Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Biogerontology ; 25(2): 341-360, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37987889

RESUMO

Telomere shortening is a well-established hallmark of cellular aging. Telomerase reverse transcriptase (TERT) plays a crucial role in maintaining the length of telomeres, which are specialised protective caps at the end of chromosomes. The lack of in vitro aging models, particularly for the central nervous system (CNS), has impeded progress in understanding aging and age-associated neurodegenerative diseases. In this study, we aimed to explore the possibility of inducing aging-associated features in cell types of the CNS using hiPSC (human induced pluripotent stem cell) technology. To achieve this, we utilised CRISPR/Cas9 to generate hiPSCs with a loss of telomerase function and shortened telomeres. Through directed differentiation, we generated motor neurons and astrocytes to investigate whether telomere shortening could lead to age-associated phenotypes. Our findings revealed that shortened telomeres induced age-associated characteristics in both motor neurons and astrocytes including increased cellular senescence, heightened inflammation, and elevated DNA damage. We also observed cell-type specific age-related morphology changes. Additionally, our study highlighted the fundamental role of TERT and telomere shortening in neural progenitor cell (NPC) proliferation and neuronal differentiation. This study serves as a proof of concept that telomere shortening can effectively induce aging-associated phenotypes, thereby providing a valuable tool to investigate age-related decline and neurodegenerative diseases.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doenças Neurodegenerativas , Telomerase , Humanos , Encurtamento do Telômero , Células-Tronco Pluripotentes Induzidas/metabolismo , Astrócitos/metabolismo , Telomerase/genética , Telômero , Neurônios Motores/metabolismo , Fenótipo
2.
Nat Commun ; 14(1): 6464, 2023 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-37833256

RESUMO

Regulation of global transcription output is important for normal development and disease, but little is known about the mechanisms involved. DNA topoisomerase I (TOP1) is an enzyme well-known for its role in relieving DNA supercoils for enabling transcription. Here, we report a non-enzymatic function of TOP1 that downregulates RNA synthesis. This function is dependent on specific DNA-interacting residues located on a conserved protein surface. A loss-of-function knock-in mutation on this surface, R548Q, is sufficient to cause hypertranscription and alter differentiation outcomes in mouse embryonic stem cells (mESCs). Hypertranscription in mESCs is accompanied by reduced TOP1 chromatin binding and change in genomic supercoiling. Notably, the mutation does not impact TOP1 enzymatic activity; rather, it diminishes TOP1-DNA binding and formation of compact protein-DNA structures. Thus, TOP1 exhibits opposing influences on transcription through distinct activities which are likely to be coordinated. This highlights TOP1 as a safeguard of appropriate total transcription levels in cells.


Assuntos
DNA Topoisomerases Tipo I , Transcrição Gênica , Animais , Camundongos , DNA Topoisomerases Tipo I/metabolismo , Replicação do DNA , Mutação , DNA/genética
3.
Nat Commun ; 14(1): 2439, 2023 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-37117180

RESUMO

Cancer cells undergo transcriptional reprogramming to drive tumor progression and metastasis. Using cancer cell lines and patient-derived tumor organoids, we demonstrate that loss of the negative elongation factor (NELF) complex inhibits breast cancer development through downregulating epithelial-mesenchymal transition (EMT) and stemness-associated genes. Quantitative multiplexed Rapid Immunoprecipitation Mass spectrometry of Endogenous proteins (qPLEX-RIME) further reveals a significant rewiring of NELF-E-associated chromatin partners as a function of EMT and a co-option of NELF-E with the key EMT transcription factor SLUG. Accordingly, loss of NELF-E leads to impaired SLUG binding on chromatin. Through integrative transcriptomic and genomic analyses, we identify the histone acetyltransferase, KAT2B, as a key functional target of NELF-E-SLUG. Genetic and pharmacological inactivation of KAT2B ameliorate the expression of EMT markers, phenocopying NELF ablation. Elevated expression of NELF-E and KAT2B is associated with poorer prognosis in breast cancer patients, highlighting the clinical relevance of our findings. Taken together, we uncover a crucial role of the NELF-E-SLUG-KAT2B epigenetic axis in breast cancer carcinogenesis.


Assuntos
Neoplasias da Mama , Feminino , Humanos , Neoplasias da Mama/patologia , Carcinogênese/genética , Linhagem Celular Tumoral , Cromatina , Epigênese Genética , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Fatores de Transcrição de p300-CBP/metabolismo , Fatores de Transcrição da Família Snail/metabolismo , Fatores de Transcrição/metabolismo
4.
Curr Top Dev Biol ; 151: 43-68, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36681477

RESUMO

Meiosis increases genetic diversity in offspring by generating genetically unique haploid gametes with reshuffled chromosomes. This process requires a specialized set of meiotic proteins, which facilitate chromosome recombination and segregation. However, re-expression of meiotic proteins in mitosis can have catastrophic oncogenic consequences and aberrant expression of meiotic proteins is a common occurrence in human tumors. Mechanistically, re-activation of meiotic genes in cancer promotes oncogenesis likely because cancers-conversely to healthy mitosis-are fueled by genetic instability which promotes tumor evolution, and evasion of immune response and treatment pressure. In this review, we explore similarities between meiotic and cancer cells with a particular focus on the oncogenic activation of meiotic genes in cancer. We emphasize the role of histones and their modifications, DNA methylation, genome organization, R-loops and the availability of distal enhancers.


Assuntos
Meiose , Neoplasias , Humanos , Meiose/genética , Cromossomos , Histonas/genética , Expressão Gênica , Neoplasias/genética
5.
STAR Protoc ; 3(4): 101684, 2022 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-36208454

RESUMO

This protocol describes an optimized workflow for generating 2-cell (2C) stage-like mouse embryonic stem cells (mESCs) for microinjection into 8-cell stage mouse embryos to evaluate the developmental potency of these cells. We detail the following steps: 1) chemical suppression of glycolysis to induce a 2C-like state in mESCs, 2) flow cytometry to enrich for 2C-like cells, 3) embryo microinjection of 2C-like mESCs into 8-cell stage mouse embryos, and finally, 4) immunofluorescence staining of the chimeric blastocysts. For complete details on the use and execution of this protocol, please refer to Hu et al. (2020).


Assuntos
Embrião de Mamíferos , Células-Tronco Embrionárias Murinas , Camundongos , Animais , Diferenciação Celular , Blastocisto
6.
Front Cell Dev Biol ; 10: 931493, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35757000

RESUMO

Epigenetics comprise a diverse array of reversible and dynamic modifications to the cell's genome without implicating any DNA sequence alterations. Both the external environment surrounding the organism, as well as the internal microenvironment of cells and tissues, contribute to these epigenetic processes that play critical roles in cell fate specification and organismal development. On the other hand, dysregulation of epigenetic activities can initiate and sustain carcinogenesis, which is often augmented by inflammation. Chronic inflammation, one of the major hallmarks of cancer, stems from proinflammatory cytokines that are secreted by tumor and tumor-associated cells in the tumor microenvironment. At the same time, inflammatory signaling can establish positive and negative feedback circuits with chromatin to modulate changes in the global epigenetic landscape. In this review, we provide an in-depth discussion of the interconnected crosstalk between epigenetics and inflammation, specifically how epigenetic mechanisms at different hierarchical levels of the genome control inflammatory gene transcription, which in turn enact changes within the cell's epigenomic profile, especially in the context of inflammation-induced cancer.

7.
Stem Cells Int ; 2021: 1624669, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34691189

RESUMO

Transposable elements (TEs) are mobile genetic sequences capable of duplicating and reintegrating at new regions within the genome. A growing body of evidence has demonstrated that these elements play important roles in host genome evolution, despite being traditionally viewed as parasitic elements. To prevent ectopic activation of TE transposition and transcription, they are epigenetically silenced in most somatic tissues. Intriguingly, a specific class of TEs-retrotransposons-is transiently expressed at discrete phases during mammalian development and has been linked to the establishment of totipotency during zygotic genome activation (ZGA). While mechanisms controlling TE regulation in somatic tissues have been extensively studied, the significance underlying the unique transcriptional reactivation of retrotransposons during ZGA is only beginning to be uncovered. In this review, we summarize the expression dynamics of key retrotransposons during ZGA, focusing on findings from in vivo totipotent embryos and in vitro totipotent-like embryonic stem cells (ESCs). We then dissect the functions of retrotransposons and discuss how their transcriptional activities are finetuned during early stages of mammalian development.

8.
Biochem J ; 478(20): 3791-3805, 2021 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-34709374

RESUMO

Meiosis facilitates diversity across individuals and serves as a major driver of evolution. However, understanding how meiosis begins is complicated by fundamental differences that exist between sexes and species. Fundamental meiotic research is further hampered by a current lack of human meiotic cells lines. Consequently, much of what we know relies on data from model organisms. However, contextualising findings from yeast, worms, flies and mice can be challenging, due to marked differences in both nomenclature and the relative timing of meiosis. In this review, we set out to combine current knowledge of signalling and transcriptional pathways that control meiosis initiation across the sexes in a variety of organisms. Furthermore, we highlight the emerging links between meiosis initiation and oncogenesis, which might explain the frequent re-expression of normally silent meiotic genes in a variety of human cancers.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Meiose , Oogênese/genética , Espermatogênese/genética , Animais , Caenorhabditis elegans/citologia , Caenorhabditis elegans/genética , Caenorhabditis elegans/crescimento & desenvolvimento , Caenorhabditis elegans/metabolismo , Carcinogênese/genética , Carcinogênese/metabolismo , Carcinogênese/patologia , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Feminino , Humanos , Masculino , Camundongos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crescimento & desenvolvimento , Saccharomyces cerevisiae/metabolismo , Fatores Sexuais , Transdução de Sinais , Fatores de Tempo , Xenopus laevis/genética , Xenopus laevis/crescimento & desenvolvimento , Xenopus laevis/metabolismo
9.
Cell Prolif ; 54(6): e13049, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33960560

RESUMO

OBJECTIVES: Mouse embryonic stem cells (ESCs) are derived from the inner cell mass of blastocyst-stage embryos and cultured in different culture media with varied pluripotency. Sporadically, a small population of ESCs exhibit 2-cell stage embryonic features in serum containing medium. However, whether ESCs can transit into 2-cell embryo-like (2C-like) cells in the chemically defined media remains largely unknown. MATERIALS AND METHODS: We established a robust in vitro induction system, based on retinoic acid (RA) containing chemically defined media, which can efficiently increase the subpopulation of 2C-like cells. Further test the pluripotency and 2C features of ESCs cultured in RA. 2C reporter-positive cells were selected by FACS; the level of protein was detected via immunofluorescence staining and western blot; the level gene expressions were measured by RNA-seq. RESULTS: Retinoic acid drives a NELFA (negative elongation factor A)-mediated 2C-like state in mouse ESCs, characterized with 2C-specific transcriptional networks and the ability to contribute trophectoderm (TE) when injected into developing embryos. In addition, RA treatment triggers DNA hypomethylation, active histone modification, suppressed glycolysis metabolism and reduced protein synthesis activity of ESCs. CONCLUSIONS: We showed that RA has a broader role in 2C-like cells state, not only is one of the upstream regulators of the 2C-like state in chemically defined media but also illuminates genetic and epigenetic regulations that govern ESCs to 2C-like transition.


Assuntos
Meios de Cultura/farmacologia , Epigênese Genética/efeitos dos fármacos , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Tretinoína/farmacologia , Animais , Blastocisto/citologia , Blastocisto/efeitos dos fármacos , Blastocisto/metabolismo , Células Cultivadas , Feminino , Camundongos , Camundongos Endogâmicos ICR , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo
10.
Nat Cell Biol ; 22(2): 175-186, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31932739

RESUMO

Mouse embryonic stem cells (ESCs) sporadically transit into an early embryonic-like state characterized by the expression of 2-cell (2C) stage-restricted transcripts. Here, we identify a maternal factor-negative elongation factor A (NELFA)-whose heterogeneous expression in mouse ESCs is coupled to 2C gene upregulation and expanded developmental potential in vivo. We show that NELFA partners with Top2a in an interaction specific to the 2C-like state, and that it drives the expression of Dux-a key 2C regulator. Accordingly, loss of NELFA and/or Top2a suppressed Dux activation. Further characterization of 2C-like cells uncovered reduced glycolytic activity; remarkably, mere chemical suppression of glycolysis was sufficient to promote a 2C-like fate, obviating the need for genetic manipulation. Global chromatin state analysis on NELFA-induced cells revealed decommissioning of ESC-specific enhancers, suggesting ESC-state impediments to 2C reversion. Our study positions NELFA as one of the earliest drivers of the 2C-like state and illuminates factors and processes that govern this transition.


Assuntos
Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Embrionárias Murinas/metabolismo , Fatores de Transcrição/genética , Animais , Diferenciação Celular , Cromatina/química , Cromatina/metabolismo , DNA Topoisomerases Tipo II/genética , DNA Topoisomerases Tipo II/metabolismo , Embrião de Mamíferos , Feminino , Glicólise/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Padrões de Herança , Masculino , Camundongos , Camundongos Transgênicos , Células-Tronco Embrionárias Murinas/citologia , Família Multigênica , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose/genética , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo
11.
Wiley Interdiscip Rev Syst Biol Med ; 11(1): e1436, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30225862

RESUMO

The germ line is a crucial cell lineage that is distinct from somatic cells, and solely responsible for the trans-generational transmission of hereditary information in metazoan sexual reproduction. Primordial germ cells (PGCs)-the precursors to functional germ cells-are among the first cell types to be allocated in embryonic development, and this lineage commitment is a critical event in partitioning germ line and somatic tissues. Classically, mammalian PGC development has been largely informed by investigations on mouse embryos and embryonic stem cells. Recent findings from corresponding nonrodent systems, however, have indicated that murine PGC specification may not be fully archetypal. In this review, we outline the current understanding of molecular mechanisms in PGC specification, emphasizing key transcriptional events, and focus on salient differences between early human and mouse PGC commitment. Beyond these latest findings, we also contemplate the future outlook of inquiries in this field, highlighting the importance of comprehensively understanding early fate decisions that underlie the segregation of this unique lineage. This article is categorized under: Developmental Biology > Stem Cell Biology and Regeneration Biological Mechanisms > Cell Fates Physiology > Mammalian Physiology in Health and Disease.


Assuntos
Diferenciação Celular/fisiologia , Embrião de Mamíferos/embriologia , Desenvolvimento Embrionário/fisiologia , Células Germinativas Embrionárias/metabolismo , Animais , Embrião de Mamíferos/citologia , Células Germinativas Embrionárias/citologia , Humanos , Camundongos
12.
Biosci Rep ; 37(2)2017 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-28351896

RESUMO

Gene expression requires successful communication between enhancer and promoter regions, whose activities are regulated by a variety of factors and associated with distinct chromatin structures; in addition, functionally related genes and their regulatory repertoire tend to be arranged in the same subchromosomal regulatory domains. In this review, we discuss the importance of enhancers, especially clusters of enhancers (such as super-enhancers), as key regulatory hubs to integrate environmental cues and encode spatiotemporal instructions for genome expression, which are critical for a variety of biological processes governing mammalian development. Furthermore, we emphasize that the enhancer-promoter interaction landscape provides a critical context to understand the aetiologies and mechanisms behind numerous complex human diseases and provides new avenues for effective transcription-based interventions.


Assuntos
Cromatina/química , Cromatina/genética , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Regiões Promotoras Genéticas , Animais , Genoma , Humanos , Transcrição Gênica
13.
J Clin Invest ; 127(3): 874-887, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28165338

RESUMO

The mechanisms underlying the neurodevelopmental deficits associated with CHARGE syndrome, which include cerebellar hypoplasia, developmental delay, coordination problems, and autistic features, have not been identified. CHARGE syndrome has been associated with mutations in the gene encoding the ATP-dependent chromatin remodeler CHD7. CHD7 is expressed in neural stem and progenitor cells, but its role in neurogenesis during brain development remains unknown. Here we have shown that deletion of Chd7 from cerebellar granule cell progenitors (GCps) results in reduced GCp proliferation, cerebellar hypoplasia, developmental delay, and motor deficits in mice. Genome-wide expression profiling revealed downregulated expression of the gene encoding the glycoprotein reelin (Reln) in Chd7-deficient GCps. Recessive RELN mutations have been associated with severe cerebellar hypoplasia in humans. We found molecular and genetic evidence that reductions in Reln expression contribute to GCp proliferative defects and cerebellar hypoplasia in GCp-specific Chd7 mouse mutants. Finally, we showed that CHD7 is necessary for maintaining an open, accessible chromatin state at the Reln locus. Taken together, this study shows that Reln gene expression is regulated by chromatin remodeling, identifies CHD7 as a previously unrecognized upstream regulator of Reln, and provides direct in vivo evidence that a mammalian CHD protein can control brain development by modulating chromatin accessibility in neuronal progenitors.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Cerebelo/embriologia , Proteínas de Ligação a DNA/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Loci Gênicos , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese , Serina Endopeptidases/metabolismo , Animais , Moléculas de Adesão Celular Neuronais/genética , Cerebelo/anormalidades , Proteínas de Ligação a DNA/genética , Deficiências do Desenvolvimento/genética , Proteínas da Matriz Extracelular/genética , Estudo de Associação Genômica Ampla , Humanos , Camundongos , Camundongos Transgênicos , Transtornos Motores/genética , Transtornos Motores/metabolismo , Proteínas do Tecido Nervoso/genética , Malformações do Sistema Nervoso/embriologia , Malformações do Sistema Nervoso/genética , Proteína Reelina , Serina Endopeptidases/genética
14.
Methods Mol Biol ; 1487: 289-301, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27924576

RESUMO

Chromatin immunoprecipitation (ChIP) is a technique used to determine the association of proteins or histone modifications with chromatin regions in living cells or tissues, and is used extensively in the chromatin biology field to study transcriptional and epigenetic mechanisms. Increasing evidence points to an epigenetic coordination of signaling cascades, such as ERK, that regulate key processes in development and disease, revealing novel principles of gene regulation. Here we describe a detailed protocol for performing chromatin immunoprecipitation followed by qPCR (ChIP-qPCR) for probing histone modifications regulated by ERK signaling in mouse ESCs.


Assuntos
Cromatina/genética , Cromatina/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Transdução de Sinais , Animais , Linhagem Celular , Imunoprecipitação da Cromatina , Sequenciamento de Nucleotídeos em Larga Escala , Código das Histonas , Histonas/metabolismo , Camundongos
15.
Development ; 141(12): 2376-90, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24917497

RESUMO

In pluripotent stem cells, the interplay between signaling cues, epigenetic regulators and transcription factors orchestrates developmental potency. Flexibility in gene expression control is imparted by molecular changes to the nucleosomes, the building block of chromatin. Here, we review the current understanding of the role of chromatin as a plastic and integrative platform to direct gene expression changes in pluripotent stem cells, giving rise to distinct pluripotent states. We will further explore the concept of epigenetic asymmetry, focusing primarily on histone stoichiometry and their associated modifications, that is apparent at both the nucleosome and chromosome-wide levels, and discuss the emerging importance of these asymmetric chromatin configurations in diversifying epigenetic states and their implications for cell fate control.


Assuntos
Cromatina/fisiologia , Células-Tronco Embrionárias/citologia , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Pluripotentes/citologia , Animais , Diferenciação Celular , Linhagem da Célula , Cromatina/química , Histonas/química , Histonas/fisiologia , Humanos , Nucleossomos/metabolismo , Oxigênio/química , Transdução de Sinais , Transcrição Gênica
16.
Cell ; 156(4): 678-90, 2014 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-24529373

RESUMO

Erk1/2 activation contributes to mouse ES cell pluripotency. We found a direct role of Erk1/2 in modulating chromatin features required for regulated developmental gene expression. Erk2 binds to specific DNA sequence motifs typically accessed by Jarid2 and PRC2. Negating Erk1/2 activation leads to increased nucleosome occupancy and decreased occupancy of PRC2 and poised RNAPII at Erk2-PRC2-targeted developmental genes. Surprisingly, Erk2-PRC2-targeted genes are specifically devoid of TFIIH, known to phosphorylate RNA polymerase II (RNAPII) at serine-5, giving rise to its initiated form. Erk2 interacts with and phosphorylates RNAPII at its serine 5 residue, which is consistent with the presence of poised RNAPII as a function of Erk1/2 activation. These findings underscore a key role for Erk1/2 activation in promoting the primed status of developmental genes in mouse ES cells and suggest that the transcription complex at developmental genes is different than the complexes formed at other genes, offering alternative pathways of regulation.


Assuntos
Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Regiões Promotoras Genéticas , RNA Polimerase II/metabolismo , Fator de Transcrição TFIIH/metabolismo , Animais , Cromatina/metabolismo , Células-Tronco Embrionárias/metabolismo , Ativação Enzimática , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Nucleossomos/metabolismo , Fosforilação , Complexo Repressor Polycomb 2/metabolismo
17.
Genes Dev ; 27(12): 1318-38, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23788621

RESUMO

Histone modifications and chromatin-associated protein complexes are crucially involved in the control of gene expression, supervising cell fate decisions and differentiation. Many promoters in embryonic stem (ES) cells harbor a distinctive histone modification signature that combines the activating histone H3 Lys 4 trimethylation (H3K4me3) mark and the repressive H3K27me3 mark. These bivalent domains are considered to poise expression of developmental genes, allowing timely activation while maintaining repression in the absence of differentiation signals. Recent advances shed light on the establishment and function of bivalent domains; however, their role in development remains controversial, not least because suitable genetic models to probe their function in developing organisms are missing. Here, we explore avenues to and from bivalency and propose that bivalent domains and associated chromatin-modifying complexes safeguard proper and robust differentiation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Regiões Promotoras Genéticas/genética , Animais , Diferenciação Celular/genética , Metilação de DNA/genética , Células-Tronco Embrionárias/citologia , Humanos
18.
Genes Dev ; 24(24): 2772-7, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21159818

RESUMO

Prmt5, an arginine methyltransferase, has multiple roles in germ cells, and possibly in pluripotency. Here we show that loss of Prmt5 function is early embryonic-lethal due to the abrogation of pluripotent cells in blastocysts. Prmt5 is also up-regulated in the cytoplasm during the derivation of embryonic stem (ES) cells together with Stat3, where they persist to maintain pluripotency. Prmt5 in association with Mep50 methylates cytosolic histone H2A (H2AR3me2s) to repress differentiation genes in ES cells. Loss of Prmt5 or Mep50 results in derepression of differentiation genes, indicating the significance of the Prmt5/Mep50 complex for pluripotency, which may occur in conjunction with the leukemia inhibitory factor (LIF)/Stat3 pathway.


Assuntos
Desenvolvimento Embrionário , Células-Tronco Embrionárias/citologia , Células-Tronco Pluripotentes/citologia , Proteínas Metiltransferases/fisiologia , Animais , Diferenciação Celular/genética , Citoplasma/química , Regulação da Expressão Gênica no Desenvolvimento , Histonas/metabolismo , Metilação , Camundongos , Proteína-Arginina N-Metiltransferases
19.
Mol Biol Cell ; 17(3): 1421-35, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16394105

RESUMO

We have identified a novel temperature-sensitive mutant of fission yeast alpha-tubulin Atb2 (atb2-983) that contains a single amino acid substitution (V260I). Atb2-983 is incorporated into the microtubules, and their overall structures are not altered noticeably, but microtubule dynamics is compromised during interphase. atb2-983 displays a high rate of chromosome missegregation and is synthetically lethal with deletions in a subset of spindle checkpoint genes including bub1, bub3, and mph1, but not with mad1, mad2, and mad3. During early mitosis in this mutant, Bub1, but not Mad2, remains for a prolonged period in the kinetochores that are situated in proximity to one of the two SPBs (spindle pole bodies). High dosage mal3(+), encoding EB1 homologue, rescues atb2-983, suggesting that Mal3 function is compromised. Consistently, Mal3 localization and binding between Mal3 and Atb2-983 are impaired significantly, and a mal3 single mutant, such as atb2-983, displays prolonged Bub1 kinetochore localization. Furthermore in atb2-983 back-and-forth centromere oscillation during prometaphase is abolished. Intriguingly, this oscillation still occurs in the mal3 mutant, indicating that there is another defect independent of Mal3. These results show that microtubule dynamics is important for coordinated execution of mitotic events, in which Mal3 plays a vital role.


Assuntos
Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/metabolismo , Fuso Acromático/fisiologia , Tubulina (Proteína)/metabolismo , Alelos , Sequência de Aminoácidos , Proteínas de Ciclo Celular/metabolismo , Cromátides/genética , Segregação de Cromossomos/genética , Dosagem de Genes/genética , Cinetocoros/metabolismo , Proteínas Mad2 , Dados de Sequência Molecular , Mutação de Sentido Incorreto/genética , Proteínas Nucleares/metabolismo , Prometáfase , Transporte Proteico , Proteínas Recombinantes de Fusão/metabolismo , Schizosaccharomyces/citologia , Proteínas de Schizosaccharomyces pombe/química , Proteínas de Schizosaccharomyces pombe/genética , Supressão Genética/genética , Tubulina (Proteína)/química , Tubulina (Proteína)/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA