Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Int J Mol Sci ; 24(15)2023 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-37569842

RESUMO

Liver X receptors (LXRα and LXRß) are oxysterol-activated nuclear receptors that play key roles in cholesterol homeostasis, the central nervous system, and the immune system. We have previously reported that LXRαß-deficient mice are more susceptible to dextran sodium sulfate (DSS)-induced colitis than their WT littermates, and that an LXR agonist protects against colitis in mice mainly via the regulation of the immune system in the gut. We now report that both LXRα and LXRß are expressed in the colonic epithelium and that in aging LXRαß-/- mice there is a reduction in the intensity of goblet cells, mucin (MUC2), TFF3, and estrogen receptor ß (ERß) levels. The cytoplasmic compartment of the surface epithelial cells was markedly reduced and there was a massive invasion of macrophages in the lamina propria. The expression and localization of ß-catenin, α-catenin, and E-cadherin were not changed, but the shrinkage of the cytoplasm led to an appearance of an increase in staining. In the colonic epithelium there was a reduction in the expression of plectin, a hemidesmosome protein whose loss in mice leads to spontaneous colitis, ELOVL1, a fatty acid elongase protein coding gene whose overexpression is found in colorectal cancer, and non-neuronal choline acetyltransferase (ChAT) involved in the regulation of epithelial cell adhesion. We conclude that in aging LXRαß-/- mice, the phenotype in the colon is due to loss of ERß expression.


Assuntos
Colite , Receptor beta de Estrogênio , Camundongos , Animais , Receptor beta de Estrogênio/metabolismo , Camundongos Knockout , Colite/induzido quimicamente , Colite/genética , Colite/metabolismo , Colo/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Mucosa Intestinal/metabolismo , Sulfato de Dextrana/toxicidade , Camundongos Endogâmicos C57BL
2.
Neuroendocrinology ; 111(1-2): 27-44, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-31991411

RESUMO

BACKGROUND: Male estrogen receptor beta (ERß) knockout (BERKO) mice display anxiety and aggression linked to, among others, altered serotonergic signaling in the basolateral amygdala and dorsal raphe, impaired cortical radial glia migration, and reduced GABAergic signaling. The effects on primary motor cortex (M1 cortex) and locomotor activity as a consequence of ERß loss have not been investigated. OBJECTIVE: The aim of this study was to determine whether locomotor activity is altered as a consequence of the changes in the M1 cortex. METHODS: The locomotor activity of male wild-type (WT) and BERKO mice was evaluated using the open-field and rotarod tests. Molecular changes in the M1 cortex were analyzed by RNA sequencing, electron microscopy, electrophysiology, and immunohistological techniques. In addition, we established oligodendrocyte (OL) cultures from WT and BERKO mouse embryonic stem cells to evaluate OL function. RESULTS: Locomotor profiling revealed that BERKO mice were more active than WT mice but had impaired motor coordination. Analysis of the M1 cortex pointed out differences in synapse function and myelination. There was a reduction in GABAergic signaling resulting in imbalanced excitatory and inhibitory neurotransmission as well as a defective OL differentiation accompanied by myelin defects. The effects of ERß loss on OL differentiation were confirmed in vitro. CONCLUSION: ERß is an important regulator of GABAergic interneurons and OL differentiation, which impacts on adult M1 cortex function and may be linked to increased locomotor activity and decreased motor coordination in BERKO mice.


Assuntos
Receptor beta de Estrogênio/genética , Locomoção/genética , Córtex Motor/fisiopatologia , Bainha de Mielina/fisiologia , Desempenho Psicomotor , Transmissão Sináptica , Animais , Expressão Gênica , Masculino , Camundongos , Camundongos Knockout , Córtex Motor/metabolismo , Oligodendroglia/patologia
3.
Proc Natl Acad Sci U S A ; 114(48): E10428-E10437, 2017 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-29133394

RESUMO

The ability to propagate mature cells and tissue from pluripotent stem cells offers enormous promise for treating many diseases, including neurodegenerative diseases. Before such cells can be used successfully in neurodegenerative diseases without causing unwanted cell growth and migration, genes regulating growth and migration of neural stem cells need to be well characterized. Estrogen receptor beta (ERß) is essential for migration of neurons and glial cells in the developing mouse brain. To examine whether ERß influences differentiation of mouse embryonic stem cells (mESC) into neural lineages, we compared control and ERß knockout (BERKO) mESCs at defined stages of neural development and examined the effects of an ERß-selective ligand (LY3201) with a combination of global and targeted gene-expression profiling and the expression of key pluripotency markers. We found that ERß was induced in embryoid bodies (EBs) and neural precursor cells (NPCs) during development. Proliferation was higher in BERKO NPCs and was inhibited by LY3201. Neurogenesis was reduced in BERKO ES cells, and oligodendrogliogenesis was enhanced. BERKO EBs expressed higher levels of key ectodermal and neural progenitor markers and lower levels of markers for mesoderm and endoderm lineages. ERß-regulated factors are involved in cell adhesion, axon guidance, and signaling of Notch and GABA receptor pathways, as well as factors important for the differentiation of neuronal precursors into dopaminergic neurons (Engrailed 1) and for the oligodendrocyte fate acquisition (Olig2). Our data suggest that ERß is an important component for differentiation into midbrain neurons as well as for preventing precocious oligodendrogliogenesis.


Assuntos
Diferenciação Celular/fisiologia , Receptor beta de Estrogênio/fisiologia , Mesencéfalo/fisiologia , Células-Tronco Embrionárias Murinas/fisiologia , Células-Tronco Neurais/fisiologia , Regeneração/fisiologia , Animais , Benzopiranos/farmacologia , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Neurônios Dopaminérgicos/fisiologia , Receptor beta de Estrogênio/agonistas , Feminino , Perfilação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Mesencéfalo/citologia , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese/fisiologia , Fator de Transcrição 2 de Oligodendrócitos/metabolismo , Oligodendroglia/fisiologia , Transdução de Sinais/fisiologia
4.
Mol Pharmacol ; 94(4): 1220-1231, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30115672

RESUMO

Recent meta-analyses found an association between prenatal exposure to the antidepressant fluoxetine (FLX) and an increased risk of autism in children. This developmental disorder has been related to dysfunctions in the brains' rewards circuitry, which, in turn, has been linked to dysfunctions in dopaminergic (DA) signaling. The present study investigated if FLX affects processes involved in dopaminergic neuronal differentiation. Mouse neuronal precursors were differentiated into midbrain dopaminergic precursor cells (mDPCs) and concomitantly exposed to clinically relevant doses of FLX. Subsequently, dopaminergic precursors were evaluated for expression of differentiation and stemness markers using quantitative polymerase chain reaction. FLX treatment led to increases in early regional specification markers orthodenticle homeobox 2 (Otx2) and homeobox engrailed-1 and -2 (En1 and En2). On the other hand, two transcription factors essential for midbrain dopaminergic (mDA) neurogenesis, LIM homeobox transcription factor 1 α (Lmx1a) and paired-like homeodomain transcription factor 3 (Pitx3) were downregulated by FLX treatment. The stemness marker nestin (Nes) was increased, whereas the neuronal differentiation marker ß3-tubulin (Tubb3) decreased. Additionally, we observed that FLX modulates the expression of several genes associated with autism spectrum disorder and downregulates the estrogen receptors (ERs) α and ß Further investigations using ERß knockout (BERKO) mDPCs showed that FLX had no or even opposite effects on several of the genes analyzed. These findings suggest that FLX affects differentiation of the dopaminergic system by increasing production of dopaminergic precursors, yet decreasing their maturation, partly via interference with the estrogen system.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Neurônios Dopaminérgicos/efeitos dos fármacos , Fluoxetina/farmacologia , Mesencéfalo/efeitos dos fármacos , Animais , Transtorno do Espectro Autista/metabolismo , Células Cultivadas , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Regulação para Baixo/efeitos dos fármacos , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteínas de Homeodomínio/metabolismo , Mesencéfalo/metabolismo , Camundongos , Neurogênese/efeitos dos fármacos , Fatores de Transcrição Otx/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Tubulina (Proteína)/metabolismo
5.
Reproduction ; 151(3): 253-9, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26700939

RESUMO

Estrogen receptor beta (ERß/ESR2) has a central role in mouse ovaries, as ERß knockout (BERKO) mice are subfertile due to an increase in fibrosis around the maturing follicle and a decrease in blood supply. This has a consequence that these follicles rarely rupture to release the mature oocyte. Matrix metalloproteinases (MMPs) are modulators of the extracellular matrix, and the expression of one specific MMP, MMP-19, is normally increased in granulosa cells during their maturation until ovulation. In this study, we demonstrate that MMP-19 levels are downregulated in BERKO mouse ovaries. Using human MCF-7 cells that overexpress ERß, we could identify MMP-19 to be a transcriptional target of ligand-bound activated ERß acting on a specificity protein-1 binding site. These data provide a molecular explanation for the observed follicle rupture defect that contributes to the subfertility of female BERKO mice.


Assuntos
Receptor beta de Estrogênio/metabolismo , Metaloproteinases da Matriz Secretadas/metabolismo , Ovário/metabolismo , Animais , Gonadotropina Coriônica , Regulação para Baixo , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Células MCF-7 , Metaloproteinases da Matriz Secretadas/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovulação , Fator de Transcrição Sp1/metabolismo
6.
Biochem Biophys Res Commun ; 466(4): 650-5, 2015 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-26393907

RESUMO

Estrogen receptor alpha (ERα) is an important regulator of the estrous cycle and mice with global ERα deletion, as well as some conditional knockout mouse lines, have an interruption in the estrous cycle. In this study we observed that conditional ERα knockout mice where the Cre gene is regulated by the rat insulin promoter (RIP), RIP-Cre/ERα(KO) mice, have a 3.7-fold increase in serum 17ß-estradiol levels, blocked estrous cycle, and develop a fluid-filled uterus (hydrometra). Using a proteomics approach, we identified three proteins, lactoferrin, complement C3 and chitinase 3-like protein 1 (CHI3L1), as highly expressed proteins in hydrometra fluid. The mRNA levels of the corresponding genes were more than 50-fold higher in RIP-Cre/ERα(KO) uterus compared to controls. High expression of CHI3L1 in the uterine fluid was not reflected as elevated levels in the serum. The high expression of lactoferrin, complement C3 and CHI3L1 in the uterine fluid, in association with elevated estrogen levels, prompted us to address if the expression of these genes is related to reproduction. However, gonadotropin treatment of mice reduced the uterine expression of these genes in a model of in vitro fertilization. Our findings identify lactoferrin, complement C3 and CHI3L1 as highly expressed proteins in hydrometra fluid in association with chronically elevated serum estradiol levels.


Assuntos
Glicoproteínas/metabolismo , Serpinas/metabolismo , Útero/metabolismo , Animais , Proteína 1 Semelhante à Quitinase-3 , Complemento C3/genética , Complemento C3/metabolismo , Estradiol/sangue , Receptor alfa de Estrogênio/deficiência , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Ciclo Estral/sangue , Ciclo Estral/genética , Ciclo Estral/metabolismo , Feminino , Expressão Gênica , Glicoproteínas/sangue , Glicoproteínas/genética , Lactoferrina/genética , Lactoferrina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Serpinas/genética , Doenças Uterinas/genética , Doenças Uterinas/metabolismo
7.
Sci Adv ; 10(4): eadj1354, 2024 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-38266095

RESUMO

The brain-specific enzyme CYP46A1 controls cholesterol turnover by converting cholesterol into 24S-hydroxycholesterol (24OH). Dysregulation of brain cholesterol turnover and reduced CYP46A1 levels are observed in Alzheimer's disease (AD). In this study, we report that CYP46A1 overexpression in aged female mice leads to enhanced estrogen signaling in the hippocampus and improved cognitive functions. In contrast, age-matched CYP46A1 overexpressing males show anxiety-like behavior, worsened memory, and elevated levels of 5α-dihydrotestosterone in the hippocampus. We report that, in neurons, 24OH contributes to these divergent effects by activating sex hormone signaling, including estrogen receptors. CYP46A1 overexpression in female mice protects from memory impairments induced by ovariectomy while having no effects in gonadectomized males. Last, we measured cerebrospinal fluid levels of 24OH in a clinical cohort of patients with AD and found that 24OH negatively correlates with neurodegeneration markers only in women. We suggest that CYP46A1 activation is a valuable pharmacological target for enhancing estrogen signaling in women at risk of developing neurodegenerative diseases.


Assuntos
Doença de Alzheimer , Transtornos da Memória , Masculino , Feminino , Humanos , Animais , Camundongos , Idoso , Colesterol 24-Hidroxilase , Transtornos da Memória/etiologia , Colesterol , Cognição , Doença de Alzheimer/genética , Estrogênios
8.
Antioxidants (Basel) ; 11(2)2022 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-35204179

RESUMO

Glutathione is an important antioxidant that plays a crucial role in the cellular protection against oxidative stress and detoxification of electrophilic mutagens, and carcinogens. Glutathione transferases are enzymes catalyzing glutathione-dependent reactions that lead to inactivation and conjugation of toxic compounds, processes followed by subsequent excretion of the detoxified products. Degeneration and loss of neuromelanin-containing dopaminergic neurons in the nigrostriatal neurons generally involves oxidative stress, neuroinflammation, alpha-synuclein aggregation to neurotoxic oligomers, mitochondrial dysfunction, protein degradation dysfunction, and endoplasmic reticulum stress. However, it is still unclear what triggers these neurodegenerative processes. It has been reported that aminochrome may elicit all of these mechanisms and, interestingly, aminochrome is formed inside neuromelanin-containing dopaminergic neurons during neuromelanin synthesis. Aminochrome is a neurotoxic ortho-quinone formed in neuromelanin synthesis. However, it seems paradoxical that the neurotoxin aminochrome is generated during neuromelanin synthesis, even though healthy seniors have these neurons intact when they die. The explanation of this paradox is the existence of protective tools against aminochrome neurotoxicity composed of the enzymes DT-diaphorase, expressed in these neurons, and glutathione transferase M2-2, expressed in astrocytes. Recently, it has been reported that dopaminergic neurons can be protected by glutathione transferase M2-2 from astrocytes, which secrete exosomes containing the protective enzyme.

9.
Neural Regen Res ; 17(9): 1861-1866, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35142659

RESUMO

Astrocytes protect neurons by modulating neuronal function and survival. Astrocytes support neurons in several ways. They provide energy through the astrocyte-neuron lactate shuttle, protect neurons from excitotoxicity, and internalize neuronal lipid droplets to degrade fatty acids for neuronal metabolic and synaptic support, as well as by their high capacity for glutamate uptake and the conversion of glutamate to glutamine. A recent reported astrocyte system for protection of dopamine neurons against the neurotoxic products of dopamine, such as aminochrome and other o-quinones, were generated under neuromelanin synthesis by oxidizing dopamine catechol structure. Astrocytes secrete glutathione transferase M2-2 through exosomes that transport this enzyme into dopaminergic neurons to protect these neurons against aminochrome neurotoxicity. The role of this new astrocyte protective mechanism in Parkinson´s disease is discussed.

10.
Vitam Horm ; 116: 313-326, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33752823

RESUMO

The female sex hormone estradiol (E2, 17ß-estradiol) has important functions in the developing brain. In addition to regulating sexual differentiation of the brain, E2 participates in the development of brain areas involved in functions unrelated to reproduction, such as cognition. E2 signals mainly thorough two estrogen receptors; estrogen receptor alpha (ERα) and beta (ERß). While ERα has distinct functions for sexual imprinting of the developing brain, ERß is considered to participate in the development of brain areas related to cognitive function. In this chapter we will focus on ERß's role during neural development. We will discuss the contributions of sex chromosomal and sex hormonal effects in this process and place it in relation to recent data on ERß obtained from stem cell models. Finally, we will discuss the lessons learned from mouse and stem cell models in understanding ERß's role in neural development and how new stem cell models, by addressing the human relevance, may help to advance our progress in this field.


Assuntos
Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Animais , Estradiol , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Feminino , Camundongos , Receptores de Estrogênio
11.
Stem Cells Transl Med ; 10(7): 987-995, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33634954

RESUMO

Hematopoietic stem cell- (HSC) and induced pluripotent stem (iPS) cell-derived natural killer (NK) cells containing engineered functions, such as chimeric antigen receptors (CAR), offer great promise for the treatment of seemingly incurable oncological malignancies. Today, some of the main challenges of CAR cell-based therapeutics are the long manufacturing time and safety of the cell sources used. Additional challenges include avoiding graft vs host disease (GVHD) and cytokine release syndrome (CRS). Here, we show compelling evidence for the use of NK cell therapeutics as a reliable off-the-shelf option, as they address key issues. Furthermore, we highlight how iPS cells and directed differentiation toward HSC and NK cells address industrial scalability and safety.


Assuntos
Células-Tronco Hematopoéticas , Imunoterapia Adotiva , Células-Tronco Pluripotentes Induzidas , Células Matadoras Naturais , Receptores de Antígenos Quiméricos , Células-Tronco Hematopoéticas/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células Matadoras Naturais/citologia
12.
Stem Cell Res ; 51: 102193, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33517122

RESUMO

KISCOi001-A is a healthy feeder-free and fully characterized human induced pluripotent stem (iPS) cell line cultured under xeno-free and defined conditions. The cell line is generated from normal human foreskin fibroblasts with non-integrating episomal plasmid vectors encoding OCT4, SOX2, KLF4, NANOG, LIN28, nontransforming L-MYC and dominant negative p53. The generated iPS cells are transgene-free and their pluripotency is confirmed by the expression of stem cell markers and capacity to differentiate into the cells of ectoderm, endoderm and mesoderm while their identity and karyotype stability is confirmed with Genomic assays.


Assuntos
Células-Tronco Pluripotentes Induzidas , Diferenciação Celular , Linhagem Celular , Fibroblastos , Prepúcio do Pênis , Humanos , Fator 4 Semelhante a Kruppel , Masculino
13.
Neurotox Res ; 39(2): 182-190, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33555546

RESUMO

The enzyme glutathione transferase M2-2, expressed in human astrocytes, increases its expression in the presence of aminochrome and catalyzes the conjugation of aminochrome, preventing its toxic effects. Secretion of the enzyme glutathione transferase M2-2 from U373MG cells, used as a cellular model for astrocytes, has been reported, and the enzyme is taken up by neuroblastoma SYSH-S7 cells and provide protection against aminochrome. The present study provides evidence that glutathione transferase M2-2 is released in exosomes from U373MG cells, thereby providing a means for intercellular transport of the enzyme. With particular relevance to Parkinson disease and other degenerative conditions, we propose a new mechanism by which astrocytes may protect dopaminergic neurons against the endogenous neurotoxin aminochrome.


Assuntos
Astrócitos/metabolismo , Exossomos/metabolismo , Glutationa Transferase/metabolismo , Transporte Proteico , Linhagem Celular Tumoral , Neurônios Dopaminérgicos/metabolismo , Humanos
14.
Brain Sci ; 7(3)2017 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-28241485

RESUMO

Many neurological disorders affect men and women differently regarding prevalence, progression, and severity. It is clear that many of these disorders may originate from defective signaling during fetal or perinatal brain development, which may affect males and females differently. Such sex-specific differences may originate from chromosomal or sex-hormone specific effects. This short review will focus on the estrogen receptor beta (ERß) signaling during perinatal brain development and put it in the context of sex-specific differences in neurodevelopmental disorders. We will discuss ERß's recent discovery in directing DNA de-methylation to specific sites, of which one such site may bear consequences for the susceptibility to the neurological reading disorder dyslexia. We will also discuss how dysregulations in sex-hormone signaling, like those evoked by endocrine disruptive chemicals, may affect this and other neurodevelopmental disorders in a sex-specific manner through ERß.

15.
Clujul Med ; 90(4): 420-424, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29151792

RESUMO

BACKGROUND AND AIMS: Selective serotonin reuptake inhibitors (SSRIs) are antidepressants increasingly prescribed against depression during and after pregnancy. However, these compounds cross the placenta and are found in breast milk, thus reaching, and possibly affecting, the fetus and infant during critical developmental stages. Fluoxetine (FLX), a widely used SSRI, can interfere with estrogen signaling, which is important for the development of female sex organs and certain brain areas, among others. Interference with estrogen signaling can take place on different levels, e.g., by affecting receptor activity or hormone levels. FLX has previously been shown to induce estrogen receptor-dependent transcription in vitro at high concentrations. In this study we set out to assess effects of FLX on estradiol levels in vitro. METHODS: FLX was tested using the OECD recommended H295R model, a human adrenocortical carcinoma cell line that is able to produce all steroid hormones found in the gonads and adrenal glands, including estradiol and testosterone. H295R cells were incubated with different doses of FLX for 48h. Subsequently, concentrations of these two steroids were measured in cell culture medium after FLX exposure, using liquid chromatography coupled with tandem mass spectrometry. Aromatase mRNA expression was assessed using qPCR. RESULTS: Fluoxetine significantly increased estradiol secretion in H295R cells after a 48h exposure at low, submicromolar concentrations, but showed no effects on testosterone levels or aromatase mRNA expression. CONCLUSION: Fluoxetine has the potential to interfere with estrogenic signaling by increasing estradiol secretion at low concentrations, which are relevant for fetal and adult human exposure.

16.
Artigo em Inglês | MEDLINE | ID: mdl-26889208

RESUMO

BACKGROUND: DNA methylation is one way to encode epigenetic information and plays a crucial role in regulating gene expression during embryonic development. DNA methylation marks are established by the DNA methyltransferases and, recently, a mechanism for active DNA demethylation has emerged involving the ten-eleven translocator proteins and thymine DNA glycosylase (TDG). However, so far it is not clear how these enzymes are recruited to, and regulate DNA methylation at, specific genomic loci. A number of studies imply that sequence-specific transcription factors are involved in targeting DNA methylation and demethylation processes. Oestrogen receptor beta (ERß) is a ligand-inducible transcription factor regulating gene expression in response to the female sex hormone oestrogen. Previously, we found that ERß deficiency results in changes in DNA methylation patterns at two gene promoters, implicating an involvement of ERß in DNA methylation. In this study, we set out to explore this involvement on a genome-wide level, and to investigate the underlying mechanisms of this function. RESULTS: Using reduced representation bisulfite sequencing, we compared genome-wide DNA methylation in mouse embryonic fibroblasts derived from wildtype and ERß knock-out mice, and identified around 8000 differentially methylated positions (DMPs). Validation and further characterisation of selected DMPs showed that differences in methylation correlated with changes in expression of the nearest gene. Additionally, re-introduction of ERß into the knock-out cells could reverse hypermethylation and reactivate expression of some of the genes. We also show that ERß is recruited to regions around hypermethylated DMPs. Finally, we demonstrate here that ERß interacts with TDG and that TDG binds ERß-dependently to hypermethylated DMPs. CONCLUSION: We provide evidence that ERß plays a role in regulating DNA methylation at specific genomic loci, likely as the result of its interaction with TDG at these regions. Our findings imply a novel function of ERß, beyond direct transcriptional control, in regulating DNA methylation at target genes. Further, they shed light on the question how DNA methylation is regulated at specific genomic loci by supporting a concept in which sequence-specific transcription factors can target factors that regulate DNA methylation patterns.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA