Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
EMBO J ; 43(8): 1388-1419, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38514807

RESUMO

Neocortex expansion during evolution is linked to higher numbers of neurons, which are thought to result from increased proliferative capacity and neurogenic potential of basal progenitor cells during development. Here, we show that EREG, encoding the growth factor EPIREGULIN, is expressed in the human developing neocortex and in gorilla cerebral organoids, but not in the mouse neocortex. Addition of EPIREGULIN to the mouse neocortex increases proliferation of basal progenitor cells, whereas EREG ablation in human cortical organoids reduces proliferation in the subventricular zone. Treatment of cortical organoids with EPIREGULIN promotes a further increase in proliferation of gorilla but not of human basal progenitor cells. EPIREGULIN competes with the epidermal growth factor (EGF) to promote proliferation, and inhibition of the EGF receptor abrogates the EPIREGULIN-mediated increase in basal progenitor cells. Finally, we identify putative cis-regulatory elements that may contribute to the observed inter-species differences in EREG expression. Our findings suggest that species-specific regulation of EPIREGULIN expression may contribute to the increased neocortex size of primates by providing a tunable pro-proliferative signal to basal progenitor cells in the subventricular zone.


Assuntos
Epirregulina , Neocórtex , Animais , Humanos , Camundongos , Proliferação de Células , Epirregulina/genética , Epirregulina/metabolismo , Gorilla gorilla/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neocórtex/citologia , Neocórtex/metabolismo , Primatas/fisiologia
2.
Mol Cell ; 58(1): 157-71, 2015 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-25801166

RESUMO

Polycomb repressive complexes PRC1 and PRC2 regulate expression of genes involved in proliferation and development. In mouse early embryos, however, canonical PRC1 localizes to paternal pericentric heterochromatin (pat-PCH), where it represses transcription of major satellite repeats. In contrast, maternal PCH (mat-PCH) is enriched for H3 lysine 9 tri-methylation (H3K9me3) and Hp1ß. How PRC1 is targeted to pat-PCH, yet excluded from mat-PCH, has remained elusive. Here, we identify a PRC1 targeting mechanism that relies on Cbx2 and Hp1ß. Cbx2 directs catalytically active PRC1 to PCH via its chromodomain (CD(Cbx2)) and neighboring AT-hook (AT(Cbx2)) binding to H3K27me3 and AT-rich major satellites, respectively. CD(Cbx2) prevents AT(Cbx2) from interacting with DNA at PCH marked by H3K9me3 and Hp1ß. Loss-of-function studies show that Hp1ß and not H3K9me3 prevents PRC1 targeting to mat-PCH. Our findings indicate that CD(Cbx2) and AT(Cbx2) separated by a short linker function together to integrate H3K9me3/HP1 and H3K27me3 states.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Heterocromatina/metabolismo , Complexo Repressor Polycomb 1/genética , Zigoto/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Centrômero , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Embrião de Mamíferos , Feminino , Heterocromatina/química , Histonas/genética , Histonas/metabolismo , Padrões de Herança , Masculino , Metiltransferases/genética , Metiltransferases/metabolismo , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Complexo Repressor Polycomb 1/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Alinhamento de Sequência , Transdução de Sinais , Zigoto/crescimento & desenvolvimento
3.
EMBO J ; 36(17): 2642-2658, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28765163

RESUMO

The generation of neocortical neurons from neural progenitor cells (NPCs) is primarily controlled by transcription factors binding to DNA in the context of chromatin. To understand the complex layer of regulation that orchestrates different NPC types from the same DNA sequence, epigenome maps with cell type resolution are required. Here, we present genomewide histone methylation maps for distinct neural cell populations in the developing mouse neocortex. Using different chromatin features, we identify potential novel regulators of cortical NPCs. Moreover, we identify extensive H3K27me3 changes between NPC subtypes coinciding with major developmental and cell biological transitions. Interestingly, we detect dynamic H3K27me3 changes on promoters of several crucial transcription factors, including the basal progenitor regulator Eomes We use catalytically inactive Cas9 fused with the histone methyltransferase Ezh2 to edit H3K27me3 at the Eomes locus in vivo, which results in reduced Tbr2 expression and lower basal progenitor abundance, underscoring the relevance of dynamic H3K27me3 changes during neocortex development. Taken together, we provide a rich resource of neocortical histone methylation data and outline an approach to investigate its contribution to the regulation of selected genes during neocortical development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Histonas/genética , Neocórtex/metabolismo , Células-Tronco Neurais/fisiologia , Animais , Epigênese Genética , Perfilação da Expressão Gênica , Genoma , Metilação , Camundongos Transgênicos , Neurogênese/fisiologia
4.
PLoS Pathog ; 11(6): e1004978, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26083387

RESUMO

Respiratory syncytial virus (RSV) infection can result in severe disease partially due to its ability to interfere with the initiation of Th1 responses targeting the production of type I interferons (IFN) and promoting a Th2 immune environment. Epigenetic modulation of gene transcription has been shown to be important in regulating inflammatory pathways. RSV-infected bone marrow-derived DCs (BMDCs) upregulated expression of Kdm5b/Jarid1b H3K4 demethylase. Kdm5b-specific siRNA inhibition in BMDC led to a 10-fold increase in IFN-ß as well as increases in IL-6 and TNF-α compared to control-transfected cells. The generation of Kdm5bfl/fl-CD11c-Cre+ mice recapitulated the latter results during in vitro DC activation showing innate cytokine modulation. In vivo, infection of Kdm5bfl/fl-CD11c-Cre+ mice with RSV resulted in higher production of IFN-γ and reduced IL-4 and IL-5 compared to littermate controls, with significantly decreased inflammation, IL-13, and mucus production in the lungs. Sensitization with RSV-infected DCs into the airways of naïve mice led to an exacerbated response when mice were challenged with live RSV infection. When Kdm5b was blocked in DCs with siRNA or DCs from Kdm5bfl/fl-CD11c-CRE mice were used, the exacerbated response was abrogated. Importantly, human monocyte-derived DCs treated with a chemical inhibitor for KDM5B resulted in increased innate cytokine levels as well as elicited decreased Th2 cytokines when co-cultured with RSV reactivated CD4+ T cells. These results suggest that KDM5B acts to repress type I IFN and other innate cytokines to promote an altered immune response following RSV infection that contributes to development of chronic disease.


Assuntos
Citocinas/biossíntese , Proteínas de Ligação a DNA/imunologia , Células Dendríticas/imunologia , Regulação da Expressão Gênica/imunologia , Histona Desmetilases com o Domínio Jumonji/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Células Cultivadas , Imunoprecipitação da Cromatina , Técnicas de Cocultura , Citocinas/imunologia , Modelos Animais de Doenças , Imunidade Inata/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA Interferente Pequeno , Reação em Cadeia da Polimerase em Tempo Real , Vírus Sinciciais Respiratórios/imunologia , Transfecção
5.
Blood ; 125(13): 2075-8, 2015 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-25655602

RESUMO

Jarid1b/KDM5b is a histone demethylase that regulates self-renewal and differentiation in stem cells and cancer; however, its function in hematopoiesis is unclear. Here, we find that Jarid1b is highly expressed in primitive hematopoietic compartments and is overexpressed in acute myeloid leukemias. Constitutive genetic deletion of Jarid1b did not impact steady-state hematopoiesis. In contrast, acute deletion of Jarid1b from bone marrow increased peripheral blood T cells and, following secondary transplantation, resulted in loss of bone marrow reconstitution. Our results reveal that deletion of Jarid1b compromises hematopoietic stem cell (HSC) self-renewal capacity and suggest that Jarid1b is a positive regulator of HSC potential.


Assuntos
Proliferação de Células/genética , Proteínas de Ligação a DNA/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Histona Desmetilases com o Domínio Jumonji/fisiologia , Animais , Diferenciação Celular/genética , Divisão Celular/genética , Proteínas de Ligação a DNA/genética , Hematopoese/genética , Histona Desmetilases com o Domínio Jumonji/genética , Masculino , Camundongos , Camundongos Knockout
6.
Arterioscler Thromb Vasc Biol ; 35(7): 1645-52, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26023081

RESUMO

OBJECTIVE: Altering endothelial biology through epigenetic modifiers is an attractive novel concept, which is, however, just in its beginnings. We therefore set out to identify chromatin modifiers important for endothelial gene expression and contributing to angiogenesis. APPROACH AND RESULTS: To identify chromatin modifying enzymes in endothelial cells, histone demethylases were screened by microarray and polymerase chain reaction. The histone 3 lysine 4 demethylase JARID1B was identified as a highly expressed enzyme at the mRNA and protein levels. Knockdown of JARID1B by shRNA in human umbilical vein endothelial cells attenuated cell migration, angiogenic sprouting, and tube formation. Similarly, pharmacological inhibition and overexpression of a catalytic inactive JARID1B mutant reduced the angiogenic capacity of human umbilical vein endothelial cells. To identify the in vivo relevance of JARID1B in the vascular system, Jarid1b knockout mice were studied. As global knockout results in increased mortality and developmental defects, tamoxifen-inducible and endothelial-specific knockout mice were generated. Acute knockout of Jarid1b attenuated retinal angiogenesis and endothelial sprout outgrowth from aortic segments. To identify the underlying mechanism, a microarray experiment was performed, which led to the identification of the antiangiogenic transcription factor HOXA5 to be suppressed by JARID1B. Importantly, downregulation or inhibition of JARID1B, but not of JARID1A and JARID1C, induced HOXA5 expression in human umbilical vein endothelial cells. Consistently, chromatin immunoprecipitation revealed that JARID1B occupies and reduces the histone 3 lysine 4 methylation levels at the HOXA5 promoter, demonstrating a direct function of JARID1B in endothelial HOXA5 gene regulation. CONCLUSIONS: JARID1B, by suppressing HOXA5, maintains the endothelial angiogenic capacity in a demethylase-dependent manner.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Epigênese Genética , Proteínas de Homeodomínio/genética , Histona Desmetilases com o Domínio Jumonji/fisiologia , Neovascularização Fisiológica/genética , Proteínas Nucleares/fisiologia , Fosfoproteínas/genética , Animais , Células Cultivadas , Células Endoteliais/fisiologia , Proteínas de Homeodomínio/fisiologia , Humanos , Camundongos Knockout , Fosfoproteínas/fisiologia , Fatores de Transcrição , Transcrição Gênica , Veias Umbilicais
7.
PLoS Genet ; 9(4): e1003461, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23637629

RESUMO

Embryonic development is tightly regulated by transcription factors and chromatin-associated proteins. H3K4me3 is associated with active transcription and H3K27me3 with gene repression, while the combination of both keeps genes required for development in a plastic state. Here we show that deletion of the H3K4me2/3 histone demethylase Jarid1b (Kdm5b/Plu1) results in major neonatal lethality due to respiratory failure. Jarid1b knockout embryos have several neural defects including disorganized cranial nerves, defects in eye development, and increased incidences of exencephaly. Moreover, in line with an overlap of Jarid1b and Polycomb target genes, Jarid1b knockout embryos display homeotic skeletal transformations typical for Polycomb mutants, supporting a functional interplay between Polycomb proteins and Jarid1b. To understand how Jarid1b regulates mouse development, we performed a genome-wide analysis of histone modifications, which demonstrated that normally inactive genes encoding developmental regulators acquire aberrant H3K4me3 during early embryogenesis in Jarid1b knockout embryos. H3K4me3 accumulates as embryonic development proceeds, leading to increased expression of neural master regulators like Pax6 and Otx2 in Jarid1b knockout brains. Taken together, these results suggest that Jarid1b regulates mouse development by protecting developmental genes from inappropriate acquisition of active histone modifications.


Assuntos
Histona Desmetilases com o Domínio Jumonji , Proteínas Repressoras , Animais , Desenvolvimento Embrionário , Genes Controladores do Desenvolvimento , Histonas/metabolismo , Histona Desmetilases com o Domínio Jumonji/genética , Camundongos , Proteínas Nucleares/genética , Proteínas do Grupo Polycomb/genética , Proteínas Repressoras/genética
8.
EMBO J ; 30(22): 4586-600, 2011 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-22020125

RESUMO

H3K4 methylation is associated with active transcription and in combination with H3K27me3 thought to keep genes regulating development in a poised state. The contribution of enzymes regulating trimethylation of lysine 4 at histone 3 (H3K4me3) levels to embryonic stem cell (ESC) self-renewal and differentiation is just starting to emerge. Here, we show that the H3K4me2/3 histone demethylase Jarid1b (Kdm5b/Plu1) is dispensable for ESC self-renewal, but essential for ESC differentiation along the neural lineage. By genome-wide location analysis, we demonstrate that Jarid1b localizes predominantly to transcription start sites of genes encoding developmental regulators, of which more than half are also bound by Polycomb group proteins. Virtually all Jarid1b target genes are associated with H3K4me3 and depletion of Jarid1b in ESCs leads to a global increase of H3K4me3 levels. During neural differentiation, Jarid1b-depleted ESCs fail to efficiently silence lineage-inappropriate genes, specifically stem and germ cell genes. Our results delineate an essential role for Jarid1b-mediated transcriptional control during ESC differentiation.


Assuntos
Células-Tronco Embrionárias/fisiologia , Histonas/metabolismo , Neurogênese , Neurônios/fisiologia , Transcrição Gênica , Animais , Anticorpos Monoclonais , Linhagem Celular , Sistema Nervoso Central/embriologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/citologia , Perfilação da Expressão Gênica , Técnicas de Inativação de Genes/métodos , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/imunologia , Histona Desmetilases com o Domínio Jumonji/metabolismo , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/citologia , Proteínas do Grupo Polycomb , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno , Proteínas Repressoras/metabolismo
10.
Nat Cell Biol ; 25(12): 1873-1883, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37996647

RESUMO

Gene expression is regulated by multiple epigenetic mechanisms, which are coordinated in development and disease. However, current multiomics methods are frequently limited to one or two modalities at a time, making it challenging to obtain a comprehensive gene regulatory signature. Here, we describe a method-3D genome, RNA, accessibility and methylation sequencing (3DRAM-seq)-that simultaneously interrogates spatial genome organization, chromatin accessibility and DNA methylation genome-wide and at high resolution. We combine 3DRAM-seq with immunoFACS and RNA sequencing in cortical organoids to map the cell-type-specific regulatory landscape of human neural development across multiple epigenetic layers. Finally, we apply a massively parallel reporter assay to profile cell-type-specific enhancer activity in organoids and to functionally assess the role of key transcription factors for human enhancer activation and function. More broadly, 3DRAM-seq can be used to profile the multimodal epigenetic landscape in rare cell types and different tissues.


Assuntos
Cromatina , Epigenoma , Humanos , Cromatina/genética , Cromatina/metabolismo , Epigênese Genética , Metilação de DNA/genética , Organoides/metabolismo
11.
Semin Cell Dev Biol ; 21(2): 209-20, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19892027

RESUMO

Cancer is perceived as a heterogeneous group of diseases that is characterized by aberrant patterns of gene expression. In the last decade, an increasing amount of data has pointed to a key role for epigenetic alterations in human cancer. In this review, we focus on a subclass of epigenetic regulators, namely histone methyltransferases (HMTs). Several HMTs have been linked to different types of cancer; however, in most cases we only have limited knowledge regarding the molecular mechanisms by which the HMTs contribute to disease development. We summarize the current knowledge regarding some of the best-validated examples of HMTs contributing to tumorigenesis and discuss their potential mechanisms of action.


Assuntos
Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Neoplasias/enzimologia , Neoplasias/genética , Epigênese Genética , Histona Metiltransferases , Humanos
12.
Dev Neurobiol ; 82(4): 345-363, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35384339

RESUMO

The neocortex is considered the seat of higher cognitive function in humans. It develops from a sheet of neural progenitor cells, most of which eventually give rise to neurons. This process of cell fate determination is controlled by precise temporal and spatial gene expression patterns that in turn are affected by epigenetic mechanisms including Polycomb group (PcG) regulation. PcG proteins assemble in multiprotein complexes and catalyze repressive posttranslational histone modifications. Their association with neurodevelopmental disease and various types of cancer of the central nervous system, as well as observations in mouse models, has implicated these epigenetic modifiers in controlling various stages of cortex development. The precise mechanisms conveying PcG-associated transcriptional repression remain incompletely understood and are an active field of research. PcG activity appears to be highly context-specific, raising the question of species-specific differences in the regulation of neural stem and progenitor regulation. In this review, we will discuss our growing understanding of how PcG regulation affects human cortex development, based on studies in murine model systems, but focusing mostly on findings obtained from examining impaired PcG activity in the context of human neurodevelopmental disorders and cancer. Furthermore, we will highlight relevant experimental approaches for functional investigations of PcG regulation in human cortex development.


Assuntos
Proteínas de Drosophila , Neoplasias , Transtornos do Neurodesenvolvimento , Animais , Encéfalo/metabolismo , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica , Humanos , Camundongos , Transtornos do Neurodesenvolvimento/genética , Proteínas do Grupo Polycomb/genética , Proteínas do Grupo Polycomb/metabolismo
13.
Sci Adv ; 7(38): eabc6792, 2021 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-34524839

RESUMO

Increase in the size of human neocortex­acquired in evolution­accounts for the unique cognitive capacity of humans. This expansion reflects the evolutionarily enhanced proliferative ability of basal progenitors (BPs), including the basal radial glia and basal intermediate progenitors (bIPs) in mammalian cortex, which may have been acquired through epigenetic alterations in BPs. However, how the epigenome in BPs differs across species is not known. Here, we report that histone H3 acetylation is a key epigenetic regulation in bIP amplification and cortical expansion. Through epigenetic profiling of sorted bIPs, we show that histone H3 lysine 9 acetylation (H3K9ac) is low in murine bIPs and high in human bIPs. Elevated H3K9ac preferentially increases bIP proliferation, increasing the size and folding of the normally smooth mouse neocortex. H3K9ac drives bIP amplification by increasing expression of the evolutionarily regulated gene, Trnp1, in developing cortex. Our findings demonstrate a previously unknown mechanism that controls cortical architecture.

14.
Front Neurosci ; 12: 359, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29896084

RESUMO

Neurogenesis is the process through which neural stem and progenitor cells generate neurons. During the development of the mouse neocortex, stem and progenitor cells sequentially give rise to neurons destined to different cortical layers and then switch to gliogenesis resulting in the generation of astrocytes and oligodendrocytes. Precise spatial and temporal regulation of neural progenitor differentiation is key for the proper formation of the complex structure of the neocortex. Dynamic changes in gene expression underlie the coordinated differentiation program, which enables the cells to generate the RNAs and proteins required at different stages of neurogenesis and across different cell types. Here, we review the contribution of epigenetic mechanisms, with a focus on Polycomb proteins, to the regulation of gene expression programs during mouse neocortical development. Moreover, we discuss the recent emerging concept of epigenetic and transcriptional pre-patterning in neocortical progenitor cells as well as post-transcriptional mechanisms for the fine-tuning of mRNA abundance.

15.
Elife ; 72018 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-30484771

RESUMO

The evolutionary increase in size and complexity of the primate neocortex is thought to underlie the higher cognitive abilities of humans. ARHGAP11B is a human-specific gene that, based on its expression pattern in fetal human neocortex and progenitor effects in embryonic mouse neocortex, has been proposed to have a key function in the evolutionary expansion of the neocortex. Here, we study the effects of ARHGAP11B expression in the developing neocortex of the gyrencephalic ferret. In contrast to its effects in mouse, ARHGAP11B markedly increases proliferative basal radial glia, a progenitor cell type thought to be instrumental for neocortical expansion, and results in extension of the neurogenic period and an increase in upper-layer neurons. Consequently, the postnatal ferret neocortex exhibits increased neuron density in the upper cortical layers and expands in both the radial and tangential dimensions. Thus, human-specific ARHGAP11B can elicit hallmarks of neocortical expansion in the developing ferret neocortex.


Assuntos
Células Ependimogliais/metabolismo , Proteínas Ativadoras de GTPase/genética , Neocórtex/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese/genética , Animais , Eletroporação , Embrião de Mamíferos , Células Ependimogliais/citologia , Feminino , Furões , Proteínas Ativadoras de GTPase/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Neocórtex/anatomia & histologia , Neocórtex/citologia , Neocórtex/crescimento & desenvolvimento , Células-Tronco Neurais/citologia , Neurônios/citologia , Neurônios/metabolismo , Tamanho do Órgão , Plasmídeos/administração & dosagem , Plasmídeos/química , Plasmídeos/metabolismo , Transgenes , Útero , Proteína Vermelha Fluorescente
16.
Elife ; 72018 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-29561261

RESUMO

Understanding the molecular basis that underlies the expansion of the neocortex during primate, and notably human, evolution requires the identification of genes that are particularly active in the neural stem and progenitor cells of the developing neocortex. Here, we have used existing transcriptome datasets to carry out a comprehensive screen for protein-coding genes preferentially expressed in progenitors of fetal human neocortex. We show that 15 human-specific genes exhibit such expression, and many of them evolved distinct neural progenitor cell-type expression profiles and levels compared to their ancestral paralogs. Functional studies on one such gene, NOTCH2NL, demonstrate its ability to promote basal progenitor proliferation in mice. An additional 35 human genes with progenitor-enriched expression are shown to have orthologs only in primates. Our study provides a resource of genes that are promising candidates to exert specific, and novel, roles in neocortical development during primate, and notably human, evolution.


Assuntos
Células-Tronco Embrionárias/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Neocórtex/metabolismo , Células-Tronco Neurais/metabolismo , Animais , Proliferação de Células/genética , Células-Tronco Embrionárias/citologia , Evolução Molecular , Humanos , Neocórtex/citologia , Neocórtex/embriologia , Células-Tronco Neurais/citologia , Neurogênese/genética , Neurônios/citologia , Neurônios/metabolismo , Primatas/classificação , Primatas/genética , Receptor Notch2/genética , Especificidade da Espécie
17.
Science ; 347(6229): 1465-70, 2015 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-25721503

RESUMO

Evolutionary expansion of the human neocortex reflects increased amplification of basal progenitors in the subventricular zone, producing more neurons during fetal corticogenesis. In this work, we analyze the transcriptomes of distinct progenitor subpopulations isolated by a cell polarity-based approach from developing mouse and human neocortex. We identify 56 genes preferentially expressed in human apical and basal radial glia that lack mouse orthologs. Among these, ARHGAP11B has the highest degree of radial glia-specific expression. ARHGAP11B arose from partial duplication of ARHGAP11A (which encodes a Rho guanosine triphosphatase-activating protein) on the human lineage after separation from the chimpanzee lineage. Expression of ARHGAP11B in embryonic mouse neocortex promotes basal progenitor generation and self-renewal and can increase cortical plate area and induce gyrification. Hence, ARHGAP11B may have contributed to evolutionary expansion of human neocortex.


Assuntos
Proteínas Ativadoras de GTPase/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Neocórtex/embriologia , Células-Tronco Neurais/citologia , Neurogênese/genética , Animais , Separação Celular , Proteínas Ativadoras de GTPase/química , Proteínas Ativadoras de GTPase/genética , Duplicação Gênica , Humanos , Ventrículos Laterais/citologia , Camundongos , Neocórtex/citologia , Neocórtex/metabolismo , Células-Tronco Neurais/metabolismo , Neuroglia/citologia , Neuroglia/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Estrutura Terciária de Proteína , Transcriptoma
18.
Nat Commun ; 5: 5868, 2014 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-25519718

RESUMO

The different configurations of maternal and paternal chromatin, acquired during oogenesis and spermatogenesis, have to be rearranged after fertilization to form a functional embryonic genome. In the paternal genome, nucleosomal chromatin domains are re-established after the protamine-to-histone exchange. We investigated the formation of constitutive heterochromatin (cHC) in human preimplantation embryos. Our results show that histones carrying canonical cHC modifications are retained in cHC regions of sperm chromatin. These modified histones are transmitted to the oocyte and contribute to the formation of paternal embryonic cHC. Subsequently, the modifications are recognized by the H3K9/HP1 pathway maternal chromatin modifiers and propagated over the embryonic cleavage divisions. These results are in contrast to what has been described for mouse embryos, in which paternal cHC lacks canonical modifications and is initially established by Polycomb group proteins. Our results show intergenerational epigenetic inheritance of the cHC structure in human embryos.


Assuntos
Proteínas Cromossômicas não Histona/genética , Heterocromatina/química , Histonas/genética , Padrões de Herança , Espermatozoides/metabolismo , Animais , Blastocisto/química , Blastocisto/citologia , Blastocisto/metabolismo , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/metabolismo , Embrião de Mamíferos , Feminino , Fertilização , Fertilização in vitro , Regulação da Expressão Gênica no Desenvolvimento , Heterocromatina/metabolismo , Histonas/metabolismo , Humanos , Masculino , Camundongos , Oócitos/metabolismo , Oócitos/ultraestrutura , Oogênese/genética , Proteínas do Grupo Polycomb/genética , Proteínas do Grupo Polycomb/metabolismo , Especificidade da Espécie , Espermatogênese/genética , Espermatozoides/ultraestrutura , Zigoto/química , Zigoto/citologia , Zigoto/metabolismo
19.
Nat Commun ; 5: 3649, 2014 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-24728135

RESUMO

The ability of PRC1 and PRC2 to promote proliferation is a main feature that links polycomb (PcG) activity to cancer. PcGs silence the expression of the tumour suppressor locus Ink4a/Arf, whose products positively regulate pRb and p53 functions. Enhanced PcG activity is a frequent feature of human tumours, and PcG inhibition has been proposed as a strategy for cancer treatment. However, the recurrent inactivation of pRb/p53 responses in human cancers raises a question regarding the ability of PcG proteins to affect cellular proliferation independently from this checkpoint. Here we demonstrate that PRCs regulate cellular proliferation and transformation independently of the Ink4a/Arf-pRb-p53 pathway. We provide evidence that PRCs localize at replication forks, and that loss of their function directly affects the progression and symmetry of DNA replication forks. Thus, we have identified a novel activity by which PcGs can regulate cell proliferation independently of major cell cycle restriction checkpoints.


Assuntos
Pontos de Checagem do Ciclo Celular/fisiologia , Replicação do DNA/fisiologia , Proteínas do Grupo Polycomb/metabolismo , Animais , Pontos de Checagem do Ciclo Celular/genética , Células Cultivadas , Ensaio Cometa , Feminino , Immunoblotting , Camundongos , Camundongos Nus , Proteínas do Grupo Polycomb/genética
20.
Curr Opin Genet Dev ; 19(2): 113-21, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19359161

RESUMO

A decade after cloning the sheep Dolly, the induction of pluripotency by transcription factors has further revolutionized the possibilities of reprogramming a cell's identity, with exciting prospects for personalized medicine. Establishing totipotency during natural reproduction remains, however, exceedingly more efficient than in reproductive cloning or in transcription factor-based reprogramming. Understanding the molecular mechanisms directing acquisition of totipotency during early embryogenesis may enable optimization of protocols for induced reprogramming. Recent studies in mouse embryonic stem cells (ESCs) show that self-renewal and pluripotency are efficiently maintained by a core set of transcription factors when intrinsic differentiation inducing signals are blocked. In early embryos, the specification of the pluripotent epiblast and two differentiating lineages (trophectoderm and primitive endoderm) is controlled by transcription factors that are regulated by autoactivating and reciprocal repressive mechanisms as well as by ERK-mediated signaling. Chromatin-based regulatory mechanisms also contribute to the identity of ESCs and early embryos. During gametogenesis, genomes undergo extensive epigenetic reprogramming. This may underlie the efficient acquisition of totipotency during subsequent preimplantation development.


Assuntos
Desenvolvimento Embrionário/genética , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Animais , Reprogramação Celular , Cromatina/metabolismo , Metilação de DNA , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA