Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 186(17): 3548-3557, 2023 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-37595564

RESUMEN

A human embryo's legal definition and its entitlement to protection vary greatly worldwide. Recently, human pluripotent stem cells have been used to form in vitro models of early embryos that have challenged legal definitions and raised questions regarding their usage. In this light, we propose a refined legal definition of an embryo, suggest "tipping points" for when human embryo models could eventually be afforded similar protection to that of embryos, and then revisit basic ethical principles that might help to draft a roadmap for the gradual, justified usage of embryo models in a manner that aims to maximize benefits to society.


Asunto(s)
Investigaciones con Embriones , Embrión de Mamíferos , Humanos , Células Madre Pluripotentes , Investigaciones con Embriones/ética
2.
Nature ; 626(7998): 357-366, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38052228

RESUMEN

Recently, several studies using cultures of human embryos together with single-cell RNA-seq analyses have revealed differences between humans and mice, necessitating the study of human embryos1-8. Despite the importance of human embryology, ethical and legal restrictions have limited post-implantation-stage studies. Thus, recent efforts have focused on developing in vitro self-organizing models using human stem cells9-17. Here, we report genetic and non-genetic approaches to generate authentic hypoblast cells (naive hPSC-derived hypoblast-like cells (nHyCs))-known to give rise to one of the two extraembryonic tissues essential for embryonic development-from naive human pluripotent stem cells (hPSCs). Our nHyCs spontaneously assemble with naive hPSCs to form a three-dimensional bilaminar structure (bilaminoids) with a pro-amniotic-like cavity. In the presence of additional naive hPSC-derived analogues of the second extraembryonic tissue, the trophectoderm, the efficiency of bilaminoid formation increases from 20% to 40%, and the epiblast within the bilaminoids continues to develop in response to trophectoderm-secreted IL-6. Furthermore, we show that bilaminoids robustly recapitulate the patterning of the anterior-posterior axis and the formation of cells reflecting the pregastrula stage, the emergence of which can be shaped by genetically manipulating the DKK1/OTX2 hypoblast-like domain. We have therefore successfully modelled and identified the mechanisms by which the two extraembryonic tissues efficiently guide the stage-specific growth and progression of the epiblast as it establishes the post-implantation landmarks of human embryogenesis.


Asunto(s)
Desarrollo Embrionario , Estratos Germinativos , Células Madre Pluripotentes , Humanos , Diferenciación Celular , Implantación del Embrión , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/genética , Desarrollo Embrionario/fisiología , Estratos Germinativos/citología , Estratos Germinativos/embriología , Estratos Germinativos/metabolismo , Células Madre Pluripotentes/citología , Interleucina-6/metabolismo , Gástrula/citología , Gástrula/embriología , Amnios/citología , Amnios/embriología , Amnios/metabolismo , Ectodermo/citología , Ectodermo/embriología , Ectodermo/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Factores de Transcripción Otx/genética , Factores de Transcripción Otx/metabolismo
3.
Nature ; 601(7894): 600-605, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34856602

RESUMEN

One week after fertilization, human embryos implant into the uterus. This event requires the embryo to form a blastocyst consisting of a sphere encircling a cavity lodging the embryo proper. Stem cells can form a blastocyst model that we called a blastoid1. Here we show that naive human pluripotent stem cells cultured in PXGL medium2 and triply inhibited for the Hippo, TGF-ß and ERK pathways efficiently (with more than 70% efficiency) form blastoids generating blastocyst-stage analogues of the three founding lineages (more than 97% trophectoderm, epiblast and primitive endoderm) according to the sequence and timing of blastocyst development. Blastoids spontaneously form the first axis, and we observe that the epiblast induces the local maturation of the polar trophectoderm, thereby endowing blastoids with the capacity to directionally attach to hormonally stimulated endometrial cells, as during implantation. Thus, we propose that such a human blastoid is a faithful, scalable and ethical model for investigating human implantation and development3,4.


Asunto(s)
Blastocisto , Células Madre Pluripotentes , Blastocisto/metabolismo , Diferenciación Celular , Linaje de la Célula , Implantación del Embrión , Desarrollo Embrionario , Femenino , Humanos
4.
Genome Res ; 2022 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-35948369

RESUMEN

The preconceptual, intrauterine, and early life environments can have a profound and long-lasting impact on the developmental trajectories and health outcomes of the offspring. Given the relatively low success rates of assisted reproductive technologies (ART; ∼25%), additives and adjuvants, such as glucocorticoids, are used to improve the success rate. Considering the dynamic developmental events that occur during this window, these exposures may alter blastocyst formation at a molecular level, and as such, affect not only the viability of the embryo and the ability of the blastocyst to implant, but also the developmental trajectory of the first three cell lineages, ultimately influencing the physiology of the embryo. In this study, we present a comprehensive single-cell transcriptome, methylome, and small RNA atlas in the day 7 human embryo. We show that, despite no change in morphology and developmental features, preimplantation glucocorticoid exposure reprograms the molecular profile of the TE lineage, and these changes are associated with an altered metabolic and inflammatory response. Our data also suggest that glucocorticoids can precociously mature the TE sublineages, supported by the presence of extravillous trophoblast markers in the polar sublineage and presence of X Chromosome dosage compensation. Further, we have elucidated that epigenetic regulation-DNA methylation and microRNAs (miRNAs)-likely underlies the transcriptional changes observed. This study suggests that exposures to exogenous compounds during preimplantation may unintentionally reprogram the human embryo, possibly leading to suboptimal development and longer-term health outcomes.

5.
Development ; 149(20)2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-35993866

RESUMEN

Embryogenesis is supported by dynamic loops of cellular interactions. Here, we create a partial mouse embryo model to elucidate the principles of epiblast (Epi) and extra-embryonic endoderm co-development (XEn). We trigger naive mouse embryonic stem cells to form a blastocyst-stage niche of Epi-like cells and XEn-like cells (3D, hydrogel free and serum free). Once established, these two lineages autonomously progress in minimal medium to form an inner pro-amniotic-like cavity surrounded by polarized Epi-like cells covered with visceral endoderm (VE)-like cells. The progression occurs through reciprocal inductions by which the Epi supports the primitive endoderm (PrE) to produce a basal lamina that subsequently regulates Epi polarization and/or cavitation, which, in return, channels the transcriptomic progression to VE. This VE then contributes to Epi bifurcation into anterior- and posterior-like states. Similarly, boosting the formation of PrE-like cells within blastoids supports developmental progression. We argue that self-organization can arise from lineage bifurcation followed by a pendulum of induction that propagates over time.


Asunto(s)
Endodermo , Estratos Germinativos , Animales , Blastocisto , Diferenciación Celular , Linaje de la Célula/fisiología , Implantación del Embrión , Embrión de Mamíferos , Ratones
7.
Nature ; 557(7703): 106-111, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29720634

RESUMEN

The blastocyst (the early mammalian embryo) forms all embryonic and extra-embryonic tissues, including the placenta. It consists of a spherical thin-walled layer, known as the trophectoderm, that surrounds a fluid-filled cavity sheltering the embryonic cells 1 . From mouse blastocysts, it is possible to derive both trophoblast 2 and embryonic stem-cell lines 3 , which are in vitro analogues of the trophectoderm and embryonic compartments, respectively. Here we report that trophoblast and embryonic stem cells cooperate in vitro to form structures that morphologically and transcriptionally resemble embryonic day 3.5 blastocysts, termed blastoids. Like blastocysts, blastoids form from inductive signals that originate from the inner embryonic cells and drive the development of the outer trophectoderm. The nature and function of these signals have been largely unexplored. Genetically and physically uncoupling the embryonic and trophectoderm compartments, along with single-cell transcriptomics, reveals the extensive inventory of embryonic inductions. We specifically show that the embryonic cells maintain trophoblast proliferation and self-renewal, while fine-tuning trophoblast epithelial morphogenesis in part via a BMP4/Nodal-KLF6 axis. Although blastoids do not support the development of bona fide embryos, we demonstrate that embryonic inductions are crucial to form a trophectoderm state that robustly implants and triggers decidualization in utero. Thus, at this stage, the nascent embryo fuels trophectoderm development and implantation.


Asunto(s)
Blastocisto/citología , Células Madre Embrionarias/citología , Animales , Blastocisto/metabolismo , Proteína Morfogenética Ósea 4/farmacología , Autorrenovación de las Células , Ectodermo/citología , Ectodermo/metabolismo , Implantación del Embrión , Células Madre Embrionarias/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Factor 6 Similar a Kruppel/deficiencia , Factor 6 Similar a Kruppel/genética , Factor 6 Similar a Kruppel/metabolismo , Masculino , Ratones , Morfogénesis , Proteína Nodal/genética , Proteína Nodal/metabolismo , Proteína Nodal/farmacología , Transcriptoma , Trofoblastos/citología , Trofoblastos/metabolismo , Útero/citología , Útero/metabolismo
8.
J Med Ethics ; 2022 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-36600611

RESUMEN

Formulating sound and acceptable embryo research policy remains challenging especially in a pluralistic world. This challenge has acquired a new dimension of complexity with the advent of so-called embryo models, which are derived from stem cells. In this article, we present a normative strategy to facilitate the process of sound policy-making in the field of human embryology. This strategy involves seeking neutral agreements on higher level theories and doctrines as well as seeking agreements on the level of concrete policy proposals. We call this strategy: going high and low. By going high and low, the plurality of reasonable moral and epistemic convictions of stakeholders involved in the domain of human embryology is respected while the process of policy-making in this area is improved.

9.
Proc Natl Acad Sci U S A ; 115(4): E610-E619, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29311336

RESUMEN

The adult mouse subependymal zone provides a niche for mammalian neural stem cells (NSCs). However, the molecular signature, self-renewal potential, and fate behavior of NSCs remain poorly defined. Here we propose a model in which the fate of active NSCs is coupled to the total number of neighboring NSCs in a shared niche. Using knock-in reporter alleles and single-cell RNA sequencing, we show that the Wnt target Tnfrsf19/Troy identifies both active and quiescent NSCs. Quantitative analysis of genetic lineage tracing of individual NSCs under homeostasis or in response to injury reveals rapid expansion of stem-cell number before some return to quiescence. This behavior is best explained by stochastic fate decisions, where stem-cell number within a shared niche fluctuates over time. Fate mapping proliferating cells using a Ki67iresCreER allele confirms that active NSCs reversibly return to quiescence, achieving long-term self-renewal. Our findings suggest a niche-based mechanism for the regulation of NSC fate and number.


Asunto(s)
Ventrículos Laterales/citología , Células-Madre Neurales/fisiología , Nicho de Células Madre , Animales , Linaje de la Célula , Proliferación Celular , Ratones , Neurogénesis , Receptores del Factor de Necrosis Tumoral/metabolismo , Análisis de la Célula Individual , Transcriptoma
10.
Development ; 149(23)2022 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-36475526
11.
Development ; 144(5): 733-736, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28246208

RESUMEN

In November 2016, developmental biologists, synthetic biologists and engineers gathered in Paris for a meeting called 'Engineering the embryo'. The participants shared an interest in exploring how synthetic systems can reveal new principles of embryonic development, and how the in vitro manipulation and modeling of development using stem cells can be used to integrate ideas and expertise from physics, developmental biology and tissue engineering. As we review here, the conference pinpointed some of the challenges arising at the intersection of these fields, along with great enthusiasm for finding new approaches and collaborations.


Asunto(s)
Biología Evolutiva/métodos , Biología Evolutiva/tendencias , Células Madre/citología , Animales , Biofisica , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Modelos Biológicos , Paris , Robótica , Biología Sintética , Biología de Sistemas , Ingeniería de Tejidos/métodos , Ingeniería de Tejidos/tendencias
13.
Curr Opin Organ Transplant ; 24(5): 562-567, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31348016

RESUMEN

PURPOSE OF REVIEW: Engineering functional organs starting from stem or progenitor cells holds promise to address the urgent need for organ transplants. However, to date, the development of complex organ structures remains an open challenge. RECENT FINDINGS: Among multiple approaches to organ regeneration that are being investigated, two main directions can be identified, namely the patterned deposition of cells to impose specific structures, using bioprinting technologies, and (ii) the spontaneous development of organoids, according to principles of self-organization. In this review, we shortly describe the advantages and limitations of these paradigms and we discuss how they can synergize their positive features to better control and robustly develop organs from stem cells, toward organogenesis by design. SUMMARY: The outlined possibilities to bring together tools and concepts of bioprinting and self-organization will be relevant not only to generate implantable organs, but also to dissect fundamental mechanisms of organogenesis and to test therapeutic strategies in modeled pathological settings.


Asunto(s)
Órganos Artificiales , Bioimpresión/métodos , Impresión Tridimensional , Ingeniería de Tejidos/métodos , Humanos , Organogénesis/fisiología , Medicina Regenerativa , Trasplantes
14.
Proc Natl Acad Sci U S A ; 109(12): 4413-8, 2012 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-22388744

RESUMEN

Large bone defects naturally regenerate via a highly vascularized tissue which progressively remodels into cartilage and bone. Current approaches in bone tissue engineering are restricted by delayed vascularization and fail to recapitulate this stepwise differentiation toward bone tissue. Here, we use the morphogen Sonic Hedgehog (Shh) to induce the in vitro organization of an endothelial capillary network in an artificial tissue. We show that endogenous Hedgehog activity regulates angiogenic genes and the formation of vascular lumens. Exogenous Shh further induces the in vitro development of the vasculature (vascular lumen formation, size, distribution). Upon implantation, the in vitro development of the vasculature improves the in vivo perfusion of the artificial tissue and is necessary to contribute to, and enhance, the formation of de novo mature bone tissue. Similar to the regenerating callus, the artificial tissue undergoes intramembranous and endochondral ossification and forms a trabecular-like bone organ including bone-marrow-like cavities. These findings open the door for new strategies to treat large bone defects by closely mimicking natural endochondral bone repair.


Asunto(s)
Huesos/metabolismo , Proteínas Hedgehog/metabolismo , Ingeniería de Tejidos/métodos , Animales , Prótesis Vascular , Células de la Médula Ósea/citología , Diferenciación Celular , Matriz Extracelular/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Neovascularización Patológica , Osteogénesis , Medicina Regenerativa/métodos , Factores de Tiempo
15.
Proc Natl Acad Sci U S A ; 109(18): 6886-91, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22511716

RESUMEN

Physical forces play a major role in the organization of developing tissues. During vascular development, physical forces originating from a fluid phase or from cells pulling on their environment can alter cellular signaling and the behavior of cells. Here, we observe how tissue deformation spatially modulates angiogenic signals and angiogenesis. Using soft lithographic templates, we assemble three-dimensional, geometric tissues. The tissues contract autonomously, change shape stereotypically and form patterns of vascular structures in regions of high deformations. We show that this emergence correlates with the formation of a long-range gradient of Vascular Endothelial Growth Factor (VEGF) in interstitial cells, the local overexpression of the corresponding receptor VEGF receptor 2 (VEGFR-2) and local differences in endothelial cells proliferation. We suggest that tissue contractility and deformation can induce the formation of gradients of angiogenic microenvironments which could contribute to the long-range patterning of the vascular system.


Asunto(s)
Neovascularización Fisiológica/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Actinas/metabolismo , Secuencia de Bases , Fenómenos Biomecánicos , Técnicas de Cocultivo , Cartilla de ADN/genética , Matriz Extracelular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Miosinas/metabolismo , Neovascularización Fisiológica/genética , Transducción de Señal/fisiología , Ingeniería de Tejidos , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología
16.
STAR Protoc ; 4(2): 102151, 2023 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-36930647

RESUMEN

Classically, culturing mouse blastocysts with FGF4/TGF-ß1, two epiblast-secreted inducers, allows for deriving trophoblast stem cells that comprise fluctuating subpopulations reflecting both pre- and post-implantation stages. However, a more complete combination of inducers (adding LPA, IL11, BMP7, Activin A, 8-Br-cAMP) captures trophectoderm stem cells with enhanced transcriptomic similarity to the blastocyst trophectoderm and self-renewal, reduced differentiation. Also, the complete combination of inducers increased potential to form blastoids and to instruct decidualization in utero, thus better reflecting the blastocyst. For complete details on the use and execution of this protocol, please refer to Seong et al.1.

17.
Curr Opin Genet Dev ; 83: 102125, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37801801

RESUMEN

Despite over 40 years following the first birth from medically assisted reproduction (MAR) technologies, mechanisms underlying the key developmental events during the first 7 days of human development, such as signaling pathway contribution, are remaining a mystery. An in-depth mechanistic understanding of how the human preimplantation embryo develops would support the optimization of embryo quality assessment methods and culturing conditions, thereby increasing the success rate of MAR. However, the limited availability of human embryos, legitimate ethical concerns, and regulations still present an obstacle toward our advancement of knowledge. Stem cell-based embryonic models, including blastoids than model blastocysts, offer unprecedented opportunities to fill knowledge gaps and complement animal models. Blastoids' predictive power depends on how faithfully they recapitulate the blastocyst. Here, we review the state of the art of human pre- and peri-implantation development and outline the specificities of human embryo research to clarify the framework for blastoid research.


Asunto(s)
Blastocisto , Implantación del Embrión , Animales , Humanos , Implantación del Embrión/genética , Blastocisto/metabolismo , Desarrollo Embrionario/genética
18.
Nat Protoc ; 18(5): 1584-1620, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36792779

RESUMEN

Human early development sets the stage for embryonic and adult life but remains difficult to investigate. A solution came from the ability of stem cells to organize into structures resembling preimplantation embryos-blastocysts-that we termed blastoids. This embryo model is available in unlimited numbers and could thus support scientific and medical advances. However, its predictive power depends on how faithfully it recapitulates the blastocyst. Here, we describe how we formed human blastoids that (1) efficiently achieve the morphology of the blastocyst and (2) form lineages according to the pace and sequence of blastocyst development, (3) ultimately forming cells that transcriptionally reflect the blastocyst (preimplantation stage). We employ three different commercially available 96- and 24-well microwell plates with results similar to our custom-made ones, and show that blastoids form in clinical in vitro fertilization medium and can be cryopreserved for shipping. Finally, we explain how blastoids replicate the directional process of implantation into endometrial organoids, specifically when these are hormonally stimulated. It takes 4 d for human blastoids to form and 10 d to prepare the endometrial implantation assay, and we have cultured blastoids up to 6 d (time-equivalent of day 13). On the basis of our experience, we anticipate that a person with ~1 year of human pluripotent stem cell culture experience and of organoid culture should be able to perform the protocol. Altogether, blastoids offer an opportunity to establish scientific and biomedical discovery programs for early pregnancy, and an ethical alternative to the use of embryos.


Asunto(s)
Blastocisto , Implantación del Embrión , Embarazo , Adulto , Femenino , Humanos , Desarrollo Embrionario , Embrión de Mamíferos , Criopreservación
19.
Biomed Microdevices ; 14(1): 95-107, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22048776

RESUMEN

This paper presents cell culture substrates in the form of microcontainer arrays with overlaid surface topographies, and a technology for their fabrication. The new fabrication technology is based on microscale thermoforming of thin polymer films whose surfaces are topographically prepatterned on a micro- or nanoscale. For microthermoforming, we apply a new process on the basis of temporary back moulding of polymer films and use the novel concept of a perforated-sheet-like mould. Thermal micro- or nanoimprinting is applied for prepatterning. The novel cell container arrays are fabricated from polylactic acid (PLA) films. The thin-walled microcontainer structures have the shape of a spherical calotte merging into a hexagonal shape at their upper circumferential edges. In the arrays, the cell containers are arranged densely packed in honeycomb fashion. The inner surfaces of the highly curved container walls are provided with various topographical micro- and nanopatterns. For a first validation of the microcontainer arrays as in vitro cell culture substrates, C2C12 mouse premyoblasts are cultured in containers with microgrooved surfaces and shown to align along the grooves in the three-dimensional film substrates. In future stem-cell-biological and tissue engineering applications, microcontainers fabricated using the proposed technology may act as geometrically defined artificial microenvironments or niches.


Asunto(s)
Polímeros/química , Ingeniería de Tejidos/métodos , Animales , Línea Celular , Ensayos Analíticos de Alto Rendimiento , Ácido Láctico/química , Ratones , Poliésteres , Propiedades de Superficie
20.
J Vis Exp ; (186)2022 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-36036618

RESUMEN

A model of the human blastocyst formed from stem cells (blastoid) would support scientific and medical advances. However, its predictive power will depend on its ability to efficiently, timely, and faithfully recapitulate the sequences of blastocyst development (morphogenesis, specification, patterning), and to form cells reflecting the blastocyst stage. Here we show that naïve human pluripotent stem cells cultured in PXGL conditions and then triply inhibited for the Hippo, transforming growth factor- ß, and extracellular signal-regulated kinase pathways efficiently undergo morphogenesis to form blastoids (>70%). Matching with developmental timing (~4 days), blastoids unroll the blastocyst sequence of specification by producing analogs of the trophoblast and epiblast, followed by the formation of analogs of the primitive endoderm and the polar trophoblasts. This results in the formation of cells transcriptionally similar to the blastocyst (>96%) and a minority of post-implantation analogs. Blastoids efficiently pattern by forming the embryonic-abembryonic axis marked by the maturation of the polar region (NR2F2+), which acquires the specific potential to directionally attach to hormonally stimulated endometrial cells, as in utero. Such a human blastoid is a scalable, versatile, and ethical model to study human development and implantation in vitro.


Asunto(s)
Implantación del Embrión , Desarrollo Embrionario , Blastocisto , Diferenciación Celular , Linaje de la Célula , Endodermo , Femenino , Estratos Germinativos , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA