Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 614(7948): 509-520, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36543322

RESUMO

The segmented body plan of vertebrates is established during somitogenesis, a well-studied process in model organisms; however, the details of this process in humans remain largely unknown owing to ethical and technical limitations. Despite recent advances with pluripotent stem cell-based approaches1-5, models that robustly recapitulate human somitogenesis in both space and time remain scarce. Here we introduce a pluripotent stem cell-derived mesoderm-based 3D model of human segmentation and somitogenesis-which we termed 'axioloid'-that captures accurately the oscillatory dynamics of the segmentation clock and the morphological and molecular characteristics of sequential somite formation in vitro. Axioloids show proper rostrocaudal patterning of forming segments and robust anterior-posterior FGF-WNT signalling gradients and retinoic acid signalling components. We identify an unexpected critical role of retinoic acid signalling in the stabilization of forming segments, indicating distinct, but also synergistic effects of retinoic acid and extracellular matrix on the formation and epithelialization of somites. Comparative analysis demonstrates marked similarities of axioloids to the human embryo, further validated by the presence of a Hox code in axioloids. Finally, we demonstrate the utility of axioloids for studying the pathogenesis of human congenital spine diseases using induced pluripotent stem cells with mutations in HES7 and MESP2. Our results indicate that axioloids represent a promising platform for the study of axial development and disease in humans.


Assuntos
Padronização Corporal , Técnicas de Cultura de Células em Três Dimensões , Somitos , Humanos , Padronização Corporal/efeitos dos fármacos , Matriz Extracelular/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Técnicas In Vitro , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Biológicos , Mutação , Somitos/citologia , Somitos/efeitos dos fármacos , Somitos/embriologia , Somitos/metabolismo , Doenças da Coluna Vertebral/patologia , Tretinoína/metabolismo , Tretinoína/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos
2.
Nature ; 580(7801): 124-129, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32238941

RESUMO

Pluripotent stem cells are increasingly used to model different aspects of embryogenesis and organ formation1. Despite recent advances in in vitro induction of major mesodermal lineages and cell types2,3, experimental model systems that can recapitulate more complex features of human mesoderm development and patterning are largely missing. Here we used induced pluripotent stem cells for the stepwise in vitro induction of presomitic mesoderm and its derivatives to model distinct aspects of human somitogenesis. We focused initially on modelling the human segmentation clock, a major biological concept believed to underlie the rhythmic and controlled emergence of somites, which give rise to the segmental pattern of the vertebrate axial skeleton. We observed oscillatory expression of core segmentation clock genes, including HES7 and DKK1, determined the period of the human segmentation clock to be around five hours, and demonstrated the presence of dynamic travelling-wave-like gene expression in in vitro-induced human presomitic mesoderm. Furthermore, we identified and compared oscillatory genes in human and mouse presomitic mesoderm derived from pluripotent stem cells, which revealed species-specific and shared molecular components and pathways associated with the putative mouse and human segmentation clocks. Using CRISPR-Cas9-based genome editing technology, we then targeted genes for which mutations in patients with segmentation defects of the vertebrae, such as spondylocostal dysostosis, have been reported (HES7, LFNG, DLL3 and MESP2). Subsequent analysis of patient-like and patient-derived induced pluripotent stem cells revealed gene-specific alterations in oscillation, synchronization or differentiation properties. Our findings provide insights into the human segmentation clock as well as diseases associated with human axial skeletogenesis.


Assuntos
Relógios Biológicos/fisiologia , Desenvolvimento Embrionário/fisiologia , Células-Tronco Pluripotentes/citologia , Somitos/citologia , Somitos/crescimento & desenvolvimento , Anormalidades Múltiplas/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Relógios Biológicos/genética , Desenvolvimento Embrionário/genética , Edição de Genes , Regulação da Expressão Gênica no Desenvolvimento/genética , Glicosiltransferases/deficiência , Glicosiltransferases/genética , Hérnia Diafragmática/genética , Humanos , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Fenótipo , Somitos/metabolismo , Fatores de Tempo
3.
BMC Bioinformatics ; 24(1): 252, 2023 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-37322439

RESUMO

BACKGROUND: Bioinformatics capability to analyze spatio-temporal dynamics of gene expression is essential in understanding animal development. Animal cells are spatially organized as functional tissues where cellular gene expression data contain information that governs morphogenesis during the developmental process. Although several computational tissue reconstruction methods using transcriptomics data have been proposed, those methods have been ineffective in arranging cells in their correct positions in tissues or organs unless spatial information is explicitly provided. RESULTS: This study demonstrates stochastic self-organizing map clustering with Markov chain Monte Carlo calculations for optimizing informative genes effectively reconstruct any spatio-temporal topology of cells from their transcriptome profiles with only a coarse topological guideline. The method, eSPRESSO (enhanced SPatial REconstruction by Stochastic Self-Organizing Map), provides a powerful in silico spatio-temporal tissue reconstruction capability, as confirmed by using human embryonic heart and mouse embryo, brain, embryonic heart, and liver lobule with generally high reproducibility (average max. accuracy = 92.0%), while revealing topologically informative genes, or spatial discriminator genes. Furthermore, eSPRESSO was used for temporal analysis of human pancreatic organoids to infer rational developmental trajectories with several candidate 'temporal' discriminator genes responsible for various cell type differentiations. CONCLUSIONS: eSPRESSO provides a novel strategy for analyzing mechanisms underlying the spatio-temporal formation of cellular organizations.


Assuntos
Perfilação da Expressão Gênica , Transcriptoma , Humanos , Animais , Camundongos , Reprodutibilidade dos Testes , Encéfalo , Análise por Conglomerados , Análise Espaço-Temporal
4.
Development ; 147(3)2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-32014865

RESUMO

The pluripotent epiblast gives rise to all tissues and organs in the adult body. Its differentiation starts at gastrulation, when the epiblast generates mesoderm and endoderm germ layers through epithelial-mesenchymal transition (EMT). Although gastrulation EMT coincides with loss of epiblast pluripotency, pluripotent cells in development and in vitro can adopt either mesenchymal or epithelial morphology. The relationship between epiblast cellular morphology and its pluripotency is not well understood. Here, using chicken epiblast and mammalian pluripotency stem cell (PSC) models, we show that PSCs undergo a mesenchymal-epithelial transition (MET) prior to EMT-associated pluripotency loss. Epiblast MET and its subsequent EMT are two distinct processes. The former, a partial MET, is associated with reversible initiation of pluripotency exit, whereas the latter, a full EMT, is associated with complete and irreversible pluripotency loss. We provide evidence that integrin-mediated cell-matrix interaction is a key player in pluripotency exit regulation. We propose that epiblast partial MET is an evolutionarily conserved process among all amniotic vertebrates and that epiblast pluripotency is restricted to an intermediate cellular state residing between the fully mesenchymal and fully epithelial states.


Assuntos
Endoderma/citologia , Transição Epitelial-Mesenquimal/fisiologia , Gastrulação/fisiologia , Mesoderma/citologia , Células-Tronco Pluripotentes/citologia , Animais , Diferenciação Celular , Linhagem Celular , Embrião de Galinha , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Morfogênese/genética
6.
Development ; 145(16)2018 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-30139810

RESUMO

Somites (SMs) comprise a transient stem cell population that gives rise to multiple cell types, including dermatome (D), myotome (MYO), sclerotome (SCL) and syndetome (SYN) cells. Although several groups have reported induction protocols for MYO and SCL from pluripotent stem cells, no studies have demonstrated the induction of SYN and D from SMs. Here, we report systematic induction of these cells from human induced pluripotent stem cells (iPSCs) under chemically defined conditions. We also successfully induced cells with differentiation capacities similar to those of multipotent mesenchymal stromal cells (MSC-like cells) from SMs. To evaluate the usefulness of these protocols, we conducted disease modeling of fibrodysplasia ossificans progressiva (FOP), an inherited disease that is characterized by heterotopic endochondral ossification in soft tissues after birth. Importantly, FOP-iPSC-derived MSC-like cells showed enhanced chondrogenesis, whereas FOP-iPSC-derived SCL did not, possibly recapitulating normal embryonic skeletogenesis in FOP and cell-type specificity of FOP phenotypes. These results demonstrate the usefulness of multipotent SMs for disease modeling and future cell-based therapies.


Assuntos
Desenvolvimento Ósseo , Condrogênese , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Biológicos , Miosite Ossificante/metabolismo , Somitos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Miosite Ossificante/patologia , Somitos/patologia
7.
J Med Genet ; 56(9): 622-628, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31015262

RESUMO

BACKGROUND: Congenital scoliosis (CS) is a common vertebral malformation. Spondylocostal dysostosis (SCD) is a rare skeletal dysplasia characterised by multiple vertebral malformations and rib anomalies. In a previous study, a compound heterozygosity for a null mutation and a risk haplotype composed by three single-nucleotide polymorphisms in TBX6 have been reported as a disease-causing model of CS. Another study identified bi-allelic missense variants in a SCD patient. The purpose of our study is to identify TBX6 variants in CS and SCD and examine their pathogenicity. METHODS: We recruited 200 patients with CS or SCD and investigated TBX6 variants. We evaluated the pathogenicity of the variants by in silico prediction and in vitro experiments. RESULTS: We identified five 16p11.2 deletions, one splice-site variant and five missense variants in 10 patients. In vitro functional assays for missense variants identified in the previous and present studies demonstrated that most of the variants caused abnormal localisation of TBX6 proteins. We confirmed mislocalisation of TBX6 proteins in presomitic mesoderm cells induced from SCD patient-derived iPS cells. In induced cells, we found decreased mRNA expressions of TBX6 and its downstream genes were involved in somite formation. All CS patients with missense variants had the risk haplotype in the opposite allele, while a SCD patient with bi-allelic missense variants did not have the haplotype. CONCLUSIONS: Our study suggests that bi-allelic loss of function variants of TBX6 cause a spectrum of phenotypes including CS and SCD, depending on the severity of the loss of TBX6 function.


Assuntos
Anormalidades Múltiplas/diagnóstico , Anormalidades Múltiplas/genética , Alelos , Hérnia Diafragmática/diagnóstico , Hérnia Diafragmática/genética , Mutação com Perda de Função , Escoliose/congênito , Escoliose/diagnóstico , Coluna Vertebral/anormalidades , Proteínas com Domínio T/genética , Biologia Computacional/métodos , Expressão Gênica , Estudos de Associação Genética , Predisposição Genética para Doença , Genótipo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Mutação de Sentido Incorreto
8.
Adv Exp Med Biol ; 1227: 9-27, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32072496

RESUMO

Notch signaling is an evolutionarily conserved pathway associated with the development and differentiation of all metazoans. It is needed for proper germ layer formation and segmentation of the embryo and controls the timing and duration of differentiation events in a dynamic manner. Perturbations of Notch signaling result in blockades of developmental cascades, developmental anomalies, and cancers. An in-depth understanding of Notch signaling is thus required to comprehend the basis of development and cancer, and can be further exploited to understand and direct the outcomes of targeted cellular differentiation into desired cell types and complex tissues from pluripotent or adult stem and progenitor cells. In this chapter, we briefly summarize the molecular, evolutionary, and developmental basis of Notch signaling. We will focus on understanding the basics of Notch signaling and its signaling control mechanisms, its developmental outcomes and perturbations leading to developmental defects, as well as have a brief look at mutations of the Notch signaling pathway causing human hereditary disorders or cancers.


Assuntos
Desenvolvimento Embrionário , Neoplasias/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Diferenciação Celular , Humanos , Neoplasias/patologia , Receptores Notch/genética , Transdução de Sinais/genética , Células-Tronco/citologia , Células-Tronco/metabolismo
9.
Phys Biol ; 16(4): 041002, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-30875695

RESUMO

Epiblast is composed of pluripotent cells which will give rise to all cell lineages in a human body. It forms a single-cell layered epithelium conserved among all amniotic vertebrates (birds, reptiles and mammals) and undergoes complex morphogenesis both before and during gastrulation. Our knowledge of the amniote epiblast is based on data acquired through cellular and molecular analyses of early chick and mouse embryos in vivo and mammalian pluripotent stem cells (PSCs) in vitro. Very few studies have been published on biomechanical characteristics of the amniote epiblast, largely due to lack of experimental tools for measuring and perturbing biomechanical properties. Also missing is a conceptual framework that can integrate both biomechanical and molecular parameters of the epiblast. This review is aimed at providing a background based on which epiblast morphogenesis, including its transition between the epithelial and mesenchymal states, can be understood from a biomechanical perspective. This simple developmental biology system is suitable for testing a multitude of theoretical models in biomechanics, leading to a better understanding of biomechanical logics and constraints governing multicellular organization.


Assuntos
Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Camadas Germinativas/citologia , Camadas Germinativas/crescimento & desenvolvimento , Morfogênese/fisiologia , Animais , Fenômenos Biomecânicos/fisiologia , Comunicação Celular/fisiologia , Linhagem Celular , Gastrulação/fisiologia , Humanos , Modelos Teóricos
10.
Zoolog Sci ; 36(1): 1-4, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31116532

RESUMO

One-egg twins, in general, initiate embryonic development at the same time, and their developmental stages proceed in parallel. Here we report a rare case of the embryonic development of the red-eared slider turtle, Trachemys scripta, in which twins at conspicuously different developmental stages developed on a single yolk. One of the twins appeared to have developed at the normal developmental rate, whereas the development of the other was markedly delayed, despite the absence of any overt anomalies. This observation suggests uncoupled or fully independent differential regulation of embryonic development from either a single or, more likely, two distinct pluripotent blastoderms sharing the same yolk and same environmental conditions.


Assuntos
Desenvolvimento Embrionário , Tartarugas/embriologia , Animais , Blastoderma , Gema de Ovo
11.
Development ; 140(13): 2691-6, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23698348

RESUMO

Mesoderm is formed during gastrulation. This process takes place at the blastopore in lower vertebrates and in the primitive streak (streak) in amniotes. The evolutionary relationship between the blastopore and the streak is unresolved, and the morphogenetic and molecular changes leading to this shift in mesoderm formation during early amniote evolution are not well understood. Using the chick model, we present evidence that the streak is dispensable for mesoderm formation in amniotes. An anamniote-like circumblastoporal mode of gastrulation can be induced in chick and three other amniote species. The induction requires cooperative activation of the FGF and Wnt pathways, and the induced mesoderm field retains anamniote-like dorsoventral patterning. We propose that the amniote streak is homologous to the blastopore in lower vertebrates and evolved from the latter in two distinct steps: an initial pan-amniote posterior restriction of mesoderm-inducing signals; and a subsequent lineage-specific morphogenetic modification of the pre-ingression epiblast.


Assuntos
Gastrulação/fisiologia , Mesoderma/citologia , Linha Primitiva/citologia , Animais , Padronização Corporal/genética , Padronização Corporal/fisiologia , Embrião de Galinha , Galinhas , Embrião não Mamífero , Gástrula/citologia , Gástrula/metabolismo , Gastrulação/genética , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Mesoderma/metabolismo , Morfogênese/genética , Morfogênese/fisiologia , Linha Primitiva/metabolismo , Codorniz , Transdução de Sinais , Tartarugas
12.
Blood ; 121(9): 1543-52, 2013 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-23293081

RESUMO

Programs that control early lineage fate decisions and transitions from embryonic to adult human cell types during development are poorly understood. Using human pluripotent stem cells (hPSCs), in the present study, we reveal reduction of Hedgehog (Hh) signaling correlates to developmental progression of hematopoiesis throughout human ontogeny. Both chemical- and gene-targeting­mediated inactivation of Hh signaling augmented hematopoietic fate and initiated transitions from embryonic to adult hematopoiesis, as measured by globin regulation in hPSCs. Inhibition of the Hh pathway resulted in truncation of Gli3 to its repressor, Gli3R, and was shown to be necessary and sufficient for initiating this transition. Our results reveal an unprecedented role for Hh signaling in the regulation of adult hematopoietic specification, thereby demonstrating the ability to modulate the default embryonic programs of hPSCs.


Assuntos
Proteínas Hedgehog/genética , Hematopoese/genética , Fatores de Transcrição Kruppel-Like/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/fisiologia , Adulto , Células-Tronco Adultas/metabolismo , Células-Tronco Adultas/fisiologia , Células Sanguíneas/metabolismo , Células Sanguíneas/fisiologia , Diferenciação Celular/genética , Células Cultivadas , Regulação para Baixo/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas Hedgehog/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Humanos , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Análise em Microsséries , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Transcriptoma , Proteína Gli3 com Dedos de Zinco
13.
Nat Ecol Evol ; 8(3): 519-535, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38216617

RESUMO

Polyploidy or whole-genome duplication (WGD) is a major event that drastically reshapes genome architecture and is often assumed to be causally associated with organismal innovations and radiations. The 2R hypothesis suggests that two WGD events (1R and 2R) occurred during early vertebrate evolution. However, the timing of the 2R event relative to the divergence of gnathostomes (jawed vertebrates) and cyclostomes (jawless hagfishes and lampreys) is unresolved and whether these WGD events underlie vertebrate phenotypic diversification remains elusive. Here we present the genome of the inshore hagfish, Eptatretus burgeri. Through comparative analysis with lamprey and gnathostome genomes, we reconstruct the early events in cyclostome genome evolution, leveraging insights into the ancestral vertebrate genome. Genome-wide synteny and phylogenetic analyses support a scenario in which 1R occurred in the vertebrate stem-lineage during the early Cambrian, and 2R occurred in the gnathostome stem-lineage, maximally in the late Cambrian-earliest Ordovician, after its divergence from cyclostomes. We find that the genome of stem-cyclostomes experienced an additional independent genome triplication. Functional genomic and morphospace analyses demonstrate that WGD events generally contribute to developmental evolution with similar changes in the regulatory genome of both vertebrate groups. However, appreciable morphological diversification occurred only in the gnathostome but not in the cyclostome lineage, calling into question the general expectation that WGDs lead to leaps of bodyplan complexity.


Assuntos
Feiticeiras (Peixe) , Animais , Filogenia , Feiticeiras (Peixe)/genética , Duplicação Gênica , Vertebrados/genética , Genoma , Lampreias/genética
14.
Development ; 137(17): 2863-74, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20667916

RESUMO

In birds and mammals, all mesoderm cells are generated from the primitive streak. Nascent mesoderm cells contain unique dorsoventral (D/V) identities according to their relative ingression position along the streak. Molecular mechanisms controlling this initial phase of mesoderm diversification are not well understood. Using the chick model, we generated high-quality transcriptomic datasets of different streak regions and analyzed their molecular heterogeneity. Fifteen percent of expressed genes exhibit differential expression levels, as represented by two major groups (dorsal to ventral and ventral to dorsal). A complete set of transcription factors and many novel genes with strong and region-specific expression were uncovered. Core components of BMP, Wnt and FGF pathways showed little regional difference, whereas their positive and negative regulators exhibited both dorsal-to-ventral and ventral-to-dorsal gradients, suggesting that robust D/V positional information is generated by fine-tuned regulation of key signaling pathways at multiple levels. Overall, our study provides a comprehensive molecular resource for understanding mesoderm diversification in vivo and targeted mesoderm lineage differentiation in vitro.


Assuntos
Perfilação da Expressão Gênica , Linha Primitiva/embriologia , Animais , Proteínas Aviárias/genética , Padronização Corporal/genética , Proteínas Morfogenéticas Ósseas/genética , Embrião de Galinha , Mapeamento Cromossômico , Fatores de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Mesoderma/embriologia , Mesoderma/metabolismo , Família Multigênica , Análise de Sequência com Séries de Oligonucleotídeos , Linha Primitiva/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética , Fator de Crescimento Transformador beta/genética , Proteínas Wnt/genética
15.
Dev Growth Differ ; 55(1): 52-9, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23157408

RESUMO

The primitive streak is where the mesoderm and definitive endoderm precursor cells ingress from the epiblast during gastrulation. It is often described as an embryological feature common to all amniotes. But such a feature has not been associated with gastrulation in any reptilian species. A parsimonious model would be that the primitive streak evolved independently in the avian and mammalian lineages. Looking beyond the primitive streak, can one find shared features of mesoderm and endoderm formation during amniote gastrulation? Here, we survey the literature on reptilian gastrulation and provide new data on Brachyury RNA and laminin protein expression in gastrula-stage turtle (Pelodiscus sinensis) embryos. We propose a model to reconcile the primitive streak-associated gastrulation in birds and the blastopore-associated gastrulation in extant reptiles.


Assuntos
Gastrulação , Regulação da Expressão Gênica no Desenvolvimento , Linha Primitiva/citologia , Tartarugas/embriologia , Animais , Evolução Biológica , Aves/embriologia , Endoderma/citologia , Endoderma/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Proteínas Fetais/genética , Gástrula/citologia , Gástrula/metabolismo , Laminina/genética , Laminina/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Filogenia , Linha Primitiva/metabolismo , Proteínas com Domínio T/genética , Tartarugas/genética
16.
Circ Res ; 109(1): 20-37, 2011 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-21566217

RESUMO

The precise and conceptual insight of circulating endothelial progenitor cell (EPC) kinetics is hampered by the absence of an assay system capable of evaluating the EPC differentiation cascade. An assay system for EPC colony formation was developed to delineate circulating EPC differentiation. EPC colony-forming assay using semisolid medium and single or bulk CD133(+) cells from umbilical cord blood exhibited the formation of two types of attaching cell colonies made of small or large cells featuring endothelial lineage potential and properties, termed small EPC colony-forming units and large EPC colony-forming units, respectively. In vitro and in vivo assays of each EPC colony-forming unit cell revealed a differentiation hierarchy from small EPC to large EPC colonies, indicating a primitive EPC stage with highly proliferative activity and a definitive EPC stage with vasculogenic properties, respectively. Experimental comparison with a conventional EPC culture assay system disclosed EPC colony-forming unit cells differentiate into noncolony-forming early EPC. The fate analysis of single CD133(+) cells into the endothelial and hematopoietic lineage was achieved by combining this assay system with a hematopoietic progenitor assay and demonstrated the development of colony-forming EPC and hematopoietic progenitor cells from a single hematopoietic stem cell. EPC colony-forming assay permits the determination of circulating EPC kinetics from single or bulk cells, based on the evaluation of hierarchical EPC colony formation. This assay further enables a proper exploration of possible links between the origin of EPC and hematopoietic stem cells, representing a novel and powerful tool to investigate the molecular signaling pathways involved in EPC biology.


Assuntos
Ensaio de Unidades Formadoras de Colônias/métodos , Células Endoteliais/citologia , Células-Tronco/citologia , Antígeno AC133 , Adulto , Animais , Antígenos CD/análise , Diferenciação Celular , Células Cultivadas , Glicoproteínas/análise , Células-Tronco Hematopoéticas/citologia , Humanos , Receptores de Lipopolissacarídeos/análise , Camundongos , Camundongos Endogâmicos BALB C , Peptídeos/análise , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/farmacologia
17.
Adv Exp Med Biol ; 727: 1-14, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22399335

RESUMO

The Notch signaling pathway is evolutionarily conserved and has been associated with numerous developmental processes, including stem cell maintenance and adult tissue homeostasis. Notably, both abnormal increases and deficiencies of Notch signaling result in human developmental anomalies and cancer development implying that the precise regulation of the intensity and duration of Notch signals is imperative. Numerous studies have demonstrated that the aberrant gain or loss of Notch signaling pathway components is critically linked to multiple human diseases. In this chapter, we will briefly summarize the molecular basis of Notch signaling, focusing on the modulation of Notch signals, and its developmental outcomes including vessel formation and the onset of cancer.


Assuntos
Receptores Notch/fisiologia , Adulto , Comunicação Celular , Diferenciação Celular , Homeostase , Humanos , Transdução de Sinais
18.
Dev Dyn ; 240(8): 2002-10, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21761483

RESUMO

An important function of the vascular system is nutrient delivery. In adult animals, this is mediated through a close contact of the mesoderm-derived vasculature with the endoderm-derived enterocytes and hepatocytes. During embryonic development, the yolk sac (YS) endoderm has been suggested to play a similar role. Physiological and molecular nature of the contact between the YS endoderm and the vasculature is not well-understood. To understand roles of the YS endoderm in early development, we used the avian model and carried out a gene expression profiling analysis of isolated area vasculosa YS endoderm tissues from embryonic day 2-4 chick embryos, covering the first 48 hr of postcirculation development. Genes involved in lipid metabolism are highly enriched, indicating an active modification of lipid components during their transfer from the yolk to the circulatory system. We also uncovered genes encoding major serum proteins and key regulators of vascular integrity. In particular, PTGDS, an enzyme controlling the last step of prostaglandin D2 production, shows high expression in the YS endoderm. Experimental introduction of prostaglandin D2 into embryonic circulation led to intraembryonic vessel rupture. These data suggest that the YS endoderm is the major, if not exclusive, source of lipid and protein constituents of the early embryonic serum and plays an important role in the regulation of vascular integrity in developing embryo.


Assuntos
Proteínas Sanguíneas/metabolismo , Embrião de Galinha/irrigação sanguínea , Embrião de Galinha/embriologia , Endoderma/química , Metabolismo dos Lipídeos , Saco Vitelino/anatomia & histologia , Animais , Proteínas Sanguíneas/genética , Desenvolvimento Embrionário/fisiologia , Endoderma/metabolismo , Perfilação da Expressão Gênica , Humanos , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/metabolismo , Lipocalinas/genética , Lipocalinas/metabolismo , Análise em Microsséries , Saco Vitelino/metabolismo
19.
Curr Opin Genet Dev ; 77: 101985, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36244078

RESUMO

The successful derivation and culture of pluripotent stem cells (PSCs) is tightly connected with the study of embryonic development, and was made largely possible by advances in in vitro fertilization and blastocyst culture during the latter half of the last century [1,2]. Since then, embryonic and induced pluripotent stem cells have been extensively used to derive a plethora of functional cell types in vitro, heavily relying on and utilizing insights into cellular differentiation won from developmental biological studies in model organisms. Excitingly, PSCs are now being increasingly used to reconstitute and analyze complex aspects of mouse and human embryonic development. These bottom-up approaches are starting to provide novel insights into core developmental processes and biological questions and may ultimately help decipher the biological principles that underlie the emergence of form and function during development. This mini review summarizes the latest advances and recent breakthroughs in this rapidly growing field of research on PSC-based in vitro models of early embryonic development.


Assuntos
Células-Tronco Pluripotentes Induzidas , Células-Tronco Pluripotentes , Gravidez , Feminino , Humanos , Gastrulação , Diferenciação Celular/genética , Desenvolvimento Embrionário/genética
20.
Stem Cells ; 28(2): 365-75, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19859984

RESUMO

Lnk is an intracellular adaptor protein reported as a negative regulator of proliferation in c-Kit positive, Sca-1 positive, lineage marker-negative (KSL) bone marrow cells. The KSL fraction in mouse bone marrow is believed to represent a population of hematopoietic and endothelial progenitor cells (EPCs). We report here that, in vitro, Lnk(-/-) KSL cells form more EPC colonies than Lnk(+/+) KSL cells and show higher expression levels of endothelial marker genes, including CD105, CD144, Tie-1, and Tie2, than their wild-type counterparts. In vivo, the administration of Lnk(+/+) KSL cells to a mouse spinal cord injury model promoted angiogenesis, astrogliosis, axon growth, and functional recovery following injury, with Lnk(-/-) KSL being significantly more effective in inducing and promoting these regenerative events. At day 3 following injury, large vessels could be observed in spinal cords treated with KSL cells, and reactive astrocytes were found to have migrated along these large vessels. We could further show that the enhancement of astrogliosis appears to be caused in conjunction with the acceleration of angiogenesis. These findings suggest that Lnk deletion reinforces the commitment of KSL cells to EPCs, promoting subsequent repair of injured spinal cord through the acceleration of angiogenesis and astrogliosis.


Assuntos
Astrócitos/fisiologia , Células da Medula Óssea/citologia , Células-Tronco Hematopoéticas/fisiologia , Neovascularização Fisiológica/fisiologia , Proteínas/fisiologia , Traumatismos da Medula Espinal/fisiopatologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana , Camundongos , Camundongos Knockout , Neovascularização Fisiológica/genética , Proteínas/genética , Traumatismos da Medula Espinal/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA