Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
EMBO J ; 41(5): e110023, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35128689

RESUMO

After entering the adult thymus, bipotent T-cell progenitors give rise to αß or γδ T cells. To determine whether the γδ T-cell receptor (TCR) has an instructive role in γδ T-cell lineage commitment or only "confirms" a pre-established γδ Τ-cell lineage state, we exploited mice lacking expression of LAT, an adaptor required for γδ TCR signaling. Although these mice showed a T-cell development block at the CD4- CD8- double-negative third (DN3) stage, 0.3% of their DN3 cells expressed intermediate levels of γδ TCR (further referred to as γδint ) at their surface. Single-cell transcriptomics of LAT-deficient DN3 γδint cells demonstrated no sign of commitment to the γδ T-cell lineage, apart from γδ TCR expression. Although the lack of LAT is thought to tightly block DN3 cell development, we unexpectedly found that 25% of LAT-deficient DN3 γδint cells were actively proliferating and progressed up to the DN4 stage. However, even those cells failed to turn on the transcriptional program associated with the γδ T-cell lineage. Therefore, the γδ TCR-LAT signaling axis builds upon a γδ T-cell uncommitted lineage state to fully instruct adult γδ T-cell lineage specification.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Linhagem da Célula/genética , Receptores de Antígenos de Linfócitos T gama-delta/genética , Transcriptoma/genética , Animais , Proliferação de Células/genética , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Transdução de Sinais/genética
2.
Development ; 149(22)2022 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-36326003

RESUMO

Stem cell-derived three-dimensional (3D) gastruloids show a remarkable capacity of self-organisation and recapitulate many aspects of gastrulation stage mammalian development. Gastruloids can be rapidly generated and offer several experimental advantages, such as scalability, observability and accessibility for manipulation. Here, we present approaches to further expand the experimental potency of murine 3D gastruloids by using functional genetics in mouse embryonic stem cells (mESCs) to generate chimeric gastruloids. In chimeric gastruloids, fluorescently labelled cells of different genotypes harbouring inducible gene expression or loss-of-function alleles are combined with wild-type cells. We showcase this experimental approach in chimeric gastruloids of mESCs carrying homozygous deletions of the Tbx transcription factor brachyury or inducible expression of Eomes. Resulting chimeric gastruloids recapitulate reported Eomes and brachyury functions, such as instructing cardiac fate and promoting posterior axial extension, respectively. Additionally, chimeric gastruloids revealed previously unrecognised phenotypes, such as the tissue sorting preference of brachyury deficient cells to endoderm and the cell non-autonomous effects of brachyury deficiency on Wnt3a patterning along the embryonic axis, demonstrating some of the advantages of chimeric gastruloids as an efficient tool for studies of mammalian gastrulation.


Assuntos
Gastrulação , Mamíferos , Animais , Camundongos , Endoderma , Células-Tronco Embrionárias Murinas , Alelos
3.
Mol Reprod Dev ; 90(2): 98-108, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36528861

RESUMO

As a highly conserved DNA polymerase (Pol), Pol δ plays crucial roles in chromosomal DNA synthesis and various DNA repair pathways. However, the function of POLD2, the second small subunit of DNA Pol δ (p50 subunit), has not been characterized in vivo during mammalian development. Here, we report for the first time, the essential role of subunit POLD2 during early murine embryogenesis. Although Pold2 mutant mouse embryos exhibit normal morphology at E3.5 blastocyst stage, they cannot be recovered at gastrulation stages. Outgrowth assays reveal that mutant blastocysts cannot hatch from the zona pellucida, indicating impaired blastocyst function. Notably, these phenotypes can be recapitulated by small interfering RNA (siRNA)-mediated knockdown, which also exhibit slowed cellular proliferation together with skewed primitive endoderm and epiblast allocation during the second cell lineage specification. In summary, our study demonstrates that POLD2 is essential for the earliest steps of mammalian development, and the retarded proliferation and embryogenesis may also alter the following cell lineage specifications in the mouse blastocyst embryos.


Assuntos
Blastocisto , DNA Polimerase III , Desenvolvimento Embrionário , Animais , Camundongos , Blastocisto/metabolismo , Linhagem da Célula , Endoderma/metabolismo , Camadas Germinativas , Mamíferos , DNA Polimerase III/metabolismo
4.
Biol Reprod ; 106(4): 699-709, 2022 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-35024788

RESUMO

Female infertility is a heterogeneous disorder with a variety of complex causes, including inflammation and oxidative stress, which are also closely associated with the pathogenesis of polycystic ovary syndrome (PCOS). As a new treatment for PCOS, berberine (BER), a natural compound from Berberis, has been clinically applied recently. However, the mechanisms underlying the association between BER and embryogenesis are still largely unknown. In this study, effects of BER on preimplantation development were evaluated under both normal and inflammatory culture conditions induced by lipopolysaccharide (LPS) in mice. Our data first suggest that BER itself (25 nM) does not affect embryo quality or future developmental potency; however, it can effectively alleviate LPS-induced embryo damage by mitigating apoptosis via reactive oxygen species (ROS)-/caspase-3-dependent pathways and by suppressing proinflammatory cytokines via inhibition of the NF-κB signaling pathway during preimplantation embryonic development. In addition, skewed cell lineage specification in the inner cell mass (ICM) and primitive endoderm (PE) caused by LPS can also be successfully rescued with BER. In summary, these findings for the first time demonstrate the nontoxicity of low doses of BER and its antiapoptotic and antioxidative properties on embryonic cells during mammalian preimplantation development.


Assuntos
Berberina , Síndrome do Ovário Policístico , Animais , Apoptose , Berberina/farmacologia , Linhagem da Célula , Feminino , Humanos , Lipopolissacarídeos/toxicidade , Mamíferos , Camundongos , Gravidez
5.
Biol Reprod ; 104(2): 325-335, 2021 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-33246328

RESUMO

Zinc finger domains of the Cys-Cys-Cys-His (CCCH) class are evolutionarily conserved proteins that bind nucleic acids and are involved in various biological processes. Nearly 60 CCCH-type zinc finger proteins have been identified in humans and mice, most have not been functionally characterized. Here, we provide the first in vivo functional characterization of ZC3H4-a novel CCCH-type zinc finger protein. Our results show that although Zc3h4 mutant embryos exhibit normal morphology at E3.5 blastocyst stage, they cannot be recovered at E7.5 early post-gastrulation stage, suggesting implantation failure. Outgrowth assays reveal that mutant blastocysts either fail to hatch from the zona pellucida, or can hatch but do not form a typical inner cell mass colony, the source of embryonic stem cells (ESCs). Although there is no change in levels of reactive oxygen species, Zc3h4 mutants display severe DNA breaks and reduced cell proliferation. Analysis of lineage specification reveals that both epiblast and primitive endoderm lineages are compromised with severe reductions in cell number and/or specification in the mutant blastocysts. In summary, these findings demonstrate the essential role of ZC3H4 during early mammalian embryogenesis.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Implantação do Embrião/fisiologia , Desenvolvimento Embrionário/fisiologia , Animais , Proliferação de Células/genética , Quebras de DNA , Proteínas de Ligação a DNA/genética , Implantação do Embrião/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Camundongos , Camundongos Knockout , Mutação
6.
Development ; 144(8): 1450-1461, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28289135

RESUMO

The separation of embryonic from extra-embryonic tissues within the inner cell mass to generate the epiblast (EPI), which will form the new organism, from the primitive endoderm (PE), which will form the yolk sac, is a crucial developmental decision. Here, we identify a chromatin modifier, Satb1, with a distinct role in this decision. Satb1 is differentially expressed within 16-cell-stage embryos, with higher expression levels in the inner cell mass progenitor cells. Depleting Satb1 increases the number of EPI cells at the expense of PE. This phenotype can be rescued by simultaneous depletion of both Satb1 and Satb2, owing to their antagonistic effect on the pluripotency regulator Nanog. Consequently, increasing Satb1 expression leads to differentiation into PE and a decrease in EPI, as a result of the modulation of expression of several pluripotency- and differentiation-related genes by Satb1. Finally, we show that Satb1 is a downstream target of the Fgf signalling pathway, linking chromatin modification and Fgf signalling. Together, these results identify a role for Satb1 in the lineage choice between pluripotency and differentiation and further our understanding of early embryonic lineage segregation.


Assuntos
Linhagem da Célula , Cromatina/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Transdução de Sinais , Animais , Contagem de Células , Linhagem da Célula/genética , Desenvolvimento Embrionário/genética , Endoderma/citologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Camadas Germinativas/citologia , Proteínas de Ligação à Região de Interação com a Matriz/genética , Camundongos Endogâmicos C57BL , Modelos Biológicos , Fenótipo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/genética , Fatores de Tempo , Fatores de Transcrição/metabolismo
7.
Biol Reprod ; 103(1): 13-23, 2020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32285100

RESUMO

Retinoblastoma-binding protein 4 (RBBP4) (also known as chromatin-remodeling factor RBAP48) is an evolutionarily conserved protein that has been involved in various biological processes. Although a variety of functions have been attributed to RBBP4 in vitro, mammalian RBBP4 has not been studied in vivo. Here we report that RBBP4 is essential during early mouse embryo development. Although Rbbp4 mutant embryos exhibit normal morphology at E3.5 blastocyst stage, they cannot be recovered at E7.5 early post-gastrulation stage, suggesting an implantation failure. Outgrowth (OG) assays reveal that mutant blastocysts cannot hatch from the zona or can hatch but then arrest without further development. We find that while there is no change in proliferation or levels of reactive oxygen species, both apoptosis and histone acetylation are significantly increased in mutant blastocysts. Analysis of lineage specification reveals that while the trophoblast is properly specified, both epiblast and primitive endoderm lineages are compromised with severe reductions in cell number and/or specification. In summary, these findings demonstrate the essential role of RBBP4 during early mammalian embryogenesis.


Assuntos
Apoptose , Blastocisto/fisiologia , Perda do Embrião , Endoderma/embriologia , Histonas/metabolismo , Proteína 4 de Ligação ao Retinoblastoma/fisiologia , Acetilação , Animais , Implantação do Embrião/fisiologia , Desenvolvimento Embrionário/fisiologia , Endoderma/citologia , Feminino , Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 4 de Ligação ao Retinoblastoma/deficiência , Proteína 4 de Ligação ao Retinoblastoma/genética
8.
FASEB J ; 33(10): 11326-11337, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31322925

RESUMO

In mammals, the early embryo travels down the oviduct to the uterus and prepares for implantation. The unique features of preimplantation development include compaction followed by blastocyst formation. This first cell lineage specification involves various proteins including cell polarity regulators, kinases, and transcription factors. In this study, a novel gene named predicted gene 11545 (Gm11545) expressed predominantly in mouse early embryos was identified and characterized at the transcript, protein, cellular, and functional levels. The Gm11545 protein localized to both cytoplasmic and membrane regions of preimplantation embryos. Remarkably, knockdown of Gm11545 led to arrest of mouse embryos at the morula stage and consequent impairment of blastocyst formation. Expression patterns of the key transcription factors critical for early lineage specification, octamer-binding transcription factor 4 and caudal type homeobox 2, were affected by Gm11545 depletion. Based on the collective findings, we propose that the novel protein identified in this study, Gm11545, is implicated in cell proliferation and cell lineage specification critical for blastocyst formation.-Kim, J., Kim, J., Jeong, J., Hong, S. H., Kim, D., Choi, S., Choi, I., Oh, J. S., Cho, C. Identification of a novel embryo-prevalent gene, Gm11545, involved in preimplantation embryogenesis in mice.


Assuntos
Blastocisto/fisiologia , Implantação do Embrião/genética , Embrião de Mamíferos/fisiologia , Desenvolvimento Embrionário/genética , Fatores de Transcrição/genética , Animais , Linhagem da Célula/genética , Polaridade Celular/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Masculino , Camundongos
9.
Biochim Biophys Acta Mol Cell Res ; 1864(11 Pt B): 2228-2239, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28739265

RESUMO

A disintegrin and metalloproteinases (ADAMs) are a family of mSultidomain, membrane-anchored proteases that regulate diverse cellular functions, including cell adhesion, migration, proteolysis and other cell signaling events. Catalytically-active ADAMs act as ectodomain sheddases that proteolytically cleave type I and type II transmembrane proteins and some GPI-anchored proteins from the cellular surface. ADAMs can also modulate other cellular signaling events through a process known as regulated intramembrane proteolysis (RIP). Through their proteolytic activity, ADAMs can rapidly modulate key cell signaling pathways in response to changes in the extracellular environment (e.g. inflammation) and play a central role in coordinating intercellular communication. Dysregulation of these processes through aberrant expression, or sustained ADAM activity, is linked to chronic inflammation, inflammation-associated cancer and tumorigenesis. ADAM10 was the first disintegrin-metalloproteinase demonstrated to have proteolytic activity and is the prototypic ADAM associated with RIP activity (e.g. sequential Notch receptor processing). ADAM10 is abundantly expressed throughout the gastrointestinal tract and during normal intestinal homeostasis ADAM10 regulates many cellular processes associated with intestinal development, cell fate specification and maintenance of intestinal stem cell/progenitor populations. In addition, several signaling pathways that undergo ectodomain shedding by ADAM10 (e.g. Notch, EGFR/ErbB, IL-6/sIL-6R) help control intestinal injury/regenerative responses and may drive intestinal inflammation and colon cancer initiation and progression. Here, I review some of the proposed functions of ADAM10 associated with intestinal crypt homeostasis and tumorigenesis within the gastrointestinal tract in vivo. This article is part of a Special Issue entitled: Proteolysis as a Regulatory Event in Pathophysiology edited by Stefan Rose-John.


Assuntos
Proteína ADAM10/genética , Secretases da Proteína Precursora do Amiloide/genética , Carcinogênese/genética , Proteínas de Membrana/genética , Neoplasias/genética , Proteólise , Proteína ADAM10/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Membrana Celular/genética , Humanos , Inflamação/genética , Inflamação/patologia , Proteínas de Membrana/metabolismo , Neoplasias/patologia
10.
Development ; 141(19): 3637-48, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25209243

RESUMO

During blastocyst formation, inner cell mass (ICM) cells differentiate into either epiblast (Epi) or primitive endoderm (PrE) cells, labeled by Nanog and Gata6, respectively, and organized in a salt-and-pepper pattern. Previous work in the mouse has shown that, in absence of Nanog, all ICM cells adopt a PrE identity. Moreover, the activation or the blockade of the Fgf/RTK pathway biases cell fate specification towards either PrE or Epi, respectively. We show that, in absence of Gata6, all ICM cells adopt an Epi identity. Furthermore, the analysis of Gata6(+/-) embryos reveals a dose-sensitive phenotype, with fewer PrE-specified cells. These results and previous findings have enabled the development of a mathematical model for the dynamics of the regulatory network that controls ICM differentiation into Epi or PrE cells. The model describes the temporal dynamics of Erk signaling and of the concentrations of Nanog, Gata6, secreted Fgf4 and Fgf receptor 2. The model is able to recapitulate most of the cell behaviors observed in different experimental conditions and provides a unifying mechanism for the dynamics of these developmental transitions. The mechanism relies on the co-existence between three stable steady states (tristability), which correspond to ICM, Epi and PrE cells, respectively. Altogether, modeling and experimental results uncover novel features of ICM cell fate specification such as the role of the initial induction of a subset of cells into Epi in the initiation of the salt-and-pepper pattern, or the precocious Epi specification in Gata6(+/-) embryos.


Assuntos
Massa Celular Interna do Blastocisto/citologia , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Fator de Transcrição GATA6/metabolismo , Redes Reguladoras de Genes/fisiologia , Modelos Biológicos , Transdução de Sinais/fisiologia , Animais , Endoderma/citologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Camadas Germinativas/citologia , Proteínas de Homeodomínio/metabolismo , Hibridização in Situ Fluorescente , Indóis , Camundongos , Microscopia Confocal , Proteína Homeobox Nanog , Transdução de Sinais/genética , Estatísticas não Paramétricas
11.
Development ; 140(20): 4129-44, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24086078

RESUMO

Sox transcription factors play widespread roles during development; however, their versatile funtions have a relatively simple basis: the binding of a Sox protein alone to DNA does not elicit transcriptional activation or repression, but requires binding of a partner transcription factor to an adjacent site on the DNA. Thus, the activity of a Sox protein is dependent upon the identity of its partner factor and the context of the DNA sequence to which it binds. In this Primer, we provide an mechanistic overview of how Sox family proteins function, as a paradigm for transcriptional regulation of development involving multi-transcription factor complexes, and we discuss how Sox factors can thus regulate diverse processes during development.


Assuntos
Diferenciação Celular , Linhagem da Célula , Fatores de Transcrição SOX/metabolismo , Células-Tronco/citologia , Animais , Proteínas de Ligação a DNA/metabolismo , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Grupo de Alta Mobilidade/metabolismo , MicroRNAs/metabolismo , Células-Tronco/metabolismo , Ativação Transcricional
12.
Biol Reprod ; 88(6): 158, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23677984

RESUMO

Mice and cattle use distinct pathways for the first cell segregation into inner cell mass (ICM) and trophectoderm (TE) lineages at the blastocyst stage. However, limited knowledge is available regarding the reliable transcriptional networks that orchestrate the complex developmental processes at this stage in nonrodent species. In order to elucidate the site-dominant transcriptomic properties of bovine blastocysts, we separated cell samples into the ICM and TE using both mechanical and chemical methods and performed in silico prescreening for candidate genes that were site-dominantly expressed in bovine blastocysts. We further performed quantitative real-time PCR and in situ hybridization using the site-specific cell samples. As a result, we identified seven ICM-dominant genes and five TE-dominant genes not found in earlier studies. Our findings provide novel insights into the mechanism of cell-fate specification in the pre-implantation bovine embryo.


Assuntos
Blastocisto/citologia , Linhagem da Célula/genética , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Animais , Blastocisto/metabolismo , Massa Celular Interna do Blastocisto/metabolismo , Bovinos , Diferenciação Celular/genética , Técnicas de Cultura Embrionária , Feminino , Fertilização in vitro
13.
Autoimmun Rev ; 20(12): 102984, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34718162

RESUMO

The mechanistic target of rapamycin (mTOR) pathway integrates metabolic cues into cell fate decisions. A particularly fateful event during the adaptive immune response is the engagement of a T cell receptor by its cognate antigen presented by an antigen-presenting cell (APC). Here, the induction of adequate T cell activation and lineage specification is critical to mount protective immunity; at the same time, inadequate activation, which could lead to autoimmunity, must be avoided. mTOR forms highly conserved protein complexes 1 and 2 that shape lineage specification by integrating signals originating from TCR engagement, co-stimulatory or co-inhibitory receptors and cytokines and availability of nutrients. If one considers autoimmunity as the result of aberrant lineage specification in response to such signals, the importance of this pathway becomes evident; this provides the conceptual basis for mTOR inhibition in the treatment of systemic autoimmunity, such as systemic lupus erythematosus (SLE). Clinical trials in SLE patients have provided preliminary evidence that mTOR blockade by sirolimus (rapamycin) can reverse pro-inflammatory lineage skewing, including the expansion of Th17 and double-negative T cells and plasma cells and the contraction of regulatory T cells. Moreover, sirolimus has shown promising efficacy in the treatment of refractory idiopathic multicentric Castleman disease, newly characterized by systemic autoimmunity due to mTOR overactivation. Alternatively, mTOR blockade enhances responsiveness to vaccination and reduces infections by influenza virus in healthy elderly subjects. Such seemingly contradictory findings highlight the importance to further evaluate the clinical effects of mTOR manipulation, including its potential role in treatment of COVID-19 infection. mTOR blockade may extend healthy lifespan by abrogating inflammation induced by viral infections and autoimmunity. This review provides a mechanistic assessment of mTOR pathway activation in lineage specification within the adaptive and innate immune systems and its role in health and autoimmunity. We then discuss some of the recent experimental and clinical discoveries implicating mTOR in viral pathogensis and aging.


Assuntos
COVID-19 , Lúpus Eritematoso Sistêmico , Idoso , Antivirais/uso terapêutico , Autoimunidade , Humanos , Longevidade , Lúpus Eritematoso Sistêmico/tratamento farmacológico , SARS-CoV-2 , Serina-Treonina Quinases TOR/uso terapêutico
14.
Anim Sci J ; 90(1): 49-54, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30358017

RESUMO

In mammalian preimplantation development, the first cell lineage segregation occurs during the blastocyst stage, when the inner cell mass and trophectoderm (TE) differentiate. Species-specific analyses are essential to elucidate the molecular mechanisms that underlie this process, since they differ between various species. We previously showed that the reciprocal regulation of CCN2 and TEAD4 is required for proper TE differentiation in bovine blastocysts; however, the function of CCN2 during early embryogenesis has remained otherwise elusive. The present study assessed the spatiotemporal expression dynamics of CCN2 in bovine embryos, and evaluated how changes to CCN2 expression (using a CCN2 knockdown (KD) blastocyst model) regulate the expression of pluripotency-related genes such as OCT4 and NANOG. The conducted quantitative PCR analysis revealed that CCN2 mRNA was expressed in bovine oocytes (at the metaphase stage of their second meiosis) and embryos. Similarly, immunostaining detected both cytoplasmic and nuclear CCN2 at all analyzed oocyte and embryonic stages. Finally, both OCT4 and NANOG expression levels were shown to be significantly reduced in CCN2 KD blastocysts. Together, these results demonstrate that bovine CCN2 exhibits unique expression patterns during preimplantation development, and is required for the proper expression of key regulatory genes in bovine blastocysts.


Assuntos
Bovinos/embriologia , Bovinos/genética , Fator de Crescimento do Tecido Conjuntivo/genética , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Expressão Gênica/genética , Animais , Blastocisto/metabolismo , Blastocisto/fisiologia , Fator de Crescimento do Tecido Conjuntivo/fisiologia , Desenvolvimento Embrionário/fisiologia , Feminino , Fertilização in vitro , Técnicas de Maturação in Vitro de Oócitos , Oócitos/metabolismo , Oócitos/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
15.
Curr Top Dev Biol ; 128: 267-294, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29477166

RESUMO

During the first days following fertilization, cells of mammalian embryo gradually lose totipotency, acquiring distinct identity. The first three lineages specified in the mammalian embryo are pluripotent epiblast, which later gives rise to the embryo proper, and two extraembryonic lineages, hypoblast (also known as primitive endoderm) and trophectoderm, which form tissues supporting development of the fetus in utero. Most of our knowledge regarding the mechanisms of early lineage specification in mammals comes from studies in the mouse. However, the growing body of evidence points to both similarities and species-specific differences. Understanding molecular and cellular mechanisms of early embryonic development in nonrodent mammals expands our understanding of basic mechanisms of differentiation and is essential for the development of effective protocols for assisted reproduction in agriculture, veterinary medicine, and for biomedical research. This review summarizes the current state of knowledge on key events in epiblast, hypoblast, and trophoblast differentiation in domestic mammals.


Assuntos
Animais Domésticos/embriologia , Desenvolvimento Embrionário , Animais , Animais Domésticos/genética , Linhagem da Célula , Embrião de Mamíferos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA