Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Methods ; 2024 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-38965443

RESUMO

Recent developments of sequencing-based spatial transcriptomics (sST) have catalyzed important advancements by facilitating transcriptome-scale spatial gene expression measurement. Despite this progress, efforts to comprehensively benchmark different platforms are currently lacking. The extant variability across technologies and datasets poses challenges in formulating standardized evaluation metrics. In this study, we established a collection of reference tissues and regions characterized by well-defined histological architectures, and used them to generate data to compare 11 sST methods. We highlighted molecular diffusion as a variable parameter across different methods and tissues, significantly affecting the effective resolutions. Furthermore, we observed that spatial transcriptomic data demonstrate unique attributes beyond merely adding a spatial axis to single-cell data, including an enhanced ability to capture patterned rare cell states along with specific markers, albeit being influenced by multiple factors including sequencing depth and resolution. Our study assists biologists in sST platform selection, and helps foster a consensus on evaluation standards and establish a framework for future benchmarking efforts that can be used as a gold standard for the development and benchmarking of computational tools for spatial transcriptomic analysis.

2.
Nat Methods ; 20(7): 1048-1057, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37231265

RESUMO

The brain is a complex tissue whose function relies on coordinated anatomical and molecular features. However, the molecular annotation of the spatial organization of the brain is currently insufficient. Here, we describe microfluidic indexing-based spatial assay for transposase-accessible chromatin and RNA-sequencing (MISAR-seq), a method for spatially resolved joint profiling of chromatin accessibility and gene expression. By applying MISAR-seq to the developing mouse brain, we study tissue organization and spatiotemporal regulatory logics during mouse brain development.


Assuntos
Cromatina , Sequenciamento de Nucleotídeos em Larga Escala , Animais , Camundongos , Cromatina/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Análise de Sequência de RNA , Encéfalo , Expressão Gênica , Perfilação da Expressão Gênica
3.
Nature ; 586(7827): E7, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32934359

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

4.
Nature ; 577(7791): E6, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31896818

RESUMO

An Amendment to this paper has been published and can be accessed via a link at the top of the paper.

5.
Development ; 149(12)2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35587592

RESUMO

Vascular establishment is one of the early events in embryogenesis. It is believed that vessel-initiating endothelial progenitors cluster to form the first primitive vessel. Understanding the molecular identity of these progenitors is crucial in order to elucidate lineage hierarchy. In this study, we identify protein C receptor (Procr) as an endothelial progenitor marker and investigate the role of Procr+ progenitors during embryonic vascular development. Using a ProcrmGFP-2A-lacZ reporter, we reveal a much earlier Procr expression (embryonic day 7.5) than previously acknowledged (embryonic day 13.5). Genetic fate-mapping experiments using ProcrCre and ProcrCreER demonstrate that Procr+ cells give rise to blood vessels throughout the entire embryo proper. Single-cell RNA-sequencing analyses place Procr+ cells at the start of endothelial commitment and maturation. Furthermore, targeted ablation of Procr+ cells results in failure of vessel formation and early embryonic lethality. Notably, genetic fate mapping and scRNA-seq pseudotime analysis support the view that Procr+ progenitors can give rise to hemogenic endothelium. In this study, we establish a Procr expression timeline and identify Procr+ vessel-initiating progenitors, and demonstrate their indispensable role in establishment of the vasculature during embryo development.


Assuntos
Hemangioblastos , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário/genética , Receptor de Proteína C Endotelial/genética , Receptor de Proteína C Endotelial/metabolismo , Hemangioblastos/metabolismo
6.
Nature ; 572(7770): 528-532, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31391582

RESUMO

During post-implantation development of the mouse embryo, descendants of the inner cell mass in the early epiblast transit from the naive to primed pluripotent state1. Concurrently, germ layers are formed and cell lineages are specified, leading to the establishment of the blueprint for embryogenesis. Fate-mapping and lineage-analysis studies have revealed that cells in different regions of the germ layers acquire location-specific cell fates during gastrulation2-5. The regionalization of cell fates preceding the formation of the basic body plan-the mechanisms of which are instrumental for understanding embryonic programming and stem-cell-based translational study-is conserved in vertebrate embryos6-8. However, a genome-wide molecular annotation of lineage segregation and tissue architecture of the post-implantation embryo has yet to be undertaken. Here we report a spatially resolved transcriptome of cell populations at defined positions in the germ layers during development from pre- to late-gastrulation stages. This spatiotemporal transcriptome provides high-resolution digitized in situ gene-expression profiles, reveals the molecular genealogy of tissue lineages and defines the continuum of pluripotency states in time and space. The transcriptome further identifies the networks of molecular determinants that drive lineage specification and tissue patterning, supports a role of Hippo-Yap signalling in germ-layer development and reveals the contribution of visceral endoderm to the endoderm in the early mouse embryo.


Assuntos
Linhagem da Célula , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Camadas Germinativas/citologia , Camadas Germinativas/embriologia , Camadas Germinativas/metabolismo , Via de Sinalização Hippo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Serina-Treonina Quinases/metabolismo , Regulon/genética , Transdução de Sinais , Transcriptoma/genética , Proteínas de Sinalização YAP
7.
Ann Rheum Dis ; 83(7): 926-944, 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38325908

RESUMO

OBJECTIVES: Single-cell and spatial transcriptomics analysis of human knee articular cartilage tissue to present a comprehensive transcriptome landscape and osteoarthritis (OA)-critical cell populations. METHODS: Single-cell RNA sequencing and spatially resolved transcriptomic technology have been applied to characterise the cellular heterogeneity of human knee articular cartilage which were collected from 8 OA donors, and 3 non-OA control donors, and a total of 19 samples. The novel chondrocyte population and marker genes of interest were validated by immunohistochemistry staining, quantitative real-time PCR, etc. The OA-critical cell populations were validated through integrative analyses of publicly available bulk RNA sequencing data and large-scale genome-wide association studies. RESULTS: We identified 33 cell population-specific marker genes that define 11 chondrocyte populations, including 9 known populations and 2 new populations, that is, pre-inflammatory chondrocyte population (preInfC) and inflammatory chondrocyte population (InfC). The novel findings that make this an important addition to the literature include: (1) the novel InfC activates the mediator MIF-CD74; (2) the prehypertrophic chondrocyte (preHTC) and hypertrophic chondrocyte (HTC) are potentially OA-critical cell populations; (3) most OA-associated differentially expressed genes reside in the articular surface and superficial zone; (4) the prefibrocartilage chondrocyte (preFC) population is a major contributor to the stratification of patients with OA, resulting in both an inflammatory-related subtype and a non-inflammatory-related subtype. CONCLUSIONS: Our results highlight InfC, preHTC, preFC and HTC as potential cell populations to target for therapy. Also, we conclude that profiling of those cell populations in patients might be used to stratify patient populations for defining cohorts for clinical trials and precision medicine.


Assuntos
Cartilagem Articular , Condrócitos , Osteoartrite do Joelho , Humanos , Condrócitos/patologia , Condrócitos/metabolismo , Osteoartrite do Joelho/patologia , Osteoartrite do Joelho/genética , Cartilagem Articular/patologia , Cartilagem Articular/metabolismo , Pessoa de Meia-Idade , Masculino , Transcriptoma , Estudo de Associação Genômica Ampla , Feminino , Análise de Célula Única/métodos , Idoso , Perfilação da Expressão Gênica/métodos , Hipertrofia/genética , Multiômica
8.
Annu Rev Genomics Hum Genet ; 21: 163-181, 2020 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-32339035

RESUMO

Embryonic development and stem cell differentiation provide a paradigm to understand the molecular regulation of coordinated cell fate determination and the architecture of tissue patterning. Emerging technologies such as single-cell RNA sequencing and spatial transcriptomics are opening new avenues to dissect cell organization, the divergence of morphological and molecular properties, and lineage allocation. Rapid advances in experimental and computational tools have enabled researchers to make many discoveries and revisit old hypotheses. In this review, we describe the use of single-cell RNA sequencing in studies of molecular trajectories and gene regulation networks for stem cell lineages, while highlighting the integratedexperimental and computational analysis of single-cell and spatial transcriptomes in the molecular annotation of tissue lineages and development during postimplantation gastrulation.


Assuntos
Linhagem da Célula , Biologia Computacional/métodos , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Análise de Célula Única/métodos , Transcriptoma , Animais , Diferenciação Celular , Humanos , Análise Espacial
9.
Development ; 146(7)2019 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-30890572

RESUMO

During embryogenesis, the stringent regulation of Wnt activity is crucial for the morphogenesis of the head and brain. The loss of function of the Wnt inhibitor Dkk1 results in elevated Wnt activity, loss of ectoderm lineage attributes from the anterior epiblast, and the posteriorisation of anterior germ layer tissue towards the mesendoderm. The modulation of Wnt signalling may therefore be crucial for the allocation of epiblast cells to ectoderm progenitors during gastrulation. To test this hypothesis, we examined the lineage characteristics of epiblast stem cells (EpiSCs) that were derived and maintained under different signalling conditions. We showed that suppression of Wnt activity enhanced the ectoderm propensity of the EpiSCs. Neuroectoderm differentiation of these EpiSCs was further empowered by the robust re-activation of Wnt activity. Therefore, during gastrulation, the tuning of the signalling activities that mediate mesendoderm differentiation is instrumental for the acquisition of ectoderm potency in the epiblast.


Assuntos
Diferenciação Celular/fisiologia , Ectoderma/citologia , Camadas Germinativas/citologia , Animais , Diferenciação Celular/genética , Células Cultivadas , Ectoderma/metabolismo , Gastrulação/genética , Gastrulação/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Camadas Germinativas/metabolismo , Camundongos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
10.
Nucleic Acids Res ; 48(4): 1828-1842, 2020 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-31853542

RESUMO

The developmental potential of cells, termed pluripotency, is highly dynamic and progresses through a continuum of naive, formative and primed states. Pluripotency progression of mouse embryonic stem cells (ESCs) from naive to formative and primed state is governed by transcription factors (TFs) and their target genes. Genomic techniques have uncovered a multitude of TF binding sites in ESCs, yet a major challenge lies in identifying target genes from functional binding sites and reconstructing dynamic transcriptional networks underlying pluripotency progression. Here, we integrated time-resolved 'trans-omic' datasets together with TF binding profiles and chromatin conformation data to identify target genes of a panel of TFs. Our analyses revealed that naive TF target genes are more likely to be TFs themselves than those of formative TFs, suggesting denser hierarchies among naive TFs. We also discovered that formative TF target genes are marked by permissive epigenomic signatures in the naive state, indicating that they are poised for expression prior to the initiation of pluripotency transition to the formative state. Finally, our reconstructed transcriptional networks pinpointed the precise timing from naive to formative pluripotency progression and enabled the spatiotemporal mapping of differentiating ESCs to their in vivo counterparts in developing embryos.


Assuntos
Desenvolvimento Embrionário/genética , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Pluripotentes/metabolismo , Fatores de Transcrição/genética , Animais , Sítios de Ligação/genética , Diferenciação Celular/genética , Cromatina/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Redes Reguladoras de Genes/genética , Genoma/genética , Camundongos
11.
PLoS Genet ; 15(2): e1007977, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30789911

RESUMO

Heart valve disease is a major clinical problem worldwide. Cardiac valve development and homeostasis need to be precisely controlled. Hippo signaling is essential for organ development and tissue homeostasis, while its role in valve formation and morphology maintenance remains unknown. VGLL4 is a transcription cofactor in vertebrates and we found it was mainly expressed in valve interstitial cells at the post-EMT stage and was maintained till the adult stage. Tissue specific knockout of VGLL4 in different cell lineages revealed that only loss of VGLL4 in endothelial cell lineage led to valve malformation with expanded expression of YAP targets. We further semi-knockout YAP in VGLL4 ablated hearts, and found hyper proliferation of arterial valve interstitial cells was significantly constrained. These findings suggest that VGLL4 is important for valve development and manipulation of Hippo components would be a potential therapy for preventing the progression of congenital valve disease.


Assuntos
Células Endoteliais/citologia , Valvas Cardíacas/crescimento & desenvolvimento , Hipertrofia Ventricular Esquerda/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Linhagem da Célula , Proliferação de Células , Células Endoteliais/metabolismo , Transição Epitelial-Mesenquimal , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Valvas Cardíacas/citologia , Valvas Cardíacas/metabolismo , Via de Sinalização Hippo , Homeostase , Hipertrofia Ventricular Esquerda/veterinária , Camundongos , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais
12.
Genesis ; 56(9): e23246, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30114334

RESUMO

Development of the embryonic head is driven by the activity of gene regulatory networks of transcription factors. LHX1 is a homeobox transcription factor that plays an essential role in the formation of the embryonic head. The loss of LHX1 function results in anterior truncation of the embryo caused by the disruption of morphogenetic movement of tissue precursors and the dysregulation of WNT signaling activity. Profiling the gene expression pattern in the Lhx1 mutant embryo revealed that tissues in anterior germ layers acquire posterior tissue characteristics, suggesting LHX1 activity is required for the allocation and patterning of head precursor tissues. Here, we used LHX1 as an entry point to delineate its transcriptional targets and interactors and construct a LHX1-anchored gene regulatory network. Using a gain-of-function approach, we identified genes that immediately respond to Lhx1 activation. Meta-analysis of the datasets of LHX1-responsive genes and genes expressed in the anterior tissues of mouse embryos at head-fold stage, in conjunction with published Xenopus embryonic LHX1 (Xlim1) ChIP-seq data, has pinpointed the putative transcriptional targets of LHX1 and an array of genetic determinants functioning together in the formation of the mouse embryonic head.


Assuntos
Redes Reguladoras de Genes , Genes Homeobox , Cabeça/embriologia , Proteínas com Homeodomínio LIM/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células-Tronco Embrionárias/metabolismo , Perfilação da Expressão Gênica , Células Germinativas/fisiologia , Transcrição Gênica , Xenopus laevis/embriologia
13.
J Biol Chem ; 292(48): 19590-19604, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28972157

RESUMO

Proper neural commitment is essential for ensuring the appropriate development of the human brain and for preventing neurodevelopmental diseases such as autism spectrum disorders, schizophrenia, and intellectual disorders. However, the molecular mechanisms underlying the neural commitment in humans remain elusive. Here, we report the establishment of a neural differentiation system based on human embryonic stem cells (hESCs) and on comprehensive RNA sequencing analysis of transcriptome dynamics during early hESC differentiation. Using weighted gene co-expression network analysis, we reveal that the hESC neurodevelopmental trajectory has five stages: pluripotency (day 0); differentiation initiation (days 2, 4, and 6); neural commitment (days 8-10); neural progenitor cell proliferation (days 12, 14, and 16); and neuronal differentiation (days 18, 20, and 22). These stages were characterized by unique module genes, which may recapitulate the early human cortical development. Moreover, a comparison of our RNA-sequencing data with several other transcriptome profiling datasets from mice and humans indicated that Module 3 associated with the day 8-10 stage is a critical window of fate switch from the pluripotency to the neural lineage. Interestingly, at this stage, no key extrinsic signals were activated. In contrast, using CRISPR/Cas9-mediated gene knockouts, we also found that intrinsic hub transcription factors, including the schizophrenia-associated SIX3 gene and septo-optic dysplasia-related HESX1 gene, are required to program hESC neural determination. Our results improve the understanding of the mechanism of neural commitment in the human brain and may help elucidate the etiology of human mental disorders and advance therapies for managing these conditions.


Assuntos
Diferenciação Celular/genética , Células-Tronco Embrionárias/metabolismo , Neurônios/citologia , Transcriptoma , Células-Tronco Embrionárias/química , Proteínas do Olho/fisiologia , Proteínas de Homeodomínio/fisiologia , Humanos , Proteínas do Tecido Nervoso/fisiologia , Fatores de Transcrição/genética , Proteína Homeobox SIX3
14.
Dev Growth Differ ; 60(8): 463-472, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30368783

RESUMO

Gastrulation is a key milestone in early mouse development when multipotent epiblast cells are allocated to progenitors of diverse tissue lineages that constitute the ensemble of building blocks of the body plan. The analysis of gene function revealed that the activity of transcription factors is likely to be the fundamental driving force underpinning the lineage specification and tissue patterning in the primary germ layers. The developmental-spatial transcriptome of the gastrulating embryo revealed the concerted and interactive activity of the gene regulatory network anchored by development-related transcription factors. The findings of the network structure offer novel insights into the regionalization of tissue fates and enable tracking of the progression of epiblast patterning, leading to the construction of molecularly annotated fate maps of epiblast during gastrulation.


Assuntos
Gastrulação/genética , Redes Reguladoras de Genes/genética , Camadas Germinativas/metabolismo , Fatores de Transcrição/metabolismo , Animais , Camadas Germinativas/citologia , Camadas Germinativas/crescimento & desenvolvimento , Camundongos
15.
Acta Biochim Biophys Sin (Shanghai) ; 50(1): 68-73, 2018 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-29190317

RESUMO

TGF-ß superfamily signaling pathways essentially contribute to the broad spectrum of early developmental events including embryonic patterning, cell fate determination and dynamic movements. In this review, we first introduced some key developmental processes that require TGF-ß signaling to show the fundamental importance of these pathways. Then we discuss how their activities are regulated, and new findings about how the TGF-ß superfamily ligands bind to the chromatin to regulate transcription during embryo development.


Assuntos
Desenvolvimento Embrionário/genética , Células-Tronco Embrionárias Murinas/metabolismo , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/genética , Animais , Diferenciação Celular/genética , Autorrenovação Celular/genética , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Fator de Crescimento Transformador beta/metabolismo
16.
J Biol Chem ; 291(29): 15212-25, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27226536

RESUMO

Mouse pluripotent cells, such as embryonic stem cells (ESCs) and epiblast stem cells (EpiSCs), provide excellent in vitro systems to study imperative pre- and postimplantation events of in vivo mammalian development. It is known that mouse ESCs are dynamic heterogeneous populations. However, it remains largely unclear whether and how EpiSCs possess heterogeneity and plasticity similar to that of ESCs. Here, we show that EpiSCs are discriminated by the expression of a specific marker T (Brachyury) into two populations. The T-positive (T(+)) and the T-negative (T(-)) populations can be interconverted within the same culture condition. In addition, the two populations display distinct responses to bone morphogenetic protein (BMP) signaling and different developmental potentials. The T(-) EpiSCs are preferentially differentiated into ectoderm lineages, whereas T(+) EpiSCs have a biased potential for mesendoderm fates. Mechanistic studies reveal that T(+) EpiSCs have an earlier and faster response to BMP4 stimulation than T(-) EpiSCs. Id1 mediates the commitment of T(-) EpiSCs to epidermal lineage during BMP4 treatment. On the other hand, Snail modulates the conversion of T(+) EpiSCs to mesendoderm fates with the presence of BMP4. Furthermore, T expression is essential for epithelial-mesenchymal transition during EpiSCs differentiation. Our findings suggest that the dynamic heterogeneity of the T(+)/T(-) subpopulation primes EpiSCs toward particular cell lineages, providing important insights into the dynamic development of the early mouse embryo.


Assuntos
Proteína Morfogenética Óssea 4/farmacologia , Proteínas Fetais/metabolismo , Camadas Germinativas/citologia , Camadas Germinativas/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Proteínas com Domínio T/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Endoderma/citologia , Epiderme/efeitos dos fármacos , Epiderme/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteínas de Fluorescência Verde/metabolismo , Mesoderma/citologia , Camundongos , Modelos Biológicos , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Transdução de Sinais/efeitos dos fármacos
17.
Genome Res ; 22(1): 142-50, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21960545

RESUMO

RNA editing enhances the diversity of gene products at the post-transcriptional level. Approaches for genome-wide identification of RNA editing face two main challenges: separating true editing sites from false discoveries and accurate estimation of editing levels. We developed an approach to analyze transcriptome sequencing data (RNA-seq) for global identification of RNA editing in cells for which whole-genome sequencing data are available. We applied the method to analyze RNA-seq data of a human glioblastoma cell line, U87MG. Around 10,000 DNA-RNA differences were identified, the majority being putative A-to-I editing sites. These predicted A-to-I events were associated with a low false-discovery rate (∼5%). Moreover, the estimated editing levels from RNA-seq correlated well with those based on traditional clonal sequencing. Our results further facilitated unbiased characterization of the sequence and evolutionary features flanking predicted A-to-I editing sites and discovery of a conserved RNA structural motif that may be functionally relevant to editing. Genes with predicted A-to-I editing were significantly enriched with those known to be involved in cancer, supporting the potential importance of cancer-specific RNA editing. A similar profile of DNA-RNA differences as in U87MG was predicted for another RNA-seq data set obtained from primary breast cancer samples. Remarkably, significant overlap exists between the putative editing sites of the two transcriptomes despite their difference in cell type, cancer type, and genomic backgrounds. Our approach enabled de novo identification of the RNA editome, which sets the stage for further mechanistic studies of this important step of post-transcriptional regulation.


Assuntos
Genoma Humano/fisiologia , Edição de RNA/fisiologia , Análise de Sequência de RNA/métodos , Transcriptoma/fisiologia , Linhagem Celular Tumoral , Humanos , Análise de Sequência de RNA/instrumentação
18.
Dev Growth Differ ; 57(2): 109-20, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25708399

RESUMO

Neural fate commitment is an early embryonic event that a group of cells in ectoderm, which do not ingress through primitive streak, acquire a neural fate but not epidermal or mesodermal lineages. Several extracellular signaling pathways initiated by the secreted proteins bone morphogenetic proteins (BMPs), fibroblast growth factors (FGFs), wingless/int class proteins (WNTs) and Nodal play essential roles in the specification of the neural plate. Accumulating evidence from the studies on mouse and pluripotent embryonic stem cells reveals that except for the extracellular signals, the intracellular molecules, including both transcriptional and epigenetic factors, participate in the modulation of neural fate commitment as well. In the review, we mainly focus on recent findings that the initiation of the nervous system is elaborately regulated by the intrinsic programs, which are mediated by transcriptional factors such as Sox2, Zfp521, Sip1 and Pou3f1, as well as epigenetic modifications, including histone methylation/demethylation, histone acetylation/deacetylation, and DNA methylation/demethylation. The discovery of the intrinsic regulatory machineries provides better understanding of the mechanisms by which the neural fate commitment is ensured by the cooperation between extracellular factors and intracellular molecules.


Assuntos
Ectoderma/embriologia , Células-Tronco Embrionárias/metabolismo , Mesoderma/embriologia , Sistema Nervoso/embriologia , Células-Tronco Pluripotentes/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células-Tronco Embrionárias/citologia , Humanos , Camundongos , Sistema Nervoso/citologia , Células-Tronco Pluripotentes/citologia
19.
Nucleic Acids Res ; 40(13): e104, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22467206

RESUMO

Establishing the functional roles of genetic variants remains a significant challenge in the post-genomic era. Here, we present a method, allele-specific alternative mRNA processing (ASARP), to identify genetically influenced mRNA processing events using transcriptome sequencing (RNA-Seq) data. The method examines RNA-Seq data at both single-nucleotide and whole-gene/isoform levels to identify allele-specific expression (ASE) and existence of allele-specific regulation of mRNA processing. We applied the methods to data obtained from the human glioblastoma cell line U87MG and primary breast cancer tissues and found that 26-45% of all genes with sufficient read coverage demonstrated ASE, with significant overlap between the two cell types. Our methods predicted potential mechanisms underlying ASE due to regulations affecting either whole-gene-level expression or alternative mRNA processing, including alternative splicing, alternative polyadenylation and alternative transcriptional initiation. Allele-specific alternative splicing and alternative polyadenylation may explain ASE in hundreds of genes in each cell type. Reporter studies following these predictions identified the causal single nucleotide variants (SNVs) for several allele-specific alternative splicing events. Finally, many genes identified in our study were also reported as disease/phenotype-associated genes in genome-wide association studies. Future applications of our approach may provide ample insights for a better understanding of the genetic basis of gene regulation underlying phenotypic diversity and disease mechanisms.


Assuntos
Alelos , Perfilação da Expressão Gênica , Processamento Pós-Transcricional do RNA , RNA Mensageiro/química , RNA Mensageiro/metabolismo , Análise de Sequência de RNA , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Feminino , Estudo de Associação Genômica Ampla , Humanos , Análise de Sequência de RNA/normas
20.
Circ Res ; 109(12): 1332-41, 2011 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-22034492

RESUMO

RATIONALE: Accurate and comprehensive de novo transcriptome profiling in heart is a central issue to better understand cardiac physiology and diseases. Although significant progress has been made in genome-wide profiling for quantitative changes in cardiac gene expression, current knowledge offers limited insights to the total complexity in cardiac transcriptome at individual exon level. OBJECTIVE: To develop more robust bioinformatic approaches to analyze high-throughput RNA sequencing (RNA-Seq) data, with the focus on the investigation of transcriptome complexity at individual exon and transcript levels. METHODS AND RESULTS: In addition to overall gene expression analysis, the methods developed in this study were used to analyze RNA-Seq data with respect to individual transcript isoforms, novel spliced exons, novel alternative terminal exons, novel transcript clusters (ie, novel genes), and long noncoding RNA genes. We applied these approaches to RNA-Seq data obtained from mouse hearts after pressure-overload-induced by transaortic constriction. Based on experimental validations, analyses of the features of the identified exons/transcripts, and expression analyses including previously published RNA-Seq data, we demonstrate that the methods are highly effective in detecting and quantifying individual exons and transcripts. Novel insights inferred from the examined aspects of the cardiac transcriptome open ways to further experimental investigations. CONCLUSIONS: Our work provided a comprehensive set of methods to analyze mouse cardiac transcriptome complexity at individual exon and transcript levels. Applications of the methods may infer important new insights to gene regulation in normal and disease hearts in terms of exon utilization and potential involvement of novel components of cardiac transcriptome.


Assuntos
Perfilação da Expressão Gênica/métodos , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/fisiopatologia , Coração/fisiologia , Análise de Sequência de RNA/métodos , Animais , Modelos Animais de Doenças , Éxons/genética , Éxons/fisiologia , Regulação da Expressão Gênica/fisiologia , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Transcrição Gênica/genética , Transcrição Gênica/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA