Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 256
Filtrar
1.
Nature ; 626(7998): 367-376, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38092041

RESUMEN

Implantation of the human embryo begins a critical developmental stage that comprises profound events including axis formation, gastrulation and the emergence of haematopoietic system1,2. Our mechanistic knowledge of this window of human life remains limited due to restricted access to in vivo samples for both technical and ethical reasons3-5. Stem cell models of human embryo have emerged to help unlock the mysteries of this stage6-16. Here we present a genetically inducible stem cell-derived embryoid model of early post-implantation human embryogenesis that captures the reciprocal codevelopment of embryonic tissue and the extra-embryonic endoderm and mesoderm niche with early haematopoiesis. This model is produced from induced pluripotent stem cells and shows unanticipated self-organizing cellular programmes similar to those that occur in embryogenesis, including the formation of amniotic cavity and bilaminar disc morphologies as well as the generation of an anterior hypoblast pole and posterior domain. The extra-embryonic layer in these embryoids lacks trophoblast and shows advanced multilineage yolk sac tissue-like morphogenesis that harbours a process similar to distinct waves of haematopoiesis, including the emergence of erythroid-, megakaryocyte-, myeloid- and lymphoid-like cells. This model presents an easy-to-use, high-throughput, reproducible and scalable platform to probe multifaceted aspects of human development and blood formation at the early post-implantation stage. It will provide a tractable human-based model for drug testing and disease modelling.


Asunto(s)
Desarrollo Embrionario , Estratos Germinativos , Hematopoyesis , Saco Vitelino , Humanos , Implantación del Embrión , Endodermo/citología , Endodermo/embriología , Estratos Germinativos/citología , Estratos Germinativos/embriología , Saco Vitelino/citología , Saco Vitelino/embriología , Mesodermo/citología , Mesodermo/embriología , Células Madre Pluripotentes Inducidas/citología , Amnios/citología , Amnios/embriología , Cuerpos Embrioides/citología , Linaje de la Célula , Biología Evolutiva/métodos , Biología Evolutiva/tendencias
2.
Nature ; 622(7983): 562-573, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37673118

RESUMEN

The ability to study human post-implantation development remains limited owing to ethical and technical challenges associated with intrauterine development after implantation1. Embryo-like models with spatially organized morphogenesis and structure of all defining embryonic and extra-embryonic tissues of the post-implantation human conceptus (that is, the embryonic disc, the bilaminar disc, the yolk sac, the chorionic sac and the surrounding trophoblast layer) remain lacking1,2. Mouse naive embryonic stem cells have recently been shown to give rise to embryonic and extra-embryonic stem cells capable of self-assembling into post-gastrulation structured stem-cell-based embryo models with spatially organized morphogenesis (called SEMs)3. Here we extend those findings to humans using only genetically unmodified human naive embryonic stem cells (cultured in human enhanced naive stem cell medium conditions)4. Such human fully integrated and complete SEMs recapitulate the organization of nearly all known lineages and compartments of post-implantation human embryos, including the epiblast, the hypoblast, the extra-embryonic mesoderm and the trophoblast layer surrounding the latter compartments. These human complete SEMs demonstrated developmental growth dynamics that resemble key hallmarks of post-implantation stage embryogenesis up to 13-14 days after fertilization (Carnegie stage 6a). These include embryonic disc and bilaminar disc formation, epiblast lumenogenesis, polarized amniogenesis, anterior-posterior symmetry breaking, primordial germ-cell specification, polarized yolk sac with visceral and parietal endoderm formation, extra-embryonic mesoderm expansion that defines a chorionic cavity and a connecting stalk, and a trophoblast-surrounding compartment demonstrating syncytium and lacunae formation. This SEM platform will probably enable the experimental investigation of previously inaccessible windows of human early post implantation up to peri-gastrulation development.


Asunto(s)
Implantación del Embrión , Embrión de Mamíferos , Desarrollo Embrionario , Células Madre Embrionarias Humanas , Humanos , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Fertilización , Gastrulación , Estratos Germinativos/citología , Estratos Germinativos/embriología , Células Madre Embrionarias Humanas/citología , Trofoblastos/citología , Saco Vitelino/citología , Saco Vitelino/embriología , Células Gigantes/citología
3.
Nature ; 622(7983): 574-583, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37369348

RESUMEN

Investigating human development is a substantial scientific challenge due to the technical and ethical limitations of working with embryonic samples. In the face of these difficulties, stem cells have provided an alternative to experimentally model inaccessible stages of human development in vitro1-13. Here we show that human pluripotent stem cells can be triggered to self-organize into three-dimensional structures that recapitulate some key spatiotemporal events of early human post-implantation embryonic development. Our system reproducibly captures spontaneous differentiation and co-development of embryonic epiblast-like and extra-embryonic hypoblast-like lineages, establishes key signalling hubs with secreted modulators and undergoes symmetry breaking-like events. Single-cell transcriptomics confirms differentiation into diverse cell states of the perigastrulating human embryo14,15 without establishing placental cell types, including signatures of post-implantation epiblast, amniotic ectoderm, primitive streak, mesoderm, early extra-embryonic endoderm, as well as initial yolk sac induction. Collectively, our system captures key features of human embryonic development spanning from Carnegie stage16 4-7, offering a reproducible, tractable and scalable experimental platform to understand the basic cellular and molecular mechanisms that underlie human development, including new opportunities to dissect congenital pathologies with high throughput.


Asunto(s)
Linaje de la Célula , Implantación del Embrión , Desarrollo Embrionario , Células Madre Pluripotentes , Femenino , Humanos , Embarazo , Diferenciación Celular , Estratos Germinativos/citología , Estratos Germinativos/enzimología , Células Madre Embrionarias Humanas/citología , Placenta/citología , Células Madre Pluripotentes/citología , Línea Primitiva/citología , Línea Primitiva/embriología , Saco Vitelino/citología , Saco Vitelino/embriología
4.
Dev Biol ; 483: 22-33, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34973175

RESUMEN

The extra-embryonic yolk sac contains adjacent layers of mesoderm and visceral endoderm. The mesodermal layer serves as the first site of embryonic hematopoiesis, while the visceral endoderm provides a means of exchanging nutrients and waste until the development of the chorioallantoic placenta. While defects in chorioallantoic fusion and yolk sac hematopoiesis have been described in Cdx mutant mouse models, little is known about the gene targets and molecular mechanisms through which Cdx members regulate these processes. To this end, we used RNA-seq to examine Cdx-dependent gene expression changes in the yolk sac. We find that loss of Cdx function impacts the expression of genes involved in yolk sac hematopoiesis, as previously described, as well as novel Cdx2 target genes. In addition, we observed Cdx-dependent changes in PRC2 subunit expression accompanied by altered H3K27me3 deposition at a subset of Cdx target genes as early as E7.5 in the embryo proper. This study identifies additional Cdx target genes and provides further evidence for Cdx-dependent epigenetic regulation of gene expression in the early embryo, and that this regulation is required to maintain gene expression programs in the extra-embryonic yolk sac at later developmental stages.


Asunto(s)
Factor de Transcripción CDX2/genética , Desarrollo Embrionario/genética , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Complejo Represivo Polycomb 2/genética , Animales , Endodermo/embriología , Femenino , Expresión Génica , Técnicas de Inactivación de Genes/métodos , Hematopoyesis/genética , Masculino , Mesodermo/embriología , Ratones , Ratones Noqueados , Embarazo , RNA-Seq/métodos , Transcripción Genética/genética , Saco Vitelino/embriología
5.
Genesis ; 59(3): e23412, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33547760

RESUMEN

The atypical PKC (aPKC) subfamily constitutes PKCζ and PKCλ in mice, and both aPKC isoforms have been proposed to be involved in regulating various endothelial cell (EC) functions. However, the physiological function of aPKC in ECs during embryonic development has not been well understood. To address this question, we utilized Tie2-Cre to delete PKCλ alone (PKCλ-SKO) or both PKCλ and PKCζ (DKO) in ECs, and found that all DKO mice died at around the embryonic day 11.5 (E11.5), whereas a small proportion of PKCλ-SKO mice survived till birth. PKCλ-SKO embryos also exhibited less phenotypic severity than DKO embryos at E10.5 and E11.5, suggesting a potential compensatory role of PKCζ for PKCλ in embryonic ECs. We then focused on DKO embryos and investigated the effects of aPKC deficiency on embryonic vascular development. At E9.5, deletion of both aPKC isoforms reduced the diameters of vitelline artery and vein, and decreased branching from both vitelline vessels in yolk sac. Ablation of both aPKC isoforms also disrupted embryonic angiogenesis in head and trunk at the same stage, increasing apoptosis of both ECs and non-ECs. Taken together, our results demonstrated that aPKC in ECs plays an essential role in regulating cell apoptosis, angiogenesis, and embryonic survival.


Asunto(s)
Inductores de la Angiogénesis/metabolismo , Desarrollo Embrionario , Células Endoteliales/metabolismo , Proteína Quinasa C/fisiología , Saco Vitelino/embriología , Saco Vitelino/metabolismo , Animales , Apoptosis , Femenino , Regulación del Desarrollo de la Expresión Génica , Ratones , Embarazo , Eliminación de Secuencia
7.
J Comp Neurol ; 529(2): 340-366, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32415669

RESUMEN

Vascular structures in the developing brain are thought to form via angiogenesis from preformed blood vessels in the cephalic mesenchyme. Immunohistochemical studies of developing mouse brain from E10.5 to E13.5 revealed the presence of avascular blood islands of primitive erythroid cells expressing hemangioblast markers (Flk1, Tal1/Scl1, platelet endothelial cell adhesion molecule 1, vascular endothelial-cadherin, and CD34) and an endothelial marker recognized by Griffonia simplicifolia isolectin B4 (IB4) in the cephalic mesenchyme. These cells formed a perineural vascular plexus from which angiogenic sprouts originated and penetrated the neuroepithelium. In addition, avascular isolated cells expressing primitive erythroid, hemangioblast and endothelial makers were visible in the neuroepithelium where they generated vasculogenic and hemogenic foci. From E10.5 to E13.5, these vasculogenic foci were a source of new blood vessel formation in the developing brain. In vitro, cultured E13.5 brain endothelial cells contained hemogenic endothelial cells capable of generating erythroid cells. Similar cells were present in primary cultures of dissociated cells from E10.5 embryonic head. Our results provide new evidence that the brain vasculature, like that of the yolk sac and the eye choriocapillaris and hyaloid vascular systems, develops at least in part via hemovasculogenesis, a process in which vasculogenesis and hematopoiesis occur simultaneously.


Asunto(s)
Encéfalo/irrigación sanguínea , Encéfalo/embriología , Endotelio Vascular/embriología , Animales , Encéfalo/citología , Endotelio Vascular/citología , Femenino , Ratones , Morfogénesis/fisiología , Embarazo , Saco Vitelino/irrigación sanguínea , Saco Vitelino/citología , Saco Vitelino/embriología
8.
Int. j. morphol ; 38(5): 1412-1420, oct. 2020. graf
Artículo en Inglés | LILACS | ID: biblio-1134457

RESUMEN

SUMMARY: Mesenchymal stem cells are characterized by in vitro high proliferation and multilineage potential maintenance. This study aimed to isolate and characterize equine YS mesenchymal stem cells and compare these with amniotic membranes. The yolk sac (YS) and amniotic membranes (AM) were obtained from 20 pregnant mares with gestational age around 30 days. Cells were cultured in α-MEM supplemented with 15 % FBS, 1 % antibiotic solution, 1 % L-glutamine and 1 % nonessential amino acids. To cell characterization we used cytogenetic analysis, fibroblast colony-forming unit assays, cell growth curves, immunophenotyping, flow cytometry, differentiation assays and teratoma formation. Results: Both cell sources presented fibroblastoid and epithelioid-like format. The YS cells have lower colony formation potential then AM ones, 3 versus 8 colonies per 103 plated cells. However, YS cells grew progressively while AM cells showed steady. Both, the YS and amnion cells immunolabeled for Oct-4, Nanog, SSEA-3, cytokeratin 18, PCNA, and vimentin. In addition, presented mesenchymal, hematopoietic, endothelial and pluripotency markers in flow cytometry. Discussion: Both cell sources presented high plasticity and differed into osteogenic, adipogenic, and chondrogenic lineages, and no tumor formation in nude mice was observed. The results suggest that horse YS may be useful for cell therapy such as amnion-derived cells.


RESUMEN: Las células madre mesenquimales se caracterizan por una alta proliferación in vitro y un mantenimiento potencial de múltiples líneas. Este estudio tuvo como objetivo aislar y caracterizar las células madre mesenquimales del saco vitelino equinas y compararlas con las membranas amnióticas. Se obtuvo el saco vitelino (SV) y las membranas amnióticas (MA) de 20 yeguas preñadas con edad gestacional de aproximadamente 30 días. Las células se cultivaron en α -MEM suplementado con 15 % de FBS, 1 % de solución antibiótica, 1 % de L-glutamina y 1 % de aminoácidos no esenciales. Para la caracterización celular utilizamos análisis citogenéticos, ensayos de unidades de colonias de fibroblastos, curvas de crecimiento celular, inmunofenotipaje, citometría de flujo, ensayos de diferenciación y formación de teratomas. Ambas fuentes celulares presentaron formato fibroblastoideo y epitelioide. Las células SV tienen un potencial de formación de colonias más bajo que las de MA, 3 versus 8 colonias por 103 células en placa. Sin embargo, las células SV crecieron progresivamente mientras que las células MA se mostraron estables. Tanto las células YS como las células amnios están inmunomarcadas para Oct-4, Nanog, SSEA-3, citoqueratina 18, PCNA y vimentina. Además, presentó marcadores mesenquimales, hematopoyéticos, endoteliales y pluripotenciales en citometría de flujo. Ambas fuentes celulares presentaron alta plasticidad y diferían en linajes osteogénicos, adipogénicos y condrogénicos, y no se observó formación de tumores en ratones. Los resultados sugieren que el SV de caballo puede ser útil para la terapia celular, como las células derivadas de amnios.


Asunto(s)
Animales , Saco Vitelino/citología , Células Madre Mesenquimatosas/citología , Caballos , Saco Vitelino/embriología , Técnicas In Vitro , Células Cultivadas , Inmunofenotipificación , Medicina Regenerativa , Desarrollo Embrionario , Citometría de Flujo , Amnios
9.
Gene Expr Patterns ; 38: 119134, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32889095

RESUMEN

NQO1, NAD(P)H: quinone oxidoreductase 1, was first identified in rat and its role has been extensively studied. Even the roles of NQO1 in the maintenance of physiological function and disease were largely addressed, whether the tissue specific functions of the NQO1 in organ development remains unknown. In the current study, we identified two NQO1 isoforms (isoform 1 and isoform 2) and examined the expression of nqo1 variants in adult zebrafish organs and embryos at different stages. In adult organs, RT-PCR result indicated that nqo1 variant 1 was mainly expressed in stomach and intestine, while nqo1 variant 2 was expressed in all organs investigated except for heart. Further, RT-PCR result showed that the nqo1 variant 1 and variant 2 were expressed at all the embryonic stages, but nqo1 variant 1 expression level was much lower than that of nqo1 variant 2. To specifically examine the expression pattern of these two different nqo1 variants, we did whole mount in situ hybridization and the results demonstrated that, both of them were maternally expressed at 8-cell stage, and they were all expressed ubiquitously at early stage. At 24 hpf, nqo1 variant 2 was mainly expressed in yolk cells, and slightly in head and eyes. At 48 hpf, nqo1 variant 2 was restricted in lateral line neuromasts. From 72 hpf to 144 hpf, nqo1 variant 2 was mainly restricted in branchial arch, liver, swimming bladder and lateral line neuromasts, while from 124 hpf to 192 hpf, nqo1 variant 2 only restricted in liver, and disappeared in lateral line neuromasts. On the contrary, at the late embryonic stage, nqo1 variant 1 was only expressed in liver and swimming bladder while not in branchial arch and lateral line neuromasts. In conclusion, we systematically analyzed the expression pattern of nqo1 variant 1 and variant 2 in zebrafish at different embryonic stages, and our data implied the possible role of nqo1 in regulating liver, branchial arch and lateral neuromasts development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , NAD(P)H Deshidrogenasa (Quinona)/genética , Proteínas de Pez Cebra/genética , Sacos Aéreos/embriología , Sacos Aéreos/metabolismo , Animales , Sistema de la Línea Lateral/embriología , Sistema de la Línea Lateral/metabolismo , Hígado/embriología , Hígado/metabolismo , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Saco Vitelino/embriología , Saco Vitelino/metabolismo , Pez Cebra , Proteínas de Pez Cebra/metabolismo
10.
Cell Rep ; 32(6): 108004, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32783932

RESUMEN

During embryogenesis, lymphoid tissue inducer (LTi) cells are essential for lymph node organogenesis. These cells are part of the innate lymphoid cell (ILC) family. Although their earliest embryonic hematopoietic origin is unclear, other innate immune cells have been shown to be derived from early hemogenic endothelium in the yolk sac as well as the aorta-gonad-mesonephros. A proper model to discriminate between these locations was unavailable. In this study, using a Cxcr4-CreERT2 lineage tracing model, we identify a major contribution from embryonic hemogenic endothelium, but not the yolk sac, toward LTi progenitors. Conversely, embryonic LTi cells are replaced by hematopoietic stem cell-derived cells in adults. We further show that, in the fetal liver, common lymphoid progenitors differentiate into highly dynamic alpha-lymphoid precursor cells that, at this embryonic stage, preferentially mature into LTi precursors and establish their functional LTi cell identity only after reaching the periphery.


Asunto(s)
Hemangioblastos/metabolismo , Hematopoyesis/fisiología , Tejido Linfoide/embriología , Receptores CXCR4/metabolismo , Animales , Desarrollo Embrionario/fisiología , Hemangioblastos/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Inmunidad Innata , Hígado/embriología , Linfocitos/metabolismo , Linfocitos T Colaboradores-Inductores/metabolismo , Saco Vitelino/embriología
11.
Nat Commun ; 11(1): 3760, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32724077

RESUMEN

Human embryogenesis is hallmarked by two phases of yolk sac development. The primate hypoblast gives rise to a transient primary yolk sac, which is rapidly superseded by a secondary yolk sac during gastrulation. Moreover, primate embryos form extraembryonic mesoderm prior to gastrulation, in contrast to mouse. The function of the primary yolk sac and the origin of extraembryonic mesoderm remain unclear. Here, we hypothesise that the hypoblast-derived primary yolk sac serves as a source for early extraembryonic mesoderm, which is supplemented with mesoderm from the gastrulating embryo. We discuss the intricate relationship between the yolk sac and the primate embryo and highlight the pivotal role of the yolk sac as a multifunctional hub for haematopoiesis, germ cell development and nutritional supply.


Asunto(s)
Desarrollo Embrionario/fisiología , Mesodermo/embriología , Primates/embriología , Saco Vitelino/embriología , Animales , Diferenciación Celular/fisiología , Células Germinales Embrionarias/fisiología , Hematopoyesis/fisiología
12.
Artículo en Inglés | MEDLINE | ID: mdl-32061751

RESUMEN

Carboxyl ester lipase (Cel), is a lipolytic enzyme secreted by the pancreas, which hydrolyzes various species of lipids in the gut. Cel is also secreted by mammary gland during lactation and exists in breast milk. It facilitates dietary fat digestion and absorption, thus contributing to normal infant development. This study aimed to examine whether the Cel in zebrafish embryos has a similar role of maternal lipid utilization as in human infants, and how Cel contributes to the utilization of yolk lipids in zebrafish. The cel1 and cel2 genes were expressed ubiquitously in the blastodisc and yolk syncytial layer before 24 hpf, and in the exocrine pancreas after 72 hpf. The cel1 and cel2 morphants exhibited developmental retardation and yolk sac retention. The total cholesterol, cholesterol ester, free cholesterol, and triglyceride were reduced in the morphants' body while accumulated in the yolk (except triglyceride). The FFA content of whole embryos was much lower in morphants than in standard controls. Moreover, the delayed development in cel (cel1/cel2) double morphants was partially rescued by FFA and cholesterol supplementation. Delayed and weakened cholesterol ester transport to the brain and eyes was observed in cel morphants. Correspondingly, shrunken midbrain tectum, microphthalmia, pigmentation-delayed eyes as well as down-regulated Shh target genes were observed in the CNS of double morphants. Interestingly, cholesterol injections reversed these CNS alterations. Our findings suggested that cel genes participate in the lipid releasing from yolk sac to developing body, thereby contributing to the normal growth rate and CNS development in zebrafish.


Asunto(s)
Carboxilesterasa/metabolismo , Regulación del Desarrollo de la Expresión Génica , Trastornos del Crecimiento/genética , Saco Vitelino/enzimología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Carboxilesterasa/genética , Sistema Nervioso Central/embriología , Colesterol/metabolismo , Ésteres del Colesterol/metabolismo , Modelos Animales de Enfermedad , Embrión no Mamífero , Desarrollo Embrionario , Técnicas de Silenciamiento del Gen , Trastornos del Crecimiento/embriología , Trastornos del Crecimiento/enzimología , Proteínas Hedgehog/metabolismo , Humanos , Metabolismo de los Lípidos , Morfolinos/administración & dosificación , Morfolinos/genética , Páncreas Exocrino/embriología , Páncreas Exocrino/enzimología , Triglicéridos/metabolismo , Saco Vitelino/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
13.
EMBO J ; 39(3): e103205, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31894879

RESUMEN

Tissue-resident macrophages (MΦTR ) originate from at least two distinct waves of erythro-myeloid progenitors (EMP) arising in the yolk sac (YS) at E7.5 and E8.5 with the latter going through a liver monocyte intermediate. The relative potential of these precursors in determining development and functional capacity of MΦTR remains unclear. Here, we studied development of alveolar macrophages (AM) after single and competitive transplantation of different precursors from YS, fetal liver, and fetal lung into neonatal Csf2ra-/- mice, which lack endogenous AM. Fetal monocytes, promoted by Myb, outcompeted primitive MΦ (pMΦ) in empty AM niches and preferentially developed to mature AM, which is associated with enhanced mitochondrial respiratory and glycolytic capacity and repression of the transcription factors c-Maf and MafB. Interestingly, AM derived from pMΦ failed to efficiently clear alveolar proteinosis and protect from fatal lung failure following influenza virus infection. Thus, our data demonstrate superior developmental and functional capacity of fetal monocytes over pMΦ in AM development and underlying mechanisms explaining replacement of pMΦ in fetal tissues.


Asunto(s)
Hígado/embriología , Pulmón/embriología , Monocitos/citología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Saco Vitelino/embriología , Animales , Células Cultivadas , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Glucólisis , Hígado/citología , Hígado/metabolismo , Pulmón/citología , Pulmón/metabolismo , Macrófagos Alveolares , Factor de Transcripción MafB/metabolismo , Ratones , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Proteínas Proto-Oncogénicas c-maf/metabolismo , Proteínas Proto-Oncogénicas c-myb/farmacología , Saco Vitelino/citología , Saco Vitelino/metabolismo
15.
Nature ; 577(7791): 537-542, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31830756

RESUMEN

Our understanding of how human embryos develop before gastrulation, including spatial self-organization and cell type ontogeny, remains limited by available two-dimensional technological platforms1,2 that do not recapitulate the in vivo conditions3-5. Here we report a three-dimensional (3D) blastocyst-culture system that enables human blastocyst development up to the primitive streak anlage stage. These 3D embryos mimic developmental landmarks and 3D architectures in vivo, including the embryonic disc, amnion, basement membrane, primary and primate unique secondary yolk sac, formation of anterior-posterior polarity and primitive streak anlage. Using single-cell transcriptome profiling, we delineate ontology and regulatory networks that underlie the segregation of epiblast, primitive endoderm and trophoblast. Compared with epiblasts, the amniotic epithelium shows unique and characteristic phenotypes. After implantation, specific pathways and transcription factors trigger the differentiation of cytotrophoblasts, extravillous cytotrophoblasts and syncytiotrophoblasts. Epiblasts undergo a transition to pluripotency upon implantation, and the transcriptome of these cells is maintained until the generation of the primitive streak anlage. These developmental processes are driven by different pluripotency factors. Together, findings from our 3D-culture approach help to determine the molecular and morphogenetic developmental landscape that occurs during human embryogenesis.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Desarrollo Embrionario , Línea Primitiva/citología , Línea Primitiva/embriología , Amnios/citología , Amnios/embriología , Blastocisto/citología , Diferenciación Celular , Linaje de la Célula , Polaridad Celular , Colágeno , Combinación de Medicamentos , Epitelio/embriología , Gastrulación , Estratos Germinativos/citología , Estratos Germinativos/embriología , Humanos , Laminina , Proteoglicanos , RNA-Seq , Análisis de la Célula Individual , Factores de Transcripción/metabolismo , Transcriptoma , Trofoblastos/citología , Saco Vitelino/citología , Saco Vitelino/embriología
16.
Evol Dev ; 21(6): 342-353, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31545014

RESUMEN

The reptilian clade Squamata is defined primarily by osteological synapomorphies, few of which are entirely unambiguous. Studies of developing squamate eggs have revealed a uniquely specialized feature not known to occur in any other amniotes. This feature-the yolk cleft/isolated yolk mass complex-lines the ventral hemisphere of the egg. During its formation, extraembryonic mesoderm penetrates the yolk and an exocoelom (the yolk cleft [YC]) forms in association with it, cutting off a thin segment of yolk (the "isolated yolk mass" [IYM]) from the main body of the yolk. The YC-IYM complex has been observed and described in more than 65 squamate species in 12 families. In viviparous species, it contributes to the "omphaloplacenta," a type of yolk sac placenta unique to squamates. The only squamates known to lack the IYM are a few highly placentotrophic skinks with minuscule eggs, viviparous species in which it clearly has been lost. Given its absence in mammals, chelonians, crocodylians, and birds, the YC-IYM complex warrants recognition as a developmental synapomorphy of the squamate clade. As in extant viviparous lizards and snakes, the YC-IYM complex presumably contributed to the placenta of extinct viviparous squamates.


Asunto(s)
Lagartos/embriología , Mesodermo/embriología , Saco Vitelino/embriología , Animales
17.
Nat Commun ; 10(1): 953, 2019 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-30814516

RESUMEN

Although hundreds of cytosolic or transmembrane molecules form the primary cilium, few secreted molecules are known to contribute to ciliogenesis. Here, homologous secreted metalloproteases ADAMTS9 and ADAMTS20 are identified as ciliogenesis regulators that act intracellularly. Secreted and furin-processed ADAMTS9 bound heparan sulfate and was internalized by LRP1, LRP2 and clathrin-mediated endocytosis to be gathered in Rab11 vesicles with a unique periciliary localization defined by super-resolution microscopy. CRISPR-Cas9 inactivation of ADAMTS9 impaired ciliogenesis in RPE-1 cells, which was restored by catalytically active ADAMTS9 or ADAMTS20 acting in trans, but not by their proteolytically inactive mutants. Their mutagenesis in mice impaired neural and yolk sac ciliogenesis, leading to morphogenetic anomalies resulting from impaired hedgehog signaling, which is transduced by primary cilia. In addition to their cognate extracellular proteolytic activity, ADAMTS9 and ADAMTS20 thus have an additional proteolytic role intracellularly, revealing an unexpected regulatory dimension in ciliogenesis.


Asunto(s)
Proteínas ADAMTS/metabolismo , Proteína ADAMTS9/metabolismo , Cilios/metabolismo , Cilios/ultraestructura , Proteínas ADAMTS/deficiencia , Proteínas ADAMTS/genética , Proteína ADAMTS9/deficiencia , Proteína ADAMTS9/genética , Animales , Línea Celular , Endocitosis , Técnicas de Inactivación de Genes , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Microscopía Electrónica de Rastreo , Modelos Biológicos , Mutación , Defectos del Tubo Neural/embriología , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , Proteolisis , Transducción de Señal , Versicanos/genética , Versicanos/metabolismo , Saco Vitelino/embriología , Saco Vitelino/metabolismo
18.
J Fish Biol ; 95(1): 214-221, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30370922

RESUMEN

Colour vision is mediated by the expression of different visual pigments in photoreceptors of the vertebrate retina. Each visual pigment is a complex of a protein (opsin) and a vitamin A chromophore; alterations to either component affects visual pigment absorbance and, potentially, the visual capabilities of an animal. Many species of fish undergo changes in opsin expression during retinal development. In the case of salmonid fishes the single cone photoreceptors undergo a switch in opsin expression from SWS1 (ultraviolet sensitive) to SWS2 (blue-light sensitive) starting at the yolk-sac alevin stage, around the time when they first experience light. Whether light may initiate this event or produce a plastic response in the various photoreceptors is unknown. In this study, Chinook salmon Oncorhynchus tshawytscha were exposed to light from the embryonic (5 days prior to hatching) into the yolk sac alevin (25 days post hatching) stage and the spectral phenotype of photoreceptors assessed with respect to that of unexposed controls by in situ hybridization with opsin riboprobes. Light exposure did not change the spectral phenotype of photoreceptors, their overall morphology or spatial arrangement. These results concur with those from a variety of fish species and suggest that plasticity in photoreceptor spectral phenotype via changes in opsin expression may not be a widespread occurrence among teleosts.


Asunto(s)
Desarrollo Embrionario/efectos de la radiación , Luz , Opsinas/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Salmón/embriología , Saco Vitelino/efectos de la radiación , Animales , Hibridación in Situ , Fenotipo , Retina/embriología , Salmón/metabolismo , Saco Vitelino/embriología , Saco Vitelino/metabolismo
19.
J Morphol ; 280(1): 35-49, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30478960

RESUMEN

The evolution of viviparity alters the physical relationship between mothers and offspring and the prevalence of viviparity among squamate reptiles presents an opportunity to uncover patterns in the evolution of placental structure. Understanding the breadth of this diversity is limited because studies of placental structure and function have emphasized a limited number of lineages. We studied placental ontogeny using light microscopy for an embryological series of the Mexican gerrhonotine lizard, Mesaspis viridiflava. This species develops an elaborate yolk sac placenta, an omphaloplacenta, which receives vascular support arising in a structure known only from other gerrhonotine lizards. A prominent feature of the omphaloplacenta is a zone of uterine and embryonic epithelial cell hyperplasia located at the upper shoulder of the yolk mass, often extending above the yolk mass. The omphaloplacenta covers more than one-half of the surface area of maternal-embryonic contact. The chorioallantoic placenta has a more restricted distribution because the allantois remains in the embryonic hemisphere of the egg throughout development and lies internal to the vascular support for the omphaloplacenta in areas where they overlap. The structural profile of the chorioallantoic placenta indicates a potential for respiratory exchange and/or hemotrophic nutritive transport, while that of the omphaloplacenta suggests that nutritive transfer is primarily via histotrophy. An eggshell is present in the earliest embryonic stages examined but regresses relatively early in development. Placental specializations of this species are consistent with a pattern of matrotrophic embryonic nutrition and have evolved in a unique lineage specific developmental pattern.


Asunto(s)
Lagartos/anatomía & histología , Placenta/anatomía & histología , Animales , Evolución Biológica , Tamaño Corporal , Embrión no Mamífero/anatomía & histología , Femenino , Fertilidad , Lagartos/embriología , México , Embarazo , Saco Vitelino/anatomía & histología , Saco Vitelino/embriología
20.
Med Ultrason ; 20(4): 487-492, 2018 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-30534657

RESUMEN

AIMS: Embryonic demise is a frequent complication of the first trimester pregnancy. The purpose of this study was to evaluate the correlation between a serum biomarker, the soluble form of the vasculo-endothelial growth factor (sFlt-1) and the distance between the yolk sac (YS) and embryo (DYSE), determined by ultrasonography. MATERIAL AND METHODS: The study was a prospective case-control study that included 2 groups of patients - the control group with 81 first-trimester pregnancies in evolution and the case group with 89 first-trimester pregnancies with a potentially reserved evolutivity. RESULTS: A correlation between the serum level of sFlt-1 and DYSE in embryos with crown-rump length (CRL) greater than 5 mm was identified, showing that a DYSE ≤3 mm correlates with a low level of sFlt-1 (p<0.05) and a DYSE> 4 mm correlates with an increased level of sFlt-1 (p<0.05). CONCLUSIONS: A low level of sFlt-1 associated with a distance between the embryo and yolk sac of small dimensions, respectively <3 mm, correlates with an increased rate of non-viable embryos. This correlation between an ultrasound and a serum parameter is of great value and brings important information about the viability of firsttrimester pregnancies.


Asunto(s)
Desarrollo Embrionario/fisiología , Ultrasonografía Prenatal/métodos , Factor A de Crecimiento Endotelial Vascular/sangre , Saco Vitelino/anatomía & histología , Adulto , Biomarcadores/sangre , Estudios de Casos y Controles , Femenino , Edad Gestacional , Humanos , Embarazo , Primer Trimestre del Embarazo , Estudios Prospectivos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Saco Vitelino/diagnóstico por imagen , Saco Vitelino/embriología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA