina/genética , Prolactina/metabolismo , Células Estromais/metabolismo
9.
J Biol Chem ; 297(4): 101150, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34478711

RESUMO

Human endometrial stromal cells (ESCs) differentiate into decidual cells by the action of progesterone, which is essential for implantation and maintenance of pregnancy. We previously reported that glucose uptake by human ESCs increases during decidualization and that glucose is indispensable for decidualization. Although glucose transporter 1 (GLUT1) is upregulated during decidualization, it remains unclear whether it is involved in glucose uptake. Here, we attempted to determine the role of GLUT1 during decidualization as well as the factors underlying its upregulation. ESCs were incubated with cAMP to induce decidualization. Knockdown of GLUT1 suppressed cAMP-increased glucose uptake and the expressions of specific markers of decidualization, IGF-binding protein-1 (IGFBP-1), and prolactin (PRL). To investigate the regulation of GLUT1 expression, we focused on CCAAT enhancer-binding protein ß (C/EBPß) and Wilms' tumor 1 (WT1) as the upstream transcription factors regulating GLUT1 expression. Knockdown of either C/EBPß or WT1 suppressed cAMP-increased GLUT1 expression and glucose uptake. cAMP treatment also increased the recruitment of C/EBPß and WT1 to the GLUT1 promoter region. Interestingly, cAMP increased the H3K27 acetylation (H3K27ac) and p300 recruitment in the GLUT1 promoter region. Knockdown of C/EBPß or WT1 inhibited these events, indicating that both C/EBPß and WT1 contribute to the increase of H3K27ac by recruiting p300 to the GLUT1 promoter region during decidualization. These findings indicate that GLUT1 is involved in glucose uptake in ESCs during decidualization, thus facilitating the establishment of pregnancy.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Decídua/metabolismo , Epigênese Genética , Transportador de Glucose Tipo 1/biossíntese , Regulação para Cima , Proteínas WT1/metabolismo , Adulto , Proteína beta Intensificadora de Ligação a CCAAT/genética , Feminino , Transportador de Glucose Tipo 1/genética , Humanos , Pessoa de Meia-Idade , Células Estromais , Proteínas WT1/genética
10.
Mol Cell Endocrinol ; 520: 111085, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33232782

RESUMO

We previously reported that H3K27 acetylation (H3K27ac) increases throughout the genome during decidualization of human endometrial stromal cells (ESCs). However, its mechanisms have not been clarified. We also reported that C/EBPß acts as a pioneer factor initiating chromatin remodeling by increasing H3K27ac of IGFBP-1 and PRL promoters. Therefore, C/EBPß may be involved in the genome-wide increase of H3K27ac during decidualization. In this study, we investigated whether C/EBPß causes genome-wide H3K27ac modifications and regulates gene expressions during decidualization. cAMP was used to induce decidualization. Three types of cells (control cells, cAMP-treated cells, and cAMP-treated + C/EBPß-knockdowned cells by siRNA) were generated. Of 4190 genes that were upregulated by cAMP, C/EBPß knockdown inhibited these upregulation in 2239 genes (53.4%), indicating that they are under the regulation of C/EBPß. cAMP increased H3K27ac in 1272 of the 2239 genes. C/EBPß knockdown abolished the increase of H3K27ac in almost all genes (1263 genes, 99.3%), suggesting that C/EBPß can upregulate gene expression by increasing H3K27ac. To investigate how C/EBPß regulates H3K27ac throughout the genome, we tested the hypothesis that C/EBPß binds to its binding regions and recruits cofactors with histone acetyltransferase activities. To do this, we collated our ChIP-sequence data with public ChIP-sequence database of transcription factors, and found that p300 is the most likely cofactor that binds to the H3K27ac-increased-regions with C/EBPß. ChIP-qPCR of several genes confirmed that C/EBPß binds to the target regions, recruits p300, and increases H3K27ac. Our genome-wide analysis revealed that C/EBPß induces H3K27ac throughout the genome and upregulates gene expressions during decidualization by recruiting p300 to the promoters.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Decídua/metabolismo , Endométrio/citologia , Genoma Humano , Histonas/metabolismo , Lisina/metabolismo , Regulação para Cima/genética , Acetilação , Adulto , AMP Cíclico/metabolismo , Regulação para Baixo/genética , Proteína p300 Associada a E1A/metabolismo , Feminino , Humanos , Pessoa de Meia-Idade , Reprodutibilidade dos Testes , Células Estromais/metabolismo
11.
J Biol Chem ; 295(14): 4673-4683, 2020 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-32098869

RESUMO

We previously reported that the transcription factor Wilms tumor 1 (WT1) regulates the expression of insulin-like growth factor-binding protein-1 (IGFBP-1) and prolactin (PRL) during decidualization of human endometrial stromal cells (ESCs). However, other roles of WT1 in decidualization remain to be fully clarified. Here, we investigated how WT1 regulates the physiological functions of human ESCs during decidualization. We incubated ESCs isolated from proliferative-phase endometrium with cAMP to induce decidualization, knocked down WT1 with siRNA, and generated three types of treatments (nontreated cells, cAMP-treated cells, and cAMP-treated + WT1-knockdown cells). To identify WT1-regulated genes, we used gene microarrays and compared the transcriptome data obtained among these three treatments. We observed that WT1 up-regulates 121 genes during decidualization, including several genes involved in lipid transport. The WT1 knockdown inhibited lipid accumulation (LA) in the cAMP-induced ESCs. To examine the mechanisms by which WT1 regulates LA, we focused on very low-density lipoprotein receptor (VLDLR), which is involved in lipoprotein uptake. We found that cAMP up-regulates VLDLR and that the WT1 knockdown inhibits it. Results of ChIP assays revealed that cAMP increases the recruitment of WT1 to the promoter region of the VLDLR gene, indicating that WT1 regulates VLDLR expression. Moreover, VLDLR knockdown inhibited cAMP-induced LA, and VLDLR overexpression reverted the suppression of LA caused by the WT1 knockdown. Taken together, our results indicate that WT1 enhances lipid storage by up-regulating VLDLR expression in human ESCs during decidualization.


Assuntos
Metabolismo dos Lipídeos , Proteínas WT1/metabolismo , Adulto , Células Cultivadas , AMP Cíclico/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Endométrio/citologia , Feminino , Regulação da Expressão Gênica , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Pessoa de Meia-Idade , Regiões Promotoras Genéticas , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores de LDL/antagonistas & inibidores , Receptores de LDL/genética , Receptores de LDL/metabolismo , Células Estromais/citologia , Células Estromais/metabolismo , Proteínas WT1/antagonistas & inibidores , Proteínas WT1/genética
12.
J Assist Reprod Genet ; 36(11): 2385-2397, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31728810

RESUMO

PURPOSE: We attempted to identify the genes involved in the pathogenesis of uterine leiomyomas, under a hypothesis that the aberrant expression of upstream regulatory genes caused by aberrant DNA methylation is involved in the onset and development of uterine leiomyomas. METHODS: To find such genes, we compared genome-wide mRNA expression and DNA methylation in uterine leiomyomas and adjacent normal myometrium. Analysis of the data by Ingenuity Pathway Analysis software identified SATB2 which is known to be an epigenetic regulator, and NRG1 as candidate upstream regulatory genes. To infer the functions of these genes, human uterine smooth muscle cell lines overexpressing SATB2 or NRG1 genes were established (SATB2 or NRG1 lines), and their transcriptomes and pathways were analyzed. RESULTS: SATB2 and NRG1 were confirmed to be hypermethylated and upregulated in most uterine leiomyoma specimens (nine to 11 of the 11 cases). Among the established cell lines, morphological changes from spindle-like forms to fibroblast-like forms with elongated protrusions were observed in only the SATB2 line. Pathway analysis revealed that WNT/ß-catenin and TGF-ß signaling pathways which are related to the pathogenesis of uterine leiomyomas were activated in both SATB2 and NRG1 lines. In addition, signaling of growth factors including VEGF, PDGF, and IGF1, and retinoic acid signaling were activated in the SATB2 and NRG1 lines, respectively. CONCLUSIONS: These results indicate that SATB2 and NRG1 overexpression induced many of the signaling pathways that are considered to be involved in the pathogenesis of uterine leiomyomas, suggesting that these genes have roles as upstream regulatory factors.


Assuntos
Leiomioma/metabolismo , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Receptor Nogo 1/metabolismo , Fatores de Transcrição/metabolismo , Neoplasias Uterinas/metabolismo , Adulto , Metilação de DNA/fisiologia , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Pessoa de Meia-Idade , Mutação/fisiologia , Miométrio/metabolismo , Transdução de Sinais/fisiologia , Regulação para Cima/fisiologia
13.
BMC Genomics ; 20(1): 324, 2019 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-31035926

RESUMO

BACKGROUND: During decidualization in endometrial stromal cells (ESCs), expressions of a number of genes and epigenetic modifications of histones are altered. However, there is little information about whether DNA methylation, which is another epigenetic mechanism, also changes during decidualization. Here, we examined the genome-wide DNA methylation profiles in ESCs during decidualization and their associations with the changes of gene expressions and histone modifications. RESULTS: ESCs were incubated with estradiol and medroxyprogesterone acetate for 14 days to induce decidualization. The genome-wide DNA methylation profiles were compared between the non-decidualized ESCs and the decidualized ESCs. Of 482,005 CpGs, only 23 CpGs (0.0048%) showed different DNA methylation statuses. The DNA methylation statuses of the differentially expressed genes and the regions with different histone modifications (H3K4 tri-methylation and H3K27 acetylation) were also compared between the ESCs. In the upregulated and downregulated genes in decidualized ESCs, DNA methylation statuses around the promoter region of the genes did not significantly differ between the ESCs. In the regions with different histone modification, DNA methylation statuses did not differ between the ESCs. The differentially expressed genes and the differential histone modification regions were hypomethylated. CONCLUSIONS: Culturing ESCs with estrogen/progesterone did not distort the physiological pattern of DNA methylation, although mRNA expression and histone modifications were dynamically altered. A genome-wide DNA methylation analysis revealed stable DNA methylation statuses during decidualization in human endometrial stromal cells. DNA hypomethylation is maintained for the variable changes of histone modifications and gene expression during decidualization.


Assuntos
Metilação de DNA , Genoma Humano , Células Cultivadas , Ilhas de CpG , Metilação de DNA/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Endométrio/citologia , Estradiol/farmacologia , Feminino , Histonas/metabolismo , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Acetato de Medroxiprogesterona/farmacologia , Células Estromais/citologia , Células Estromais/metabolismo , Regulação para Cima/efeitos dos fármacos
14.
Reproduction ; 157(5): 457-464, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30817321

RESUMO

Decidualization stimuli activate the insulin signaling pathway and increase the glucose uptake in human endometrial stromal cells (ESCs). The inductions of prolactin (PRL) and IGF-binding protein-1 (IGFBP1), specific markers of decidualization, were inhibited by incubating ESCs under low glucose concentrations. These results suggested that decidualization stimuli activate the insulin signaling pathway, which contributes to decidualization through the increase of glucose uptake. Here, we investigated the mechanisms by which glucose regulates decidualization. ESCs were incubated with cAMP to induce decidualization. We examined whether low glucose affects the expression levels of transcription factors that induce decidualization. Forkhead box O1 (FOXO1) expression was significantly suppressed under low glucose conditions. Knockdown of FOXO1 by siRNA inhibited the expression levels of PRL and IGFBP1 during decidualization. Taken together, our results showed that low glucose inhibits decidualization by decreasing FOXO1 expression. We also examined the levels of histone H3K27 acetylation (H3K27ac), which is related to active transcription, of the promoter regions of FOXO1, PRL and IGFBP1 by ChIP assay. The H3K27ac levels of these promoter regions were increased by decidualization under normal glucose conditions, but not under low glucose conditions. Thus, our results show that glucose is indispensable for decidualization by activating the histone modification status of the promoters of PRL, IGFBP1 and FOXO1.


Assuntos
Decídua/efeitos dos fármacos , Glucose/farmacologia , Histona Acetiltransferases/metabolismo , Histonas/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Células Estromais/efeitos dos fármacos , Acetilação/efeitos dos fármacos , Adulto , Células Cultivadas , Decídua/metabolismo , Implantação do Embrião/efeitos dos fármacos , Implantação do Embrião/genética , Endométrio/efeitos dos fármacos , Endométrio/metabolismo , Feminino , Histonas/efeitos dos fármacos , Humanos , Insulina/metabolismo , Pessoa de Meia-Idade , Cultura Primária de Células , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Células Estromais/metabolismo
15.
J Ovarian Res ; 12(1): 14, 2019 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-30728052

RESUMO

BACKGROUND: In ovarian endometriomas (OE), the expression statuses of various steroid hormone receptors are altered compared with their expression statuses in eutopic endometrium (EE). For example, in OE, the expressions of estrogen receptor 1 (ESR1), which encodes ERα, and progesterone receptor (PGR) are downregulated, while the expression of ESR2, which encodes ERß, is upregulated. The causes of these changes are unclear. DNA methylation of a specific region of a gene can result in tissue-specific gene expression. Such regions are called tissue-dependent and differentially methylated regions (T-DMRs). We previously reported that the tissue-specific expression of ESR1 is regulated by DNA methylation of a T-DMR in normal tissues. In the present study, we examined whether aberrant DNA methylation of the T-DMR is associated with the altered expressions of ESR1, ESR2 and PGR in OE. RESULTS: Gene expression levels of ESR1, ESR2 and PGR were measured by quantitative RT-PCR. The expression levels of ESR1 and PGR were significantly lower and the expression level of ESR2 was significantly higher in OE than in EE. DNA methylation statuses were examined with an Infinium HumanMethylation450K BeadChip and sodium bisulfite sequencing. DNA methylation at the T-DMRs of ESR1 were significantly higher in OE than in EE, but no significant differences were observed in the DNA methylation statuses of ESR2 and PGR. CONCLUSIONS: Aberrant DNA methylation of the T-DMR was associated with the impaired expression of ESR1, but not the altered expressions of ESR2 and PGR, in OE.


Assuntos
Metilação de DNA/genética , Endometriose/genética , Receptor alfa de Estrogênio/genética , Regulação da Expressão Gênica/genética , Doenças Ovarianas/genética , Adulto , Endometriose/metabolismo , Endometriose/patologia , Endométrio/metabolismo , Endométrio/patologia , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Feminino , Humanos , Doenças Ovarianas/metabolismo , Doenças Ovarianas/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo
16.
Sci Rep ; 9(1): 714, 2019 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-30679486

RESUMO

The ovulatory LH-surge increases Vegf gene expression in granulosa cells (GCs) undergoing luteinization during ovulation. To understand the factors involved in this increase, we examined the roles of two transcription factors and epigenetic mechanisms in rat GCs. GCs were obtained from rats treated with eCG before, 4 h, 8 h, 12 h and 24 h after hCG injection. Vegf mRNA levels gradually increased after hCG injection and reached a peak at 12 h. To investigate the mechanism by which Vegf is up-regulated after hCG injection, we focused on C/EBPß and HIF1α. Their protein expression levels were increased at 12 h. The binding activity of C/EBPß to the Vegf promoter region increased after hCG injection whereas that of HIF1α did not at this time point. The C/EBPß binding site had transcriptional activities whereas the HIF1α binding sites did not have transcriptional activities under cAMP stimulation. The levels of H3K9me3 and H3K27me3, which are transcriptional repression markers, decreased in the C/EBPß binding region after hCG injection. The chromatin structure of this region becomes looser after hCG injection. These results show that C/EBPß regulates Vegf gene expression with changes in histone modifications and chromatin structure of the promoter region in GCs undergoing luteinization during ovulation.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Regulação da Expressão Gênica , Células da Granulosa/metabolismo , Luteinização/fisiologia , Ovulação/fisiologia , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteína beta Intensificadora de Ligação a CCAAT/genética , Células Cultivadas , Epigênese Genética , Feminino , Granuloma de Células Gigantes/genética , Granuloma de Células Gigantes/metabolismo , Granuloma de Células Gigantes/patologia , Células da Granulosa/citologia , Código das Histonas , Processamento de Proteína Pós-Traducional , Ratos , Ratos Sprague-Dawley , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
J Biol Chem ; 293(14): 5270-5280, 2018 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-29453285

RESUMO

We have previously shown that decidualization of human endometrial stromal cells (ESCs) causes a genome-wide increase in the levels of acetylation of histone-H3 Lys-27 (H3K27ac). We also reported that the distal gene regions, more than 3 kb up- or downstream of gene transcription start sites have increased H3K27ac levels. Insulin-like growth factor-binding protein-1 (IGFBP-1) is a specific decidualization marker and has increased H3K27ac levels in its distal upstream region (-4701 to -7501 bp). Here, using a luciferase reporter gene construct containing this IGFBP-1 upstream region, we tested the hypothesis that it is an IGFBP-1 enhancer. To induce decidualization, we incubated ESCs with cAMP and found that cAMP increased luciferase expression, indicating that decidualization increased the transcriptional activity from the IGFBP-1 upstream region. Furthermore, CRISPR/Cas9-mediated deletion of this region in HepG2 cells significantly reduced IGFBP-1 expression, confirming its role as an IGFBP-1 enhancer. A ChIP assay revealed that cAMP increased the recruitment of the transcriptional regulators CCAAT enhancer-binding protein ß (C/EBPß), forkhead box O1 (FOXO1), and p300 to the IGFBP-1 enhancer in ESCs. Of note, C/EBPß knockdown inhibited the stimulatory effects of cAMP on the levels of H3K27ac, chromatin opening, and p300 recruitment at the IGFBP-1 enhancer. These results indicate that the region -4701 to -7501 bp upstream of IGFBP-1 functions as an enhancer for IGFBP-1 expression in ESCs undergoing decidualization, that C/EBPß and FOXO1 bind to the enhancer region to up-regulate IGFBP-1 expression, and that C/EBPß induces H3K27ac by recruiting p300 to the IGFBP-1 enhancer.


Assuntos
Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Acetilação , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Sistemas CRISPR-Cas , AMP Cíclico/metabolismo , Decídua/metabolismo , Proteína p300 Associada a E1A , Implantação do Embrião , Endométrio/metabolismo , Elementos Facilitadores Genéticos/genética , Células Epiteliais/metabolismo , Feminino , Proteína Forkhead Box O1 , Regulação da Expressão Gênica/genética , Células Hep G2/metabolismo , Humanos , Prolactina/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Células Estromais/metabolismo , Transcriptoma/genética
18.
Endocrinology ; 158(10): 3696-3707, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28977591

RESUMO

The Wilms tumor suppressor gene (WT1) encodes an essential transcription factor regulating mammalian urogenital development. However, the function of WT1 in human endometrium is still unclear. The current study examined the involvement of WT1 in the regulation of IGF-binding protein-1 (IGFBP-1) and prolactin (PRL), which are specific markers of decidualization, in human endometrial stromal cells (ESCs) undergoing decidualization. ESCs isolated from proliferative-phase endometrium were incubated with cyclic adenosine monophosphate (cAMP) to induce decidualization. cAMP increased WT1 expression with the induction of IGFBP-1 and PRL. Knockdown of WT1 by small interfering RNA inhibited cAMP-induced expression of IGFBP-1 and PRL. cAMP also induced the recruitment of WT1 to the IGFBP-1 and PRL promoters. To investigate the mechanism by which WT1 is upregulated by cAMP, we focused on C/EBPß, a gene that regulates the expression of many genes during decidualization. Knockdown of C/EBPß decreased cAMP-increased WT1 expression. cAMP increased the recruitment of C/EBPß to the WT1 enhancer that is located approximately 14,000 bp downstream from the transcription start site. To test the endogenous function of the WT1 enhancer region on WT1 expression, the endogenous WT1 enhancer region was deleted by CRISPR/Cas9 system in HEK293 cells. The increase of WT1 expression by cAMP was not observed in the enhancer-deleted clones. Chromatin immunoprecipitation assay revealed that this enhancer region has high levels of H3K27ac and H3K4me1, which are active enhancer marks. These results show the role of WT1 in regulating decidualization in human ESCs. C/EBPß is an upstream gene that regulates WT1 expression by binding to the novel enhancer region.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/genética , AMP Cíclico/metabolismo , Decídua/metabolismo , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Prolactina/metabolismo , Células Estromais/metabolismo , Proteínas WT1/genética , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Sistemas CRISPR-Cas , Imunoprecipitação da Cromatina , Decídua/citologia , Endométrio/citologia , Endométrio/metabolismo , Elementos Facilitadores Genéticos , Feminino , Técnicas de Silenciamento de Genes , Células HEK293 , Histonas/metabolismo , Humanos , Proteínas WT1/metabolismo
19.
Sci Rep ; 6: 30652, 2016 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-27498619

RESUMO

Differential diagnosis of uterine leiomyomas and leiomyosarcomas is needed to determine whether the uterus can be retained. Therefore, biomarkers for uterine leiomyomas, and reliable and objective diagnostic methods have been desired besides the pathological diagnosis. In the present study, we identified 12 genes specific to uterine leiomyomas based on DNA methylation. Using these marker genes specific to uterine leiomyomas, we established a hierarchical clustering system based on the DNA methylation level of the marker genes, which could completely differentiate between uterine leiomyomas and normal myometrium. Furthermore, our hierarchical clustering system completely discriminated uterine cancers and differentiated between uterine leiomyosarcomas and leiomyomas with more than 70% accuracy. In conclusion, this study identified DNA methylation-based marker genes specific to uterine leiomyomas, and our hierarchical clustering system using these marker genes was useful for differential diagnosis of uterine leiomyomas and leiomyosarcomas.


Assuntos
Biomarcadores Tumorais/genética , Leiomioma/diagnóstico , Leiomioma/genética , Leiomiossarcoma/diagnóstico , Leiomiossarcoma/genética , Miométrio/patologia , Neoplasias Uterinas/diagnóstico , Neoplasias Uterinas/genética , Linhagem Celular Tumoral , DNA/genética , Metilação de DNA/genética , Diagnóstico Diferencial , Feminino , Marcadores Genéticos/genética , Humanos , Leiomioma/patologia , Leiomiossarcoma/patologia , Complexo Mediador/genética , Miométrio/metabolismo , Neoplasias Uterinas/patologia
20.
Mol Endocrinol ; 30(3): 335-47, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26683811

RESUMO

The mechanism controlling tissue-specific expression of estrogen receptor 1 (ESR1) is unclear. In other genes, DNA methylation of a region called the tissue-dependent and differentially methylated region (T-DMR) has been associated with tissue-specific gene expression. This study investigated whether human ESR1 has a T-DMR and whether DNA methylation of the T-DMR regulates its expression. ESR1 expression was tissue-specific, being high in the endometrium and mammary gland and low/nil in the placenta and skin. Therefore, DNA methylation profiles of the promoter of ESR1 were analyzed in these tissues and in breast cancer tissues. In all of the normal tissues, the proximal promoter regions were unmethylated. On the other hand, the distal regions (T-DMR) were unmethylated in the endometrium and mammary gland, but were moderately methylated and hypermethylated in the placenta and skin, respectively. T-DMR-methylated reporter assay was performed to examine whether DNA methylation at the T-DMR suppresses ESR1 transcription. T-DMR, but not the promoter region, had transcriptional activities and DNA methylation of the T-DMR suppressed ESR1 transcription. Early growth response protein 1 was shown to be a possible transcription factor to bind the T-DMR and up-regulate ESR1 expression. ESR1 has several upstream exons, and each upstream exon, Exon-A/Exon-B/Exon-C, had its own T-DMR. In some breast cancer cases and breast cancer cell lines, ESR1 expression was not regulated by DNA methylation at T-DMR as it is in normal tissues. In conclusion, ESR1 has a T-DMR. DNA methylation status at the T-DMR is involved in tissue-specific ESR1 expression in normal tissues but not always in breast cancer.


Assuntos
Metilação de DNA/genética , Receptor alfa de Estrogênio/genética , Especificidade de Órgãos/genética , Sequência de Bases , Neoplasias da Mama/genética , Receptor alfa de Estrogênio/metabolismo , Éxons/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Genes Reporter , Histonas/metabolismo , Humanos , Células MCF-7 , Regiões Promotoras Genéticas , Ligação Proteica , Processamento de Proteína Pós-Traducional , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA