Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 3993, 2023 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-37414772

RESUMO

A lingering question in developmental biology has centered on how transcription factors with widespread distribution in vertebrate embryos can perform tissue-specific functions. Here, using the murine hindlimb as a model, we investigate the elusive mechanisms whereby PBX TALE homeoproteins, viewed primarily as HOX cofactors, attain context-specific developmental roles despite ubiquitous presence in the embryo. We first demonstrate that mesenchymal-specific loss of PBX1/2 or the transcriptional regulator HAND2 generates similar limb phenotypes. By combining tissue-specific and temporally controlled mutagenesis with multi-omics approaches, we reconstruct a gene regulatory network (GRN) at organismal-level resolution that is collaboratively directed by PBX1/2 and HAND2 interactions in subsets of posterior hindlimb mesenchymal cells. Genome-wide profiling of PBX1 binding across multiple embryonic tissues further reveals that HAND2 interacts with subsets of PBX-bound regions to regulate limb-specific GRNs. Our research elucidates fundamental principles by which promiscuous transcription factors cooperate with cofactors that display domain-restricted localization to instruct tissue-specific developmental programs.


Assuntos
Redes Reguladoras de Genes , Fatores de Transcrição , Animais , Camundongos , Proteínas de Homeodomínio/metabolismo , Fator de Transcrição 1 de Leucemia de Células Pré-B/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
2.
J Dev Biol ; 9(4)2021 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-34940502

RESUMO

The highly conserved HOX homeodomain (HD) transcription factors (TFs) establish the identity of different body parts along the antero-posterior axis of bilaterian animals. Segment diversification and the morphogenesis of different structures is achieved by generating precise patterns of HOX expression along the antero-posterior axis and by the ability of different HOX TFs to instruct unique and specific transcriptional programs. However, HOX binding properties in vitro, characterised by the recognition of similar AT-rich binding sequences, do not account for the ability of different HOX to instruct segment-specific transcriptional programs. To address this problem, we previously compared HOXA2 and HOXA3 binding in vivo. Here, we explore if sequence motif enrichments observed in vivo are explained by binding affinities in vitro. Unexpectedly, we found that the highest enriched motif in HOXA2 peaks was not recognised by HOXA2 in vitro, highlighting the importance of investigating HOX binding in its physiological context. We also report the ability of HOXA2 and HOXA3 to heterodimerise, which may have functional consequences for the HOX patterning function in vivo.

4.
Oncogene ; 40(5): 875-884, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33262459

RESUMO

Meningiomas are the most common primary brain tumor and their incidence and prevalence is increasing. This review summarizes current evidence regarding the embryogenesis of the human meninges in the context of meningioma pathogenesis and anatomical distribution. Though not mutually exclusive, chromosomal instability and pathogenic variants affecting the long arm of chromosome 22 (22q) result in meningiomas in neural-crest cell-derived meninges, while variants affecting Hedgehog signaling, PI3K signaling, TRAF7, KLF4, and POLR2A result in meningiomas in the mesodermal-derived meninges of the midline and paramedian anterior, central, and ventral posterior skull base. Current evidence regarding the common pathways for genetic pathogenesis and the anatomical distribution of meningiomas is presented alongside existing understanding of the embryological origins for the meninges prior to proposing next steps for this work.


Assuntos
Proteínas Hedgehog/genética , Fatores de Transcrição Kruppel-Like/genética , Neoplasias Meníngeas/genética , Meningioma/genética , Genótipo , Humanos , Fator 4 Semelhante a Kruppel , Neoplasias Meníngeas/patologia , Meningioma/patologia , Mutação/genética , Fenótipo , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Transdução de Sinais/genética
5.
PLoS Genet ; 16(12): e1009162, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33315856

RESUMO

Gene expression programs determine cell fate in embryonic development and their dysregulation results in disease. Transcription factors (TFs) control gene expression by binding to enhancers, but how TFs select and activate their target enhancers is still unclear. HOX TFs share conserved homeodomains with highly similar sequence recognition properties, yet they impart the identity of different animal body parts. To understand how HOX TFs control their specific transcriptional programs in vivo, we compared HOXA2 and HOXA3 binding profiles in the mouse embryo. HOXA2 and HOXA3 directly cooperate with TALE TFs and selectively target different subsets of a broad TALE chromatin platform. Binding of HOX and tissue-specific TFs convert low affinity TALE binding into high confidence, tissue-specific binding events, which bear the mark of active enhancers. We propose that HOX paralogs, alone and in combination with tissue-specific TFs, generate tissue-specific transcriptional outputs by modulating the activity of TALE TFs at selected enhancers.


Assuntos
Elementos Facilitadores Genéticos , Proteínas de Homeodomínio/metabolismo , Motivos de Aminoácidos , Animais , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/genética , Camundongos , Especificidade de Órgãos , Ligação Proteica , Fatores de Transcrição/metabolismo , Ativação Transcricional , Peixe-Zebra
6.
Nat Commun ; 11(1): 3920, 2020 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-32764605

RESUMO

How the genome activates or silences transcriptional programmes governs organ formation. Little is known in human embryos undermining our ability to benchmark the fidelity of stem cell differentiation or cell programming, or interpret the pathogenicity of noncoding variation. Here, we study histone modifications across thirteen tissues during human organogenesis. We integrate the data with transcription to build an overview of how the human genome differentially regulates alternative organ fates including by repression. Promoters from nearly 20,000 genes partition into discrete states. Key developmental gene sets are actively repressed outside of the appropriate organ without obvious bivalency. Candidate enhancers, functional in zebrafish, allow imputation of tissue-specific and shared patterns of transcription factor binding. Overlaying more than 700 noncoding mutations from patients with developmental disorders allows correlation to unanticipated target genes. Taken together, the data provide a comprehensive genomic framework for investigating normal and abnormal human development.


Assuntos
Deficiências do Desenvolvimento/genética , Epigênese Genética , Organogênese/genética , Animais , Animais Geneticamente Modificados , Bases de Dados Genéticas , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica no Desenvolvimento , Código das Histonas/genética , Humanos , Modelos Genéticos , Mutação , Organogênese/fisiologia , Regiões Promotoras Genéticas , Distribuição Tecidual , Fatores de Transcrição/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética
7.
Nucleic Acids Res ; 48(5): e27, 2020 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-31974574

RESUMO

Transcription factors (TFs) can bind DNA in a cooperative manner, enabling a mutual increase in occupancy. Through this type of interaction, alternative binding sites can be preferentially bound in different tissues to regulate tissue-specific expression programmes. Recently, deep learning models have become state-of-the-art in various pattern analysis tasks, including applications in the field of genomics. We therefore investigate the application of convolutional neural network (CNN) models to the discovery of sequence features determining cooperative and differential TF binding across tissues. We analyse ChIP-seq data from MEIS, TFs which are broadly expressed across mouse branchial arches, and HOXA2, which is expressed in the second and more posterior branchial arches. By developing models predictive of MEIS differential binding in all three tissues, we are able to accurately predict HOXA2 co-binding sites. We evaluate transfer-like and multitask approaches to regularizing the high-dimensional classification task with a larger regression dataset, allowing for the creation of deeper and more accurate models. We test the performance of perturbation and gradient-based attribution methods in identifying the HOXA2 sites from differential MEIS data. Our results show that deep regularized models significantly outperform shallow CNNs as well as k-mer methods in the discovery of tissue-specific sites bound in vivo.


Assuntos
Região Branquial/metabolismo , Aprendizado Profundo , Proteínas de Homeodomínio/genética , Proteína Meis1/genética , RNA/genética , Animais , Sítios de Ligação , Região Branquial/crescimento & desenvolvimento , Imunoprecipitação da Cromatina , Biologia Computacional/métodos , Biologia Computacional/estatística & dados numéricos , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento , Sequenciamento de Nucleotídeos em Larga Escala , Proteínas de Homeodomínio/metabolismo , Camundongos , Modelos Genéticos , Proteína Meis1/metabolismo , Especificidade de Órgãos , Distribuição de Poisson , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA/metabolismo
8.
Dev Biol ; 459(2): 161-180, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-31862379

RESUMO

Animal embryogenesis is initiated by maternal factors, but zygotic genome activation (ZGA) shifts regulatory control to the embryo during blastula stages. ZGA is thought to be mediated by maternally provided transcription factors (TFs), but few such TFs have been identified in vertebrates. Here we report that NF-Y and TALE TFs bind zebrafish genomic elements associated with developmental control genes already at ZGA. In particular, co-regulation by NF-Y and TALE is associated with broadly acting genes involved in transcriptional control, while regulation by either NF-Y or TALE defines genes in specific developmental processes, such that NF-Y controls a cilia gene expression program while TALE controls expression of hox genes. We also demonstrate that NF-Y and TALE-occupied genomic elements function as enhancers during embryogenesis. We conclude that combinatorial use of NF-Y and TALE at developmental enhancers permits the establishment of distinct gene expression programs at zebrafish ZGA.


Assuntos
Fator de Ligação a CCAAT/metabolismo , Expressão Gênica , Genoma , Proteínas de Homeodomínio/metabolismo , Ativação Transcricional , Peixe-Zebra/embriologia , Zigoto/metabolismo , Animais , Cílios/genética , Desenvolvimento Embrionário/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Masculino , Proteínas de Peixe-Zebra
9.
Elife ; 72018 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-29911973

RESUMO

TALE factors are broadly expressed embryonically and known to function in complexes with transcription factors (TFs) like Hox proteins at gastrula/segmentation stages, but it is unclear if such generally expressed factors act by the same mechanism throughout embryogenesis. We identify a TALE-dependent gene regulatory network (GRN) required for anterior development and detect TALE occupancy associated with this GRN throughout embryogenesis. At blastula stages, we uncover a novel functional mode for TALE factors, where they occupy genomic DECA motifs with nearby NF-Y sites. We demonstrate that TALE and NF-Y form complexes and regulate chromatin state at genes of this GRN. At segmentation stages, GRN-associated TALE occupancy expands to include HEXA motifs near PBX:HOX sites. Hence, TALE factors control a key GRN, but utilize distinct DNA motifs and protein partners at different stages - a strategy that may also explain their oncogenic potential and may be employed by other broadly expressed TFs.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Genes Essenciais/genética , Proteínas de Homeodomínio/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Animais , Blástula/embriologia , Blástula/metabolismo , Fator de Ligação a CCAAT/genética , Fator de Ligação a CCAAT/metabolismo , Técnicas de Silenciamento de Genes , Redes Reguladoras de Genes , Proteínas de Homeodomínio/metabolismo , Ligação Proteica , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
10.
Stem Cell Reports ; 9(5): 1387-1394, 2017 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-29056335

RESUMO

To interrogate the alternative fates of pancreas and liver in the earliest stages of human organogenesis, we developed laser capture, RNA amplification, and computational analysis of deep sequencing. Pancreas-enriched gene expression was less conserved between human and mouse than for liver. The dorsal pancreatic bud was enriched for components of Notch, Wnt, BMP, and FGF signaling, almost all genes known to cause pancreatic agenesis or hypoplasia, and over 30 unexplored transcription factors. SOX9 and RORA were imputed as key regulators in pancreas compared with EP300, HNF4A, and FOXA family members in liver. Analyses implied that current in vitro human stem cell differentiation follows a dorsal rather than a ventral pancreatic program and pointed to additional factors for hepatic differentiation. In summary, we provide the transcriptional codes regulating the start of human liver and pancreas development to facilitate stem cell research and clinical interpretation without inter-species extrapolation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Fígado/embriologia , Pâncreas/embriologia , Ativação Transcricional , Transcriptoma , Diferenciação Celular , Perfilação da Expressão Gênica , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Fígado/metabolismo , Pâncreas/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
11.
Elife ; 62017 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-28952437

RESUMO

Connection of the heart to the systemic circulation is a critical developmental event that requires selective preservation of embryonic vessels (aortic arches). However, why some aortic arches regress while others are incorporated into the mature aortic tree remains unclear. By microdissection and deep sequencing in mouse, we find that neural crest (NC) only differentiates into vascular smooth muscle cells (SMCs) around those aortic arches destined for survival and reorganization, and identify the transcription factor Gata6 as a crucial regulator of this process. Gata6 is expressed in SMCs and its target genes activation control SMC differentiation. Furthermore, Gata6 is sufficient to promote SMCs differentiation in vivo, and drive preservation of aortic arches that ought to regress. These findings identify Gata6-directed differentiation of NC to SMCs as an essential mechanism that specifies the aortic tree, and provide a new framework for how mutations in GATA6 lead to congenital heart disorders in humans.


Assuntos
Aorta/embriologia , Diferenciação Celular , Fator de Transcrição GATA6/metabolismo , Miócitos de Músculo Liso/fisiologia , Crista Neural/embriologia , Animais , Expressão Gênica , Camundongos
12.
J Cell Biol ; 216(9): 2603-2605, 2017 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-28801318

RESUMO

How transcription factors (TFs) control enhancer and promoter functions to effect changes in gene expression is an important question. In this issue, Hau et al. (2017. J. Cell Biol. https://doi.org/10.1083/jcb.201701154) show that the TALE TF MEIS recruits the histone modifier PARP1/ARTD1 at promoters to decompact chromatin and activate transcription.


Assuntos
Fatores de Transcrição/genética , Ativação Transcricional , Cromatina , DNA , Regiões Promotoras Genéticas
13.
Curr Opin Genet Dev ; 43: 1-8, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27768937

RESUMO

Homeodomain proteins are evolutionary conserved proteins present in the entire eukaryote kingdom. They execute functions that are essential for life, both in developing and adult organisms. Most homeodomain proteins act as transcription factors and bind DNA to control the activity of other genes. In contrast to their similar DNA binding specificity, homeodomain proteins execute highly diverse and context-dependent functions. Several factors, including genome accessibility, DNA shape, combinatorial binding and the ability to interact with many transcriptional partners, diversify the activity of homeodomain proteins and culminate in the activation of highly dynamic, context-specific transcriptional programs. Clarifying how homeodomain transcription factors work is central to our understanding of development, disease and evolution.


Assuntos
Proteínas de Ligação a DNA/genética , DNA/genética , Proteínas de Homeodomínio/genética , Transcrição Gênica , Sequência de Aminoácidos/genética , Sítios de Ligação/genética , Eucariotos/genética , Humanos
14.
Elife ; 52016 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-27557446

RESUMO

Human organogenesis is when severe developmental abnormalities commonly originate. However, understanding this critical embryonic phase has relied upon inference from patient phenotypes and assumptions from in vitro stem cell models and non-human vertebrates. We report an integrated transcriptomic atlas of human organogenesis. By lineage-guided principal components analysis, we uncover novel relatedness of particular developmental genes across different organs and tissues and identified unique transcriptional codes which correctly predicted the cause of many congenital disorders. By inference, our model pinpoints co-enriched genes as new causes of developmental disorders such as cleft palate and congenital heart disease. The data revealed more than 6000 novel transcripts, over 90% of which fulfil criteria as long non-coding RNAs correlated with the protein-coding genome over megabase distances. Taken together, we have uncovered cryptic transcriptional programs used by the human embryo and established a new resource for the molecular understanding of human organogenesis and its associated disorders.


Assuntos
Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Organogênese , Transcriptoma , Humanos
15.
Development ; 143(14): 2582-92, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27287804

RESUMO

Hmx1 encodes a homeodomain transcription factor expressed in the developing lateral craniofacial mesenchyme, retina and sensory ganglia. Mutation or mis-regulation of Hmx1 underlies malformations of the eye and external ear in multiple species. Deletion or insertional duplication of an evolutionarily conserved region (ECR) downstream of Hmx1 has recently been described in rat and cow, respectively. Here, we demonstrate that the impact of Hmx1 loss is greater than previously appreciated, with a variety of lateral cranioskeletal defects, auriculofacial nerve deficits, and duplication of the caudal region of the external ear. Using a transgenic approach, we demonstrate that a 594 bp sequence encompassing the ECR recapitulates specific aspects of the endogenous Hmx1 lateral facial expression pattern. Moreover, we show that Hoxa2, Meis and Pbx proteins act cooperatively on the ECR, via a core 32 bp sequence, to regulate Hmx1 expression. These studies highlight the conserved role for Hmx1 in BA2-derived tissues and provide an entry point for improved understanding of the causes of the frequent lateral facial birth defects in humans.


Assuntos
Pareamento de Bases/genética , Pavilhão Auricular/metabolismo , Evolução Molecular , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Morfogênese/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Sequência Conservada/genética , Anormalidades Craniofaciais/genética , Pavilhão Auricular/anormalidades , Pavilhão Auricular/patologia , Elementos Facilitadores Genéticos/genética , Face/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Camundongos Mutantes , Camundongos Transgênicos , Especificidade de Órgãos/genética , Estimulação Física , Fator de Transcrição 1 de Leucemia de Células Pré-B , Ligação Proteica/genética , Células Receptoras Sensoriais/patologia
16.
Diabetes ; 64(12): 4184-97, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26324181

RESUMO

Recruitment of innate immune cells from the bone marrow (BM) to an injury site is required for effective repair. In diabetes, this process is altered, leading to excessive recruitment and retention of dysfunctional myeloid cells that fail to promote angiogenesis, prolong inflammation, and block healing. The aberrant myeloid phenotype is partially mediated by stable intrinsic changes to developing cells in the BM that are induced by the diabetic (db) environment, but the exact mechanisms remain largely unknown. Here, we show that the db-derived Gr-1(+)CD11b(+) immature myeloid population has widespread misexpression of chromatin-remodeling enzymes and myeloid differentiation factors. Crucially, diabetes represses transcription of the key myeloid transcription factor CEBPA via diminished H3 Lys 27 promoter acetylation, leading to a failure in monocyte and granulocyte maturation. Restoring Cebpa expression by granulocyte colony-stimulating factor reverses the db phenotype and rescues myeloid maturation. Importantly, our data demonstrate a possible link between myeloid cell maturation and chronic inflammation.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Regulação para Baixo , Hematopoese , Imunidade Inata , Células Mieloides/metabolismo , Adulto , Animais , Biomarcadores/sangue , Biomarcadores/metabolismo , Proteínas Estimuladoras de Ligação a CCAAT/agonistas , Proteínas Estimuladoras de Ligação a CCAAT/sangue , Proteínas Estimuladoras de Ligação a CCAAT/genética , Células Cultivadas , Cruzamentos Genéticos , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/imunologia , Diabetes Mellitus Tipo 2/patologia , Feminino , Granulócitos/imunologia , Granulócitos/metabolismo , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos , Células Mieloides/imunologia , Células Mieloides/patologia , Receptores de Quimiocinas/sangue , Receptores de Quimiocinas/metabolismo , Proteínas Recombinantes/metabolismo
17.
Dev Cell ; 32(3): 265-77, 2015 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-25640223

RESUMO

Hox transcription factors (TFs) are essential for vertebrate development, but how these evolutionary conserved proteins function in vivo remains unclear. Because Hox proteins have notoriously low binding specificity, they are believed to bind with cofactors, mainly homeodomain TFs Pbx and Meis, to select their specific targets. We mapped binding of Meis, Pbx, and Hoxa2 in the branchial arches, a series of segments in the developing vertebrate head. Meis occupancy is largely similar in Hox-positive and -negative arches. Hoxa2, which specifies second arch (IIBA) identity, recognizes a subset of Meis prebound sites that contain Hox motifs. Importantly, at these sites Meis binding is strongly increased. This enhanced Meis binding coincides with active enhancers, which are linked to genes highly expressed in the IIBA and regulated by Hoxa2. These findings show that Hoxa2 operates as a tissue-specific cofactor, enhancing Meis binding to specific sites that provide the IIBA with its anatomical identity.


Assuntos
Região Branquial/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/metabolismo , Animais , Linhagem Celular , Camundongos , Proteína Meis1 , Proteínas de Neoplasias/metabolismo , Fatores de Transcrição/metabolismo
18.
Methods Mol Biol ; 1196: 231-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25151167

RESUMO

Regulation of gene expression must be tightly controlled during embryonic development. A central mechanism to control gene expression is the binding of sequence-specific transcription factors to cis-regulatory elements in the genome. Chromatin immunoprecipitation (ChIP) is a widely used technique to analyze binding of transcription factors and histone modifications on chromatin; however, it is limited to looking at a small number of genes. ChIP with massively parallel sequencing (ChIP-seq) is a recently developed powerful tool to analyze genome-wide binding of transcription factors and histone modifications and provides a vast amount of information into the regulation of gene expression. This chapter describes how ChIP and ChIP-seq are performed on mouse embryonic tissue.


Assuntos
Imunoprecipitação da Cromatina , Embrião de Mamíferos/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Animais , Cromatina/metabolismo , Imunoprecipitação da Cromatina/métodos , DNA/isolamento & purificação , DNA/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Camundongos , Fatores de Transcrição/metabolismo
19.
Stem Cells ; 32(11): 2869-79, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25074424

RESUMO

Mouse embryonic stem cells (mESCs) and epiblast stem cells represent the naïve and primed pluripotent states, respectively. These cells self-renew via distinct signaling pathways and can transition between the two states in the presence of appropriate growth factors. Manipulation of signaling pathways has therefore allowed the isolation of novel pluripotent cell types such as Fibroblast growth factor, Activin and BIO-derived stem cells and IESCs. However, the effect of cell seeding density on pluripotency remains unexplored. In this study, we have examined whether mESCs can epigenetically regulate E-cadherin to enter a primed-like state in response to low cell seeding density. We show that low density seeding in the absence of leukaemia inhibitory factor (LIF) induces decreased apoptosis and maintenance of pluripotency via Activin/Nodal, concomitant with loss of E-cadherin, Signal transducer and activator of transcription phosphorylation, and chimera-forming ability. These cells, E-cadherin negative proliferating stem cells (ENPSCs) can be reverted to a naïve phenotype by addition of LIF or forced E-cadherin expression. However, prolonged culture of ENPSCs without LIF leads to methylation of the E-cadherin promoter (ENPSC(M)), which cannot be reversed by LIF supplementation, and increased histone H3K27 and decreased H3K4 trimethylation. Transcript analysis of ENPSC(M) revealed a primed-like phenotype and their differentiation leads to enrichment of neuroectoderm cells. The generation of ENPSCs is similar to tumorigenesis as ENPSCs exhibit transcript alterations associated with neoplasia, hyperplasia, carcinoma, and metastasis. We therefore describe a novel cell model to elucidate the role of E-cadherin in pluripotency and to investigate epigenetic regulation of this gene during mESC differentiation and tumor metastasis.


Assuntos
Caderinas/metabolismo , Diferenciação Celular/fisiologia , Metilação de DNA , Células-Tronco Embrionárias/metabolismo , Células-Tronco Pluripotentes/citologia , Regiões Promotoras Genéticas , Transdução de Sinais/fisiologia , Animais , Separação Celular , Células Cultivadas , Epigênese Genética/efeitos dos fármacos , Humanos , Fator Inibidor de Leucemia/metabolismo , Camundongos da Linhagem 129 , Células-Tronco Pluripotentes/metabolismo
20.
PLoS One ; 9(6): e100091, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24971829

RESUMO

BACKGROUND AND AIMS: Liver fibrosis is a major cause of morbidity and mortality. It is characterised by excessive extracellular matrix (ECM) deposition from activated hepatic stellate cells (HSCs). Although potentially reversible, treatment remains limited. Understanding how ECM influences the pathogenesis of the disease may provide insight into novel therapeutic targets for the disease. The extracellular protein Epimorphin (EPIM) has been implicated in tissue repair mechanisms in several tissues, partially, through its ability to manipulate proteases. In this study, we have identified that EPIM modulates the ECM environment produced by activated hepatic stellate cells (HSCs), in part, through down-regulation of pro-fibrotic Sex-determining region Y-box 9 (SOX9). METHODS: Influence of EPIM on ECM was investigated in cultured primary rat HSCs. Activated HSCs were treated with recombinant EPIM or SOX9 siRNA. Core fibrotic factors were evaluated by immunoblotting, qPCR and chromatin immunoprecipitation (ChIP). RESULTS: During HSC activation EPIM became significantly decreased in contrast to pro-fibrotic markers SOX9, Collagen type 1 (COL1), and α-Smooth muscle actin (α-SMA). Treatment of activated HSCs with recombinant EPIM caused a reduction in α-SMA, SOX9, COL1 and Osteopontin (OPN), while increasing expression of the collagenase matrix metalloproteinase 13 (MMP13). Sox9 abrogation in activated HSCs increased EPIM and MMP13 expression. CONCLUSION: These data provide evidence for EPIM and SOX9 functioning by mutual negative feedback to regulate attributes of the quiescent or activated state of HSCs. Further understanding of EPIM's role may lead to opportunities to modulate SOX9 as a therapeutic avenue for liver fibrosis.


Assuntos
Células Estreladas do Fígado/metabolismo , Metaloproteinase 13 da Matriz/metabolismo , Glicoproteínas de Membrana/genética , Fatores de Transcrição SOX9/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Colágeno/metabolismo , Modelos Animais de Doenças , Expressão Gênica , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Cirrose Hepática/genética , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Metaloproteinase 13 da Matriz/genética , Glicoproteínas de Membrana/metabolismo , Motivos de Nucleotídeos , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Ligação Proteica , Proteólise , Ratos , Fatores de Transcrição SOX9/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...