Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2728: 195-222, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38019403

RESUMO

The human placenta provides the site of exchange between the maternal and fetal bloodstreams, acts as an endocrine organ, and has immunological functions. The majority of pregnancy disorders including preeclampsia and fetal growth restriction have their roots in pathological placentation. Yet, the underlying molecular causes of these complications remain largely unknown, not least due to the lack of reliable in vitro models. Recent establishment of 2D human trophoblast stem cells and 3D trophoblast organoids has been a major advancement that opened new avenues for trophoblast research. Here we provide a protocol detailing isolation of cytotrophoblast from the first trimester human placenta, establishment of trophoblast organoids, their culture and differentiation conditions. Overall, we describe an in vitro system that offers an excellent model to study the molecular basis of placental development and disease.


Assuntos
Placenta , Placentação , Gravidez , Humanos , Feminino , Trofoblastos , Diferenciação Celular , Organoides
2.
J Integr Bioinform ; 20(4)2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-38127662

RESUMO

During early pregnancy, extravillous trophoblasts (EVTs) play a crucial role in modifying the maternal uterine environment. Failures in EVT lineage formation and differentiation can lead to pregnancy complications such as preeclampsia, fetal growth restriction, and pregnancy loss. Despite recent advances, our knowledge on molecular and external factors that control and affect EVT development remains incomplete. Using trophoblast organoid in vitro models, we recently discovered that coordinated manipulation of the transforming growth factor beta (TGFß) signaling is essential for EVT development. To further investigate gene networks involved in EVT function and development, we performed weighted gene co-expression network analysis (WGCNA) on our RNA-Seq data. We identified 10 modules with a median module membership of over 0.8 and sizes ranging from 1005 (M1) to 72 (M27) network genes associated with TGFß activation status or in vitro culturing, the latter being indicative for yet undiscovered factors that shape the EVT phenotypes. Lastly, we hypothesized that certain therapeutic drugs might unintentionally interfere with placentation by affecting EVT-specific gene expression. We used the STRING database to map correlations and the Drug-Gene Interaction database to identify drug targets. Our comprehensive dataset of drug-gene interactions provides insights into potential risks associated with certain drugs in early gestation.


Assuntos
Redes Reguladoras de Genes , Placenta , Gravidez , Humanos , Feminino , Placenta/metabolismo , Trofoblastos/metabolismo , Placentação/genética , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
3.
Nat Commun ; 14(1): 2559, 2023 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-37137875

RESUMO

Placental development relies on coordinated cell fate decisions governed by signalling inputs. However, little is known about how signalling cues are transformed into repressive mechanisms triggering lineage-specific transcriptional signatures. Here, we demonstrate that upon inhibition of the Fgf/Erk pathway in mouse trophoblast stem cells (TSCs), the Ets2 repressor factor (Erf) interacts with the Nuclear Receptor Co-Repressor Complex 1 and 2 (NCoR1/2) and recruits it to key trophoblast genes. Genetic ablation of Erf or Tbl1x (a component of the NCoR1/2 complex) abrogates the Erf/NCoR1/2 interaction. This leads to mis-expression of Erf/NCoR1/2 target genes, resulting in a TSC differentiation defect. Mechanistically, Erf regulates expression of these genes by recruiting the NCoR1/2 complex and decommissioning their H3K27ac-dependent enhancers. Our findings uncover how the Fgf/Erf/NCoR1/2 repressive axis governs cell fate and placental development, providing a paradigm for Fgf-mediated transcriptional control.


Assuntos
Fator 2 de Crescimento de Fibroblastos , Trofoblastos , Camundongos , Animais , Feminino , Gravidez , Placenta , Diferenciação Celular/fisiologia , Regulação da Expressão Gênica , Correpressor 1 de Receptor Nuclear , Correpressor 2 de Receptor Nuclear
5.
Cell Rep ; 42(1): 111977, 2023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36640334

RESUMO

During human pregnancy, placenta-derived extravillous trophoblasts (EVTs) invade the decidua and communicate with maternal immune cells. The decidua distinguishes into basalis (decB) and parietalis (decP). The latter remains unaffected by EVT invasion. By defining a specific gating strategy, we report the accumulation of macrophages in decB. We describe a decidua basalis-associated macrophage (decBAM) population with a differential transcriptome and secretome compared with decidua parietalis-associated macrophages (decPAMs). decBAMs are CD11chi and efficient inducers of Tregs, proliferate in situ, and secrete high levels of CXCL1, CXCL5, M-CSF, and IL-10. In contrast, decPAMs exert a dendritic cell-like, motile phenotype characterized by induced expression of HLA class II molecules, enhanced phagocytosis, and the ability to activate T cells. Strikingly, EVT-conditioned media convert decPAMs into a decBAM phenotype. These findings assign distinct macrophage phenotypes to decidual areas depending on placentation and further highlight a critical role for EVTs in the induction of decB-associated macrophage polarization.


Assuntos
Decídua , Trofoblastos , Gravidez , Feminino , Humanos , Primeiro Trimestre da Gravidez/fisiologia , Decídua/metabolismo , Trofoblastos/metabolismo , Fenótipo , Macrófagos/metabolismo
6.
Proc Natl Acad Sci U S A ; 119(28): e2120667119, 2022 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-35867736

RESUMO

Abnormal placentation has been noticed in a variety of pregnancy complications such as miscarriage, early-onset preeclampsia, and fetal growth restriction. Defects in the developmental program of extravillous trophoblasts (EVTs), migrating from placental anchoring villi into the maternal decidua and its vessels, is thought to be an underlying cause. Yet, key regulatory mechanisms controlling commitment and differentiation of the invasive trophoblast lineage remain largely elusive. Herein, comparative gene expression analyses of HLA-G-purified EVTs, isolated from donor-matched placenta, decidua, and trophoblast organoids (TB-ORGs), revealed biological processes and signaling pathways governing EVT development. In particular, bioinformatics analyses and manipulations in different versatile trophoblast cell models unraveled transforming growth factor-ß (TGF-ß) signaling as a crucial pathway driving differentiation of placental EVTs into decidual EVTs, the latter showing enrichment of a secretory gene signature. Removal of Wingless signaling and subsequent activation of the TGF-ß pathway were required for the formation of human leukocyte antigen-G+ (HLA-G+) EVTs in TB-ORGs that resemble in situ EVTs at the level of global gene expression. Accordingly, TGF-ß-treated EVTs secreted enzymes, such as DAO and PAPPA2, which were predominantly expressed by decidual EVTs. Their genes were controlled by EVT-specific induction and genomic binding of the TGF-ß downstream effector SMAD3. In summary, TGF-ß signaling plays a key role in human placental development governing the differentiation program of EVTs.


Assuntos
Placentação , Fator de Crescimento Transformador beta , Trofoblastos , Feminino , Antígenos HLA-G/metabolismo , Humanos , Gravidez , Fator de Crescimento Transformador beta/metabolismo , Trofoblastos/citologia , Trofoblastos/metabolismo
7.
Cell Mol Life Sci ; 79(6): 337, 2022 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-35657505

RESUMO

The placenta sustains embryonic development and is critical for a successful pregnancy outcome. It provides the site of exchange between the mother and the embryo, has immunological functions and is a vital endocrine organ. To perform these diverse roles, the placenta comprises highly specialized trophoblast cell types, including syncytiotrophoblast and extravillous trophoblast. The coordinated actions of transcription factors (TFs) regulate their emergence during development, subsequent specialization, and identity. These TFs integrate diverse signaling cues, form TF networks, associate with chromatin remodeling and modifying factors, and collectively determine the cell type-specific characteristics. Here, we summarize the general properties of TFs, provide an overview of TFs involved in the development and function of the human trophoblast, and address similarities and differences to their murine orthologs. In addition, we discuss how the recent establishment of human in vitro models combined with -omics approaches propel our knowledge and transform the human trophoblast field.


Assuntos
Fatores de Transcrição , Trofoblastos , Animais , Diferenciação Celular/fisiologia , Feminino , Regulação da Expressão Gênica , Humanos , Camundongos , Placenta/metabolismo , Gravidez , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Trofoblastos/metabolismo
8.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34507999

RESUMO

Multiple placental pathologies are associated with failures in trophoblast differentiation, yet the underlying transcriptional regulation is poorly understood. Here, we discovered msh homeobox 2 (MSX2) as a key transcriptional regulator of trophoblast identity using the human trophoblast stem cell model. Depletion of MSX2 resulted in activation of the syncytiotrophoblast transcriptional program, while forced expression of MSX2 blocked it. We demonstrated that a large proportion of the affected genes were directly bound and regulated by MSX2 and identified components of the SWItch/Sucrose nonfermentable (SWI/SNF) complex as strong MSX2 interactors and target gene cobinders. MSX2 cooperated specifically with the SWI/SNF canonical BAF (cBAF) subcomplex and cooccupied, together with H3K27ac, a number of differentiation genes. Increased H3K27ac and cBAF occupancy upon MSX2 depletion imply that MSX2 prevents premature syncytiotrophoblast differentiation. Our findings established MSX2 as a repressor of the syncytiotrophoblast lineage and demonstrated its pivotal role in cell fate decisions that govern human placental development and disease.


Assuntos
Diferenciação Celular , Proteínas Cromossômicas não Histona/metabolismo , Histonas/metabolismo , Proteínas de Homeodomínio/metabolismo , Placenta/citologia , Células-Tronco Pluripotentes/citologia , Fatores de Transcrição/metabolismo , Trofoblastos/citologia , Proteínas Cromossômicas não Histona/genética , Feminino , Histonas/genética , Proteínas de Homeodomínio/genética , Humanos , Placenta/metabolismo , Placentação , Células-Tronco Pluripotentes/metabolismo , Gravidez , Fatores de Transcrição/genética , Trofoblastos/metabolismo
9.
Endocrinology ; 160(10): 2282-2297, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31290979

RESUMO

The human endometrium is the inner lining of the uterus consisting of stromal and epithelial (secretory and ciliated) cells. It undergoes a hormonally regulated monthly cycle of growth, differentiation, and desquamation. However, how these cyclic changes control the balance between secretory and ciliated cells remains unclear. Here, we established endometrial organoids to investigate the estrogen (E2)-driven control of cell fate decisions in human endometrial epithelium. We demonstrate that they preserve the structure, expression patterns, secretory properties, and E2 responsiveness of their tissue of origin. Next, we show that the induction of ciliated cells is orchestrated by the coordinated action of E2 and NOTCH signaling. Although E2 is the primary driver, inhibition of NOTCH signaling provides a permissive environment. However, inhibition of NOTCH alone is not sufficient to trigger ciliogenesis. Overall, we provide insights into endometrial biology and propose endometrial organoids as a robust and powerful model for studying ciliogenesis in vitro.


Assuntos
Cílios/fisiologia , Endométrio/fisiologia , Estrogênios/metabolismo , Organoides/metabolismo , Feminino , Regulação da Expressão Gênica/fisiologia , Humanos , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais , Técnicas de Cultura de Tecidos
10.
Stem Cell Reports ; 11(2): 537-551, 2018 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-30078556

RESUMO

Defective placentation is the underlying cause of various pregnancy complications, such as severe intrauterine growth restriction and preeclampsia. However, studies on human placental development are hampered by the lack of a self-renewing in vitro model that would recapitulate formation of trophoblast progenitors and differentiated subtypes, syncytiotrophoblast (STB) and invasive extravillous trophoblast (EVT), in a 3D orientation. Hence, we established long-term expanding organoid cultures from purified first-trimester cytotrophoblasts (CTBs). Molecular analyses revealed that the CTB organoid cultures (CTB-ORGs) express markers of trophoblast stemness and proliferation and are highly similar to primary CTBs at the level of global gene expression. Whereas CTB-ORGs spontaneously generated STBs, withdrawal of factors for self-renewal induced trophoblast outgrowth, expressing the EVT progenitor marker NOTCH1, and provoked formation of adjacent, distally located HLA-G+ EVTs. In summary, we established human CTB-ORGs that grow and differentiate under defined culture conditions, allowing future human placental disease modeling.


Assuntos
Diferenciação Celular , Autorrenovação Celular , Organoides/citologia , Placenta/citologia , Trofoblastos/citologia , Biomarcadores , Proliferação de Células , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Gravidez , Trofoblastos/metabolismo , Via de Sinalização Wnt
11.
Development ; 143(20): 3650-3660, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27802134

RESUMO

Trophoblast stem cells (TSCs) retain the capacity to self-renew indefinitely and harbour the potential to differentiate into all trophoblast subtypes of the placenta. Recent studies have shown how signalling cascades integrate with transcription factor circuits to govern the fine balance between TSC self-renewal and differentiation. In addition, breakthroughs in reprogramming strategies have enabled the generation of TSCs from fibroblasts, opening up exciting new avenues that may allow the isolation of this stem cell type from other species, notably humans. Here, we review these recent advances in light of their importance for understanding placental pathologies and developing personalised medicine approaches for pregnancy complications.


Assuntos
Placenta/citologia , Células-Tronco/citologia , Trofoblastos/citologia , Animais , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Placenta/metabolismo , Gravidez , Transdução de Sinais/fisiologia , Células-Tronco/metabolismo , Trofoblastos/metabolismo
12.
Genes Dev ; 29(23): 2435-48, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26584622

RESUMO

Elf5 is a transcription factor with pivotal roles in the trophoblast compartment, where it reinforces a trophoblast stem cell (TSC)-specific transcriptional circuit. However, Elf5 is also present in differentiating trophoblast cells that have ceased to express other TSC genes such as Cdx2 and Eomes. In the present study, we aimed to elucidate the context-dependent role of Elf5 at the interface between TSC self-renewal and the onset of differentiation. We demonstrate that precise levels of Elf5 are critical for normal expansion of the TSC compartment and embryonic survival, as Elf5 overexpression triggers precocious trophoblast differentiation. Through integration of protein interactome, transcriptome, and genome-wide chromatin immunoprecipitation data, we reveal that this abundance-dependent function is mediated through a shift in preferred Elf5-binding partners; in TSCs, Elf5 interaction with Eomes recruits Tfap2c to triply occupied sites at TSC-specific genes, driving their expression. In contrast, the Elf5 and Tfap2c interaction becomes predominant as their protein levels increase. This triggers binding to double- and single-occupancy sites that harbor the cognate Tfap2c motif, causing activation of the associated differentiation-promoting genes. These data place Elf5 at the center of a stoichiometry-sensitive transcriptional network, where it acts as a molecular switch governing the balance between TSC proliferation and differentiation.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Redes Reguladoras de Genes , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Trofoblastos/citologia , Animais , Diferenciação Celular/genética , Linhagem Celular , Autorrenovação Celular/genética , Proteínas de Ligação a DNA/química , Regulação da Expressão Gênica no Desenvolvimento/genética , Camundongos , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Fatores de Transcrição/química , Trofoblastos/metabolismo
13.
Nat Commun ; 6: 7776, 2015 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-26206133

RESUMO

Esrrb (oestrogen-related receptor beta) is a transcription factor implicated in embryonic stem (ES) cell self-renewal, yet its knockout causes intrauterine lethality due to defects in trophoblast development. Here we show that in trophoblast stem (TS) cells, Esrrb is a downstream target of fibroblast growth factor (Fgf) signalling and is critical to drive TS cell self-renewal. In contrast to its occupancy of pluripotency-associated loci in ES cells, Esrrb sustains the stemness of TS cells by direct binding and regulation of TS cell-specific transcription factors including Elf5 and Eomes. To elucidate the mechanisms whereby Esrrb controls the expression of its targets, we characterized its TS cell-specific interactome using mass spectrometry. Unlike in ES cells, Esrrb interacts in TS cells with the histone demethylase Lsd1 and with the RNA Polymerase II-associated Integrator complex. Our findings provide new insights into both the general and context-dependent wiring of transcription factor networks in stem cells by master transcription factors.


Assuntos
Células-Tronco Embrionárias/fisiologia , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Receptores de Estrogênio/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Epigênese Genética , Redes Reguladoras de Genes , Histona Desmetilases/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Mapeamento de Interação de Proteínas , RNA Polimerase II/metabolismo , Trofoblastos/citologia
14.
Nucleic Acids Res ; 42(14): 8914-27, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25034692

RESUMO

Embryonic stem (ES) cells are in a dynamic equilibrium of distinct functional states, characterized by the heterogeneous expression of critical pluripotency factors and regulated by a spectrum of reversible histone modifications. Maintenance of this equilibrium is a hallmark of pluripotency. Here we find that the ADP-ribosyltransferases Parp1 and Parp7 play a critical role in safeguarding this state by occupying key pluripotency genes, notably Nanog, Pou5f1, Sox2, Stella, Tet1 and Zfp42, thereby protecting them from progressive epigenetic repression. In the absence of either Parp1 or Parp7, or upon inhibition of the ADP-ribosylating activity, ES cells exhibit a decrease in ground state pluripotency as they cannot maintain the typical heterogeneity characteristic of the metastable state. As a consequence, they display a higher propensity to differentiate. These findings place Parp1 and Parp7 at the genetic-epigenetic interface of pluripotency networks, fine-tuning the transcriptional heterogeneity and thereby determining the developmental plasticity of ES cells.


Assuntos
ADP Ribose Transferases/fisiologia , Células-Tronco Embrionárias/enzimologia , Células-Tronco Pluripotentes/enzimologia , Poli(ADP-Ribose) Polimerases/fisiologia , ADP Ribose Transferases/genética , ADP Ribose Transferases/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Cromatina/química , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/citologia , Metilação , Camundongos , Mutação , Células-Tronco Pluripotentes/citologia , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Fatores de Transcrição/metabolismo , Trofoblastos/citologia , Trofoblastos/enzimologia
15.
Science ; 338(6113): 1469-72, 2012 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-23239737

RESUMO

Mammalian imprinted genes often cluster with long noncoding (lnc) RNAs. Three lncRNAs that induce parental-specific silencing show hallmarks indicating that their transcription is more important than their product. To test whether Airn transcription or product silences the Igf2r gene, we shortened the endogenous lncRNA to different lengths. The results excluded a role for spliced and unspliced Airn lncRNA products and for Airn nuclear size and location in silencing Igf2r. Instead, silencing only required Airn transcriptional overlap of the Igf2r promoter, which interferes with RNA polymerase II recruitment in the absence of repressive chromatin. Such a repressor function for lncRNA transcriptional overlap reveals a gene silencing mechanism that may be widespread in the mammalian genome, given the abundance of lncRNA transcripts.


Assuntos
Inativação Gênica , Impressão Genômica , RNA Longo não Codificante/metabolismo , Receptor IGF Tipo 2/genética , Transcrição Gênica , Processamento Alternativo , Animais , Células Cultivadas , Camundongos , Família Multigênica , Regiões Promotoras Genéticas , RNA Polimerase II/metabolismo , RNA Longo não Codificante/genética
16.
Biol Open ; 1(4): 341-52, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23213424

RESUMO

Embryonic Stem (ES) cells are able to give rise to the three germ layers of the embryo but are prevented from contributing to the trophoblast. The molecular nature of this barrier between embryonic and trophectodermal cell fates is not clear, but is known to involve DNA methylation. Here we demonstrate that the Nucleosome Remodeling and Deacetylation (NuRD) co-repressor complex maintains the developmental barrier between embryonic and trophectodermal cell fates by maintaining transcriptional silencing of trophectoderm determinant genes in ES cells. We further show that NuRD activity facilitates DNA methylation of several of its target promoters, where it acts non-redundantly with DNA methylation to enforce transcriptional silencing. NuRD-deficient ES cells fail to completely silence expression of the trophectoderm determinant genes Elf5 and Eomes, but this alone is not sufficient to induce transdifferentiation towards the trophectoderm fate. Rather this leaves ES cells capable of activating expression of trophectoderm-specific genes in response to appropriate extracellular signals, enabling them to commit to a trophectodermal cell fate. Our findings clarify the molecular nature of the developmental barrier between the embryonic and trophoblast cell fates, and establish a role for NuRD activity in specifying sites for de novo DNA methylation.

17.
Cell Stem Cell ; 10(5): 583-94, 2012 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-22560079

RESUMO

Transcriptional heterogeneity within embryonic stem cell (ESC) populations has been suggested as a mechanism by which a seemingly homogeneous cell population can initiate differentiation into an array of different cell types. Chromatin remodeling proteins have been shown to control transcriptional variability in yeast and to be important for mammalian ESC lineage commitment. Here we show that the Nucleosome Remodeling and Deacetylation (NuRD) complex, which is required for ESC lineage commitment, modulates both transcriptional heterogeneity and the dynamic range of a set of pluripotency genes in ESCs. In self-renewing conditions, the influence of NuRD at these genes is balanced by the opposing action of self-renewal factors. Upon loss of self-renewal factors, the action of NuRD is sufficient to silence transcription of these pluripotency genes, allowing cells to exit self-renewal. We propose that modulation of transcription levels by NuRD is key to maintaining the differentiation responsiveness of pluripotent cells.


Assuntos
Células-Tronco Embrionárias/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Células-Tronco Pluripotentes/fisiologia , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Células Cultivadas , Proteínas de Ligação a DNA/genética , Heterogeneidade Genética , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/genética , Camundongos , Camundongos Knockout , Fatores de Transcrição/genética
18.
PLoS Genet ; 8(3): e1002540, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22396659

RESUMO

A CpG island (CGI) lies at the 5' end of the Airn macro non-protein-coding (nc) RNA that represses the flanking Igf2r promoter in cis on paternally inherited chromosomes. In addition to being modified on maternally inherited chromosomes by a DNA methylation imprint, the Airn CGI shows two unusual organization features: its position immediately downstream of the Airn promoter and transcription start site and a series of tandem direct repeats (TDRs) occupying its second half. The physical separation of the Airn promoter from the CGI provides a model to investigate if the CGI plays distinct transcriptional and epigenetic roles. We used homologous recombination to generate embryonic stem cells carrying deletions at the endogenous locus of the entire CGI or just the TDRs. The deleted Airn alleles were analyzed by using an ES cell imprinting model that recapitulates the onset of Igf2r imprinted expression in embryonic development or by using knock-out mice. The results show that the CGI is required for efficient Airn initiation and to maintain the unmethylated state of the Airn promoter, which are both necessary for Igf2r repression on the paternal chromosome. The TDRs occupying the second half of the CGI play a minor role in Airn transcriptional elongation or processivity, but are essential for methylation on the maternal Airn promoter that is necessary for Igf2r to be expressed from this chromosome. Together the data indicate the existence of a class of regulatory CGIs in the mammalian genome that act downstream of the promoter and transcription start.


Assuntos
Ilhas de CpG/genética , Metilação de DNA , Epigênese Genética , Impressão Genômica , Regiões Promotoras Genéticas , Precursores de RNA/genética , RNA não Traduzido/genética , Animais , Diferenciação Celular , Células Cultivadas , Desenvolvimento Embrionário , Células-Tronco Embrionárias , Regulação da Expressão Gênica , Recombinação Homóloga , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/metabolismo , Camundongos , Deleção de Sequência , Sequências de Repetição em Tandem , Sítio de Iniciação de Transcrição
19.
RNA Biol ; 6(2): 100-6, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19229135

RESUMO

In mammals, imprinted genes are clustered and at least one gene in each imprinted cluster is a long i.e., macro non-coding (nc) RNA. Most genes in a cluster show concordant parental-specific expression but the ncRNA is the odd one out, and is expressed from the opposite parental chromosome. While reciprocal expression between imprinted macro non-coding RNAs and flanking mRNA genes is indicative of a functional role, only two of three tested macro ncRNAs have been shown to induce imprinted gene expression. The two known functional imprinted macro non-coding RNAs are both RNAPII transcripts with unusual transcriptional properties that may be functionally relevant and their analysis may shed light on the function of non-coding RNAs that have been shown to comprise the majority of the mammalian transcriptome.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Impressão Genômica , RNA não Traduzido/fisiologia , Animais , Embrião de Mamíferos/fisiologia , Epigênese Genética , Feminino , Família Multigênica , Placenta/fisiologia , Gravidez
20.
Development ; 136(3): 437-48, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19141673

RESUMO

Genomic imprinting is an epigenetic process that results in parental-specific gene expression. Advances in understanding the mechanism that regulates imprinted gene expression in mammals have largely depended on generating targeted manipulations in embryonic stem (ES) cells that are analysed in vivo in mice. However, genomic imprinting consists of distinct developmental steps, some of which occur in post-implantation embryos, indicating that they could be studied in vitro in ES cells. The mouse Igf2r gene shows imprinted expression only in post-implantation stages, when repression of the paternal allele has been shown to require cis-expression of the Airn non-coding (nc) RNA and to correlate with gain of DNA methylation and repressive histone modifications. Here we follow the gain of imprinted expression of Igf2r during in vitro ES cell differentiation and show that it coincides with the onset of paternal-specific expression of the Airn ncRNA. Notably, although Airn ncRNA expression leads, as predicted, to gain of repressive epigenetic marks on the paternal Igf2r promoter, we unexpectedly find that the paternal Igf2r promoter is expressed at similar low levels throughout ES cell differentiation. Our results further show that the maternal and paternal Igf2r promoters are expressed equally in undifferentiated ES cells, but during differentiation expression of the maternal Igf2r promoter increases up to 10-fold, while expression from the paternal Igf2r promoter remains constant. This indicates, contrary to expectation, that the Airn ncRNA induces imprinted Igf2r expression not by silencing the paternal Igf2r promoter, but by generating an expression bias between the two parental alleles.


Assuntos
Células-Tronco Embrionárias/citologia , Impressão Genômica , RNA não Traduzido/biossíntese , Receptor IGF Tipo 2/biossíntese , Alelos , Animais , Diferenciação Celular , Células Cultivadas , Ilhas de CpG , Metilação de DNA , Células-Tronco Embrionárias/fisiologia , Epigênese Genética , Histonas/genética , Histonas/metabolismo , Camundongos , Regiões Promotoras Genéticas , RNA não Traduzido/genética , Receptor IGF Tipo 2/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...