Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Intervalo de año de publicación
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.
bioRxiv ; 2023 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-37398391

RESUMEN

Implantation of the human embryo commences a critical developmental stage that comprises profound morphogenetic alteration of embryonic and extra-embryonic tissues, axis formation, and gastrulation events. Our mechanistic knowledge of this window of human life remains limited due to restricted access to in vivo samples for both technical and ethical reasons. Additionally, human stem cell models of early post-implantation development with both embryonic and extra-embryonic tissue morphogenesis are lacking. Here, we present iDiscoid, produced from human induced pluripotent stem cells via an engineered a synthetic gene circuit. iDiscoids exhibit reciprocal co-development of human embryonic tissue and engineered extra-embryonic niche in a model of human post-implantation. They exhibit unanticipated self-organization and tissue boundary formation that recapitulates yolk sac-like tissue specification with extra-embryonic mesoderm and hematopoietic characteristics, the formation of bilaminar disc-like embryonic morphology, the development of an amniotic-like cavity, and acquisition of an anterior-like hypoblast pole and posterior-like axis. iDiscoids offer an easy-to-use, high-throughput, reproducible, and scalable platform to probe multifaceted aspects of human early post-implantation development. Thus, they have the potential to provide a tractable human model for drug testing, developmental toxicology, and disease modeling.

3.
bioRxiv ; 2023 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-37333168

RESUMEN

Gastrulation is considered the sine qua non of embryogenesis, establishing a multidimensional structure and the spatial coordinates upon which all later developmental events transpire. At this time, the embryo adopts a heavy reliance on glucose metabolism to support rapidly accelerating changes in morphology, proliferation, and differentiation. However, it is currently unknown how this conserved metabolic shift maps onto the three-dimensional landscape of the growing embryo and whether it is spatially linked to the orchestrated cellular and molecular processes necessary for gastrulation. Here we identify that glucose is utilised during mouse gastrulation via distinct metabolic pathways to instruct local and global embryonic morphogenesis, in a cell type and stage-specific manner. Through detailed mechanistic studies and quantitative live imaging of mouse embryos, in parallel with tractable in vitro stem cell differentiation models and embryo-derived tissue explants, we discover that cell fate acquisition and the epithelial-to-mesenchymal transition (EMT) relies on the Hexosamine Biosynthetic Pathway (HBP) branch of glucose metabolism, while newly-formed mesoderm requires glycolysis for correct migration and lateral expansion. This regional and tissue-specific difference in glucose metabolism is coordinated with Fibroblast Growth Factor (FGF) activity, demonstrating that reciprocal crosstalk between metabolism and growth factor signalling is a prerequisite for gastrulation progression. We expect these studies to provide important insights into the function of metabolism in other developmental contexts and may help uncover mechanisms that underpin embryonic lethality, cancer, and congenital disease.

4.
Elife ; 102021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34569938

RESUMEN

Apico-basal polarization of cells within the embryo is critical for the segregation of distinct lineages during mammalian development. Polarized cells become the trophectoderm (TE), which forms the placenta, and apolar cells become the inner cell mass (ICM), the founding population of the fetus. The cellular and molecular mechanisms leading to polarization of the human embryo and its timing during embryogenesis have remained unknown. Here, we show that human embryo polarization occurs in two steps: it begins with the apical enrichment of F-actin and is followed by the apical accumulation of the PAR complex. This two-step polarization process leads to the formation of an apical domain at the 8-16 cell stage. Using RNA interference, we show that apical domain formation requires Phospholipase C (PLC) signaling, specifically the enzymes PLCB1 and PLCE1, from the eight-cell stage onwards. Finally, we show that although expression of the critical TE differentiation marker GATA3 can be initiated independently of embryo polarization, downregulation of PLCB1 and PLCE1 decreases GATA3 expression through a reduction in the number of polarized cells. Therefore, apical domain formation reinforces a TE fate. The results we present here demonstrate how polarization is triggered to regulate the first lineage segregation in human embryos.


Asunto(s)
Tipificación del Cuerpo , Diferenciación Celular , Linaje de la Célula , Polaridad Celular , Embrión de Mamíferos/enzimología , Actinas/metabolismo , Adulto , Técnicas de Cultivo de Embriones , Femenino , Factor de Transcripción GATA3/metabolismo , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Humanos , Fosfoinositido Fosfolipasa C , Fosfolipasa C beta , Embarazo , Transducción de Señal , Factores de Tiempo , Adulto Joven
5.
Nat Commun ; 12(1): 5550, 2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34548496

RESUMEN

Understanding human development is of fundamental biological and clinical importance. Despite its significance, mechanisms behind human embryogenesis remain largely unknown. Here, we attempt to model human early embryo development with expanded pluripotent stem cells (EPSCs) in 3-dimensions. We define a protocol that allows us to generate self-organizing cystic structures from human EPSCs that display some hallmarks of human early embryogenesis. These structures mimic polarization and cavitation characteristic of pre-implantation development leading to blastocyst morphology formation and the transition to post-implantation-like organization upon extended culture. Single-cell RNA sequencing of these structures reveals subsets of cells bearing some resemblance to epiblast, hypoblast and trophectoderm lineages. Nevertheless, significant divergences from natural blastocysts persist in some key markers, and signalling pathways point towards ways in which morphology and transcriptional-level cell identities may diverge in stem cell models of the embryo. Thus, this stem cell platform provides insights into the design of stem cell models of embryogenesis.


Asunto(s)
Blastocisto/citología , Técnicas de Cultivo de Célula , Embrión de Mamíferos/citología , Desarrollo Embrionario/genética , Modelos Biológicos , Células Madre Pluripotentes/citología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Biomarcadores/metabolismo , Blastocisto/metabolismo , Linaje de la Célula/genética , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/metabolismo , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/metabolismo , Expresión Génica , Humanos , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , Células Madre Pluripotentes/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/metabolismo , Análisis de Secuencia de ARN , Análisis de la Célula Individual
6.
J Assist Reprod Genet ; 36(10): 2121-2133, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31396850

RESUMEN

PURPOSE: The aim of the present study is to investigate role of FoxO transcription factors in preimplantation embryo development by knocking down FoxO1, FoxO3, and FoxO4 genes and also to assess cell cycle arrest related proteins, p53 and p21, and apoptosis-related proteins, fas ligand (FASL), and cleaved caspase 3. METHODS: Knockdown of FoxOs using siRNA was confirmed utilizing RT-PCR and qRT-PCR in gene level and using immunofluorescence in protein level. Following knockdown of FoxO1, FoxO3, and FoxO4 in two-cell mouse embryos with or without resveratrol treatment; developmental competence of embryos and expression patterns of SIRT1, p53, p21, FASL, and CLEAVED CASPASE 3 proteins in embryos by immunofluorescence were assessed after 48 h. ROS levels were measured in knockdown embryos. Terminal deoxynucleotidyl transferase dUTP nick end labeling assay was used to determine resveratrol dose. RESULTS: Successful knockdown of FoxO genes in mouse embryos utilizing a non-invasive siRNA method was achieved. Significantly, knockdown of FoxO genes impaired preimplantation embryo development which cannot be prevented by resveratrol treatment. Immunofluorescence results showed that resveratrol could protect embryos from cell cycle arrest and apoptosis. FOXO proteins regulate apoptosis and cell cycle related proteins in mouse preimplantation embryos. Moreover, there might be an autofeedback mechanism where FOXO1, FOXO3, and FOXO4 regulate SIRT1 protein expression. CONCLUSIONS: These results suggest that FOXO transcription factors could contribute to mouse preimplantation embryo development, and it remains to investigate whether they have crucial roles in human preimplantation embryo and infertility.


Asunto(s)
Proteínas de Ciclo Celular/genética , Desarrollo Embrionario/genética , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O3/genética , Factores de Transcripción Forkhead/genética , Animales , Apoptosis/genética , Blastocisto/metabolismo , Puntos de Control del Ciclo Celular/genética , Proteína Ligando Fas/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Ratones , Embarazo , Sirtuina 1/genética , Proteína p53 Supresora de Tumor/genética , Quinasas p21 Activadas/genética
7.
J Assist Reprod Genet ; 33(3): 335-348, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26843391

RESUMEN

PURPOSE: Azoospermia is one of the major causes of male infertility and is basically classified into obstructive (OA) and non-obstructive azoospermia (NOA). The molecular background of NOA still largely remains elusive. It has been shown that the poly(A)-binding proteins (PABPs) essentially play critical roles in stabilization and translational control of the mRNAs during spermatogenesis. METHODS: In the present study, we aim to evaluate expression levels of the PABP genes, EPAB, PABPC1, and PABPC3, in the testicular biopsy samples and in the isolated spermatocyte (SC) and round spermatid (RS) fractions obtained from men with various types of NOA including hypospermatogenesis (hyposperm), RS arrest, SC arrest, and Sertoli cell-only syndrome (SCO). RESULTS: In the testicular biopsy samples, both PABPC1 and PABPC3 mRNA expressions were gradually decreased from hyposperm to SCO groups (P < 0.05), whereas there was no remarkable difference for the EPAB expression among groups. The expression levels of cytoplasmically localized PABPC1 and PABPC3 proteins dramatically reduced from hyposperm to SCO groups (P < 0.05). In the isolated SC and RS fractions, the EPAB, PABPC1, and PABPC3 mRNA expressions were gradually decreased from hyposperm to SC arrest groups (P < 0.05). Similarly, both PABPC1 and PABPC3 proteins were expressed at higher levels in the SC and RS fractions from hyposperm group when compared to the SC and RS fractions from either RS arrest or SC arrest group (P < 0.05). CONCLUSION: Our findings suggest that observed significant alterations in the PABPs expression may have an implication for development of different NOA forms.


Asunto(s)
Azoospermia/genética , Proteína I de Unión a Poli(A)/genética , Proteínas de Unión a Poli(A)/genética , Testículo/fisiología , Adulto , Anciano , Biopsia , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Oligospermia/genética , Oligospermia/patología , Proteína I de Unión a Poli(A)/metabolismo , Proteínas de Unión a Poli(A)/metabolismo , Síndrome de Sólo Células de Sertoli/genética , Síndrome de Sólo Células de Sertoli/patología , Testículo/fisiopatología
8.
Mol Hum Reprod ; 20(1): 15-30, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23928157

RESUMEN

Telomeres are located at the ends of all eukaryotic chromosomes and protect them from deleterious events such as inappropriate DNA repair, illegitimate recombination or improper segregation of the chromosomes during mitotic or meiotic divisions. However, telomeres gradually shorten primarily due to successive rounds of genomic DNA replication and also as the result of the adverse effects of oxidative stress, genotoxic agents, diseases related to ageing and environmental factors on the nuclear materials of dividing or non-dividing cells. Germline cells, proliferative granulosa cells, early embryos, stem cells, highly proliferative somatic cells and many cancer cells contain the enzyme telomerase so that they are capable of elongating the shortened telomeres. Although numerous studies have revealed the length of telomeres and telomerase activity in oocytes, granulosa cells and early embryos, only a few studies have analyzed and compared the work performed on distinct mammalian species. In this comprehensive review article, we compare and discuss telomere length and telomerase activity in oocytes, granulosa cells and early embryos in different mammalian species including mice, bovines and humans.


Asunto(s)
Desarrollo Embrionario , Células de la Granulosa/citología , Oocitos/crecimiento & desarrollo , Telomerasa/metabolismo , Homeostasis del Telómero/genética , Envejecimiento , Animales , Bovinos , Femenino , Humanos , Ratones , Oocitos/citología , Estrés Oxidativo , Telómero/genética , Telómero/metabolismo , Acortamiento del Telómero/genética , Proteínas de Unión a Telómeros
9.
Clin Neurol Neurosurg ; 115(4): 438-44, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22776801

RESUMEN

OBJECTIVE: Investigation of the structure of vascular malformations highlights the pathogenic mechanisms underlying their clinical behavior. One of the vascular malformations is called cerebral cavernous malformation (CCM). However, the ultrastructural features of the vascular malformations are not defined in detail. METHODS: We aimed to investigate the ultrastructural features of CCMs using transmission (TEM), scanning (SEM) electron microscopy, and also immunohistochemistry methods with antibodies against CCM proteins such as CCM2 and CCM3. CCM tissues (n=6) microsurgically excised from patients for conventional indications. RESULTS: CCM2 and CCM3 were strongly detected in the vascular endothelium. However, there was a very weak immunostaining in stroma. SEM observations revealed that there were ruptures and damages in the luminal endothelium, possibly due to the damage of intercellular junctions. TEM observations also showed a few ruptures and detachments between the endothelium and basal lamina as observed with partially damages and disconnections. The architecture of pericytes showed protrusions and shrinkages. Our results suggest that the thin vessel walls of CCMs were lacking of subendothelial support and intact basal lamina underlying the endothelial cells. CONCLUSION: This study is so far the first study attempting to show human CCM lesions with SEM. We believe that an understanding of the ultrastructural features of these lesions by light and electron microscopy techniques would help to understand the pathology of these diseases.


Asunto(s)
Vasos Sanguíneos/patología , Vasos Sanguíneos/ultraestructura , Encéfalo/ultraestructura , Malformaciones Arteriovenosas Intracraneales/patología , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/inmunología , Encéfalo/patología , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Circulación Cerebrovascular , Mapeo Cromosómico , Endotelio Vascular/patología , Endotelio Vascular/ultraestructura , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Malformaciones Arteriovenosas Intracraneales/genética , Malformaciones Arteriovenosas Intracraneales/fisiopatología , Malformaciones Arteriovenosas Intracraneales/cirugía , Proteína KRIT1 , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/inmunología , Neuroglía/patología , Neuroglía/ultraestructura , Pericitos/patología , Pericitos/ultraestructura , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/inmunología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/inmunología , Células del Estroma/patología , Células del Estroma/ultraestructura
10.
Int J Dev Neurosci ; 29(5): 509-14, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21569831

RESUMEN

Cerebral cavernous malformation (CCM) is one of the most common types of vascular malformations of the central nervous system, affecting nearly one in 200 people. CCM lesions are characterized by grossly dilated vascular channels lined by a single layer of endothelium. Genetic linkage analyses have mapped three CCM loci to CCM1, CCM2 and CCM3. All three causative genes have now been identified allowing new insights into CCM pathophysiology. We focused on the CCM2 protein that might take place in blood vessel formation; we report here the expression patterns of CCM2 in prenatal development and adult human neocortex by means of immunohistochemistry and Western blot analysis. CCM2 was obviously detected in vascular endothelium and neuroglial precursor cells during development and also observed in arterial endothelium, neurons, some of the glial cells in adult neocortex. The expression patterns suggest that it could be one of the arterial markers whether this is a cause or a consequence of an altered vascular identity. CCM2 might play a role during vasculogenesis and angiogenesis during human brain development. Furthermore, with this study, CCM2 have been described for the first time in developing human neocortex.


Asunto(s)
Proteínas Portadoras/metabolismo , Neocórtex/embriología , Neocórtex/crecimiento & desarrollo , Neocórtex/metabolismo , Adulto , Proteínas Portadoras/genética , Circulación Cerebrovascular/fisiología , Hemangioma Cavernoso del Sistema Nervioso Central/genética , Hemangioma Cavernoso del Sistema Nervioso Central/patología , Hemangioma Cavernoso del Sistema Nervioso Central/fisiopatología , Humanos , Neocórtex/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA