Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Cell ; 187(13): 3224-3228, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38906097

RESUMEN

The next 50 years of developmental biology will illuminate exciting new discoveries but are also poised to provide solutions to important problems society faces. Ten scientists whose work intersects with developmental biology in various capacities tell us about their vision for the future.


Asunto(s)
Biología Evolutiva , Biología Evolutiva/tendencias , Humanos , Células Madre/citología , Animales , Investigación con Células Madre
2.
Cell ; 186(25): 5587-5605.e27, 2023 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-38029745

RESUMEN

The number one cause of human fetal death are defects in heart development. Because the human embryonic heart is inaccessible and the impacts of mutations, drugs, and environmental factors on the specialized functions of different heart compartments are not captured by in vitro models, determining the underlying causes is difficult. Here, we established a human cardioid platform that recapitulates the development of all major embryonic heart compartments, including right and left ventricles, atria, outflow tract, and atrioventricular canal. By leveraging 2D and 3D differentiation, we efficiently generated progenitor subsets with distinct first, anterior, and posterior second heart field identities. This advance enabled the reproducible generation of cardioids with compartment-specific in vivo-like gene expression profiles, morphologies, and functions. We used this platform to unravel the ontogeny of signal and contraction propagation between interacting heart chambers and dissect how mutations, teratogens, and drugs cause compartment-specific defects in the developing human heart.


Asunto(s)
Cardiopatías , Ventrículos Cardíacos , Corazón , Humanos , Transcriptoma/genética , Línea Celular , Regulación del Desarrollo de la Expresión Génica , Cardiopatías/genética , Cardiopatías/metabolismo
3.
Cell ; 184(12): 3299-3317.e22, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34019794

RESUMEN

Organoids capable of forming tissue-like structures have transformed our ability to model human development and disease. With the notable exception of the human heart, lineage-specific self-organizing organoids have been reported for all major organs. Here, we established self-organizing cardioids from human pluripotent stem cells that intrinsically specify, pattern, and morph into chamber-like structures containing a cavity. Cardioid complexity can be controlled by signaling that instructs the separation of cardiomyocyte and endothelial layers and by directing epicardial spreading, inward migration, and differentiation. We find that cavity morphogenesis is governed by a mesodermal WNT-BMP signaling axis and requires its target HAND1, a transcription factor linked to developmental heart chamber defects. Upon cryoinjury, cardioids initiated a cell-type-dependent accumulation of extracellular matrix, an early hallmark of both regeneration and heart disease. Thus, human cardioids represent a powerful platform to mechanistically dissect self-organization, congenital heart defects and serve as a foundation for future translational research.


Asunto(s)
Corazón/embriología , Organogénesis , Organoides/embriología , Activinas/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Calcio/metabolismo , Línea Celular , Linaje de la Célula , Pollos , Células Endoteliales/citología , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Fibroblastos/citología , Proteína Homeótica Nkx-2.5/metabolismo , Humanos , Masculino , Mesodermo/embriología , Modelos Biológicos , Miocardio/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas Wnt/metabolismo
4.
Mol Cell Proteomics ; 22(12): 100665, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37839701

RESUMEN

Multiplexed and label-free mass spectrometry-based approaches with single-cell resolution have attributed surprising heterogeneity to presumed homogenous cell populations. Even though specialized experimental designs and instrumentation have demonstrated remarkable advances, the efficient sample preparation of single cells still lags. Here, we introduce the proteoCHIP, a universal option for single-cell proteomics sample preparation including multiplexed labeling up to 16-plex with high sensitivity and throughput. The automated processing using a commercial system combining single-cell isolation and picoliter dispensing, the cellenONE, reduces final sample volumes to low nanoliters submerged in a hexadecane layer simultaneously eliminating error-prone manual sample handling and overcoming evaporation. The specialized proteoCHIP design allows direct injection of single cells via a standard autosampler resulting in around 1500 protein groups per TMT10-plex with reduced or eliminated need for a carrier proteome. We evaluated the effect of wider precursor isolation windows at single-cell input levels and found that using 2 Da isolation windows increased overall sensitivity without significantly impacting interference. Using the dedicated mass spectrometry acquisition strategies detailed here, we identified on average close to 2000 proteins per TMT10-plex across 170 multiplexed single cells that readily distinguished human cell types. Overall, our workflow combines highly efficient sample preparation, chromatographic and ion mobility-based filtering, rapid wide-window data-dependent acquisition analysis, and intelligent data analysis for optimal multiplexed single-cell proteomics. This versatile and automated proteoCHIP-based sample preparation approach is sufficiently sensitive to drive biological applications of single-cell proteomics and can be readily adopted by proteomics laboratories.


Asunto(s)
Proteoma , Proteómica , Humanos , Proteómica/métodos , Flujo de Trabajo , Espectrometría de Masas/métodos , Proteoma/metabolismo
5.
Development ; 148(16)2021 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-34423833

RESUMEN

Cardiac congenital disabilities are the most common organ malformations, but we still do not understand how they arise in the human embryo. Moreover, although cardiovascular disease is the most common cause of death globally, the development of new therapies is lagging compared with other fields. One major bottleneck hindering progress is the lack of self-organizing human cardiac models that recapitulate key aspects of human heart development, physiology and disease. Current in vitro cardiac three-dimensional systems are either engineered constructs or spherical aggregates of cardiomyocytes and other cell types. Although tissue engineering enables the modeling of some electro-mechanical properties, it falls short of mimicking heart development, morphogenetic defects and many clinically relevant aspects of cardiomyopathies. Here, we review different approaches and recent efforts to overcome these challenges in the field using a new generation of self-organizing embryonic and cardiac organoids.


Asunto(s)
Corazón/embriología , Modelos Cardiovasculares , Miocitos Cardíacos/metabolismo , Organogénesis/fisiología , Organoides/embriología , Ingeniería de Tejidos/métodos , Animales , Enfermedades Cardiovasculares , Técnicas de Cocultivo/métodos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo
6.
Nature ; 555(7695): 256-259, 2018 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-29489750

RESUMEN

The TGFß pathway has essential roles in embryonic development, organ homeostasis, tissue repair and disease. These diverse effects are mediated through the intracellular effectors SMAD2 and SMAD3 (hereafter SMAD2/3), whose canonical function is to control the activity of target genes by interacting with transcriptional regulators. Therefore, a complete description of the factors that interact with SMAD2/3 in a given cell type would have broad implications for many areas of cell biology. Here we describe the interactome of SMAD2/3 in human pluripotent stem cells. This analysis reveals that SMAD2/3 is involved in multiple molecular processes in addition to its role in transcription. In particular, we identify a functional interaction with the METTL3-METTL14-WTAP complex, which mediates the conversion of adenosine to N6-methyladenosine (m6A) on RNA. We show that SMAD2/3 promotes binding of the m6A methyltransferase complex to a subset of transcripts involved in early cell fate decisions. This mechanism destabilizes specific SMAD2/3 transcriptional targets, including the pluripotency factor gene NANOG, priming them for rapid downregulation upon differentiation to enable timely exit from pluripotency. Collectively, these findings reveal the mechanism by which extracellular signalling can induce rapid cellular responses through regulation of the epitranscriptome. These aspects of TGFß signalling could have far-reaching implications in many other cell types and in diseases such as cancer.


Asunto(s)
Adenosina/análogos & derivados , Diferenciación Celular/genética , Células Madre Pluripotentes/metabolismo , ARN Mensajero/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Activinas/metabolismo , Adenosina/metabolismo , Animales , Proteínas de Ciclo Celular , Epigénesis Genética , Humanos , Metilación , Metiltransferasas/química , Metiltransferasas/metabolismo , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Proteína Homeótica Nanog/metabolismo , Proteína Nodal/metabolismo , Proteínas Nucleares/metabolismo , Células Madre Pluripotentes/citología , Unión Proteica , Factores de Empalme de ARN , ARN Mensajero/química , ARN Mensajero/genética , Transducción de Señal , Transcriptoma
7.
Mol Cell Proteomics ; 21(1): 100177, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34793982

RESUMEN

Single-cell transcriptomics has revolutionized our understanding of basic biology and disease. Since transcript levels often do not correlate with protein expression, it is crucial to complement transcriptomics approaches with proteome analyses at single-cell resolution. Despite continuous technological improvements in sensitivity, mass-spectrometry-based single-cell proteomics ultimately faces the challenge of reproducibly comparing the protein expression profiles of thousands of individual cells. Here, we combine two hitherto opposing analytical strategies, DIA and Tandem-Mass-Tag (TMT)-multiplexing, to generate highly reproducible, quantitative proteome signatures from ultralow input samples. We developed a novel, identification-independent proteomics data-analysis pipeline that allows to quantitatively compare DIA-TMT proteome signatures across hundreds of samples independent of their biological origin to identify cell types and single protein knockouts. These proteome signatures overcome the need to impute quantitative data due to accumulating detrimental amounts of missing data in standard multibatch TMT experiments. We validate our approach using integrative data analysis of different human cell lines and standard database searches for knockouts of defined proteins. Our data establish a novel and reproducible approach to markedly expand the numbers of proteins one detects from ultralow input samples.


Asunto(s)
Proteoma , Espectrometría de Masas en Tándem , Línea Celular , Humanos , Procesamiento Proteico-Postraduccional , Proteoma/metabolismo , Proteómica
8.
Genes Dev ; 29(7): 702-17, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25805847

RESUMEN

Stem cells can self-renew and differentiate into multiple cell types. These characteristics are maintained by the combination of specific signaling pathways and transcription factors that cooperate to establish a unique epigenetic state. Despite the broad interest of these mechanisms, the precise molecular controls by which extracellular signals organize epigenetic marks to confer multipotency remain to be uncovered. Here, we use human embryonic stem cells (hESCs) to show that the Activin-SMAD2/3 signaling pathway cooperates with the core pluripotency factor NANOG to recruit the DPY30-COMPASS histone modifiers onto key developmental genes. Functional studies demonstrate the importance of these interactions for correct histone 3 Lys4 trimethylation and also self-renewal and differentiation. Finally, genetic studies in mice show that Dpy30 is also necessary to maintain pluripotency in the pregastrulation embryo, thereby confirming the existence of similar regulations in vivo during early embryonic development. Our results reveal the mechanisms by which extracellular factors coordinate chromatin status and cell fate decisions in hESCs.


Asunto(s)
Activinas/metabolismo , Diferenciación Celular/genética , Cromatina/genética , Histonas/genética , Proteínas de Homeodominio/metabolismo , Proteína Nodal/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Cromatina/metabolismo , Embrión de Mamíferos , Células Madre Embrionarias , Epigénesis Genética/genética , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Ratones , Proteína Homeótica Nanog , Proteína Smad2/metabolismo , Proteína smad3/metabolismo
9.
Anal Chem ; 94(5): 2434-2443, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-34967612

RESUMEN

Single-cell proteomics workflows have considerably improved in sensitivity and reproducibility to characterize as-yet unknown biological phenomena. With the emergence of multiplexed single-cell proteomics, studies increasingly present single-cell measurements in conjunction with an abundant congruent carrier to improve the precursor selection and enhance identifications. While these extreme carrier spikes are often >100× more abundant than the investigated samples, the total ion current undoubtably increases but the quantitative accuracy possibly is affected. We here focus on narrowly titrated carrier spikes (i.e., <20×) and assess their elimination for a comparable sensitivity with superior accuracy. We find that subtle changes in the carrier ratio can severely impact the measurement variability and describe alternative multiplexing strategies to evaluate data quality. Lastly, we demonstrate elevated replicate overlap while preserving acquisition throughput at an improved quantitative accuracy with DIA-TMT and discuss optimized experimental designs for multiplexed proteomics of trace samples. This comprehensive benchmarking gives an overview of currently available techniques and guides the conceptualization of the optimal single-cell proteomics experiment.


Asunto(s)
Proteoma , Proteómica , Proteómica/métodos , Reproducibilidad de los Resultados , Flujo de Trabajo
10.
Development ; 142(12): 2121-35, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-26015544

RESUMEN

The transcription factor brachyury (T, BRA) is one of the first markers of gastrulation and lineage specification in vertebrates. Despite its wide use and importance in stem cell and developmental biology, its functional genomic targets in human cells are largely unknown. Here, we use differentiating human embryonic stem cells to study the role of BRA in activin A-induced endoderm and BMP4-induced mesoderm progenitors. We show that BRA has distinct genome-wide binding landscapes in these two cell populations, and that BRA interacts and collaborates with SMAD1 or SMAD2/3 signalling to regulate the expression of its target genes in a cell-specific manner. Importantly, by manipulating the levels of BRA in cells exposed to different signalling environments, we demonstrate that BRA is essential for mesoderm but not for endoderm formation. Together, our data illuminate the function of BRA in the context of human embryonic development and show that the regulatory role of BRA is context dependent. Our study reinforces the importance of analysing the functions of a transcription factor in different cellular and signalling environments.


Asunto(s)
Células Madre Embrionarias/citología , Proteínas Fetales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Neurogénesis/fisiología , Proteína Smad1/metabolismo , Proteínas de Dominio T Box/metabolismo , Animales , Proteína Morfogenética Ósea 4/metabolismo , Línea Celular , Células Madre Embrionarias/metabolismo , Endodermo/citología , Gastrulación/fisiología , Humanos , Mesodermo/citología , Ratones , Ratones Transgénicos , Proteína Smad2/metabolismo , Proteína smad3/metabolismo
11.
Stem Cells ; 30(2): 161-8, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22109880

RESUMEN

Mouse epiblast stem cells (EpiSCs) derived from postimplantation embryos are developmentally and functionally different from embryonic stem cells (ESCs) generated from blastocysts. EpiSCs require Activin A and FGF2 signaling for self-renewal, similar to human ESCs (hESCs), while mouse ESCs require LIF and BMP4. Unlike ESCs, EpiSCs have undergone X-inactivation, similar to the tendency of hESCs. The shared self-renewal and X-inactivation properties of EpiSCs and hESCs suggest that they have an epigenetic state distinct from ESCs. This hypothesis predicts that EpiSCs would have monoallelic expression of most imprinted genes, like that observed in hESCs. Here, we confirm this prediction. By contrast, we find that mouse induced pluripotent stem cells (iPSCs) tend to lose imprinting similar to mouse ESCs. These findings reveal that iPSCs have an epigenetic status associated with their pluripotent state rather than their developmental origin. Our results also reinforce the view that hESCs and EpiSCs are in vitro counterparts, sharing an epigenetic status distinct from ESCs and iPSCs.


Asunto(s)
Epigénesis Genética , Impresión Genómica , Células Madre Pluripotentes/metabolismo , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Células Cultivadas , Metilación de ADN , Células Madre Embrionarias/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Estratos Germinativos/citología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
12.
Sci Adv ; 9(13): eade1792, 2023 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-36989351

RESUMEN

The blueprints of developing organs are preset at the early stages of embryogenesis. Transcriptional and epigenetic mechanisms are proposed to preset developmental trajectories. However, we reveal that the competence for the future cardiac fate of human embryonic stem cells (hESCs) is preset in pluripotency by a specialized mRNA translation circuit controlled by RBPMS. RBPMS is recruited to active ribosomes in hESCs to control the translation of essential factors needed for cardiac commitment program, including Wingless/Integrated (WNT) signaling. Consequently, RBPMS loss specifically and severely impedes cardiac mesoderm specification, leading to patterning and morphogenetic defects in human cardiac organoids. Mechanistically, RBPMS specializes mRNA translation, selectively via 3'UTR binding and globally by promoting translation initiation. Accordingly, RBPMS loss causes translation initiation defects highlighted by aberrant retention of the EIF3 complex and depletion of EIF5A from mRNAs, thereby abrogating ribosome recruitment. We demonstrate how future fate trajectories are programmed during embryogenesis by specialized mRNA translation.


Asunto(s)
Células Madre Embrionarias Humanas , Humanos , Células Madre Embrionarias Humanas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ribosomas/metabolismo , Corazón , Transducción de Señal , Proteínas de Unión al ARN/metabolismo
13.
Nat Cell Biol ; 25(1): 42-55, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36604593

RESUMEN

ZNF462 haploinsufficiency is linked to Weiss-Kruszka syndrome, a genetic disorder characterized by neurodevelopmental defects, including autism. Though conserved in vertebrates and essential for embryonic development, the molecular functions of ZNF462 remain unclear. We identified its murine homologue ZFP462 in a screen for mediators of epigenetic gene silencing. Here we show that ZFP462 safeguards neural lineage specification of mouse embryonic stem cells (ESCs) by targeting the H3K9-specific histone methyltransferase complex G9A/GLP to silence meso-endodermal genes. ZFP462 binds to transposable elements that are potential enhancers harbouring pluripotency and meso-endoderm transcription factor binding sites. Recruiting G9A/GLP, ZFP462 seeds heterochromatin, restricting transcription factor binding. Loss of ZFP462 in ESCs results in increased chromatin accessibility at target sites and ectopic expression of meso-endodermal genes. Taken together, ZFP462 confers lineage and locus specificity to the broadly expressed epigenetic regulator G9A/GLP. Our results suggest that aberrant activation of lineage non-specific genes in the neuronal lineage underlies ZNF462-associated neurodevelopmental pathology.


Asunto(s)
Heterocromatina , N-Metiltransferasa de Histona-Lisina , Animales , Ratones , Heterocromatina/genética , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Cromatina , Células Madre Embrionarias , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas de Unión al ADN/genética , Proteínas del Tejido Nervioso/genética
14.
Cell Stem Cell ; 28(2): 180-181, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33545075

RESUMEN

Current in vitro systems are powerful tools for studying early heart specification but lack the ability to model morphological events. Reporting in this issue of Cell Stem Cell, Rossi et al. (2021) present a patterned embryonic organoid model (gastruloid) that mimics aspects of early cardiogenesis.


Asunto(s)
Corazón , Organogénesis , Organoides
15.
Stem Cell Reports ; 16(3): 641-655, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33606988

RESUMEN

Increasing brown adipose tissue (BAT) mass and activation is a therapeutic strategy to treat obesity and complications. Obese and diabetic patients possess low amounts of BAT, so an efficient way to expand their mass is necessary. There is limited knowledge about how human BAT develops, differentiates, and is optimally activated. Accessing human BAT is challenging, given its low volume and anatomical dispersion. These constraints make detailed BAT-related developmental and functional mechanistic studies in humans virtually impossible. We have developed and characterized functionally and molecularly a new chemically defined protocol for the differentiation of human pluripotent stem cells (hPSCs) into brown adipocytes (BAs) that overcomes current limitations. This protocol recapitulates step by step the physiological developmental path of human BAT. The BAs obtained express BA and thermogenic markers, are insulin sensitive, and responsive to ß-adrenergic stimuli. This new protocol is scalable, enabling the study of human BAs at early stages of development.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipogénesis , Tejido Adiposo Pardo/metabolismo , Técnicas de Cultivo de Célula/métodos , Células Madre Pluripotentes/metabolismo , Termogénesis , Factores de Transcripción/metabolismo , Biomarcadores/metabolismo , Diferenciación Celular , Línea Celular , Regulación del Desarrollo de la Expresión Génica , Humanos , Reproducibilidad de los Resultados
16.
Endocrinology ; 160(10): 2282-2297, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31290979

RESUMEN

The human endometrium is the inner lining of the uterus consisting of stromal and epithelial (secretory and ciliated) cells. It undergoes a hormonally regulated monthly cycle of growth, differentiation, and desquamation. However, how these cyclic changes control the balance between secretory and ciliated cells remains unclear. Here, we established endometrial organoids to investigate the estrogen (E2)-driven control of cell fate decisions in human endometrial epithelium. We demonstrate that they preserve the structure, expression patterns, secretory properties, and E2 responsiveness of their tissue of origin. Next, we show that the induction of ciliated cells is orchestrated by the coordinated action of E2 and NOTCH signaling. Although E2 is the primary driver, inhibition of NOTCH signaling provides a permissive environment. However, inhibition of NOTCH alone is not sufficient to trigger ciliogenesis. Overall, we provide insights into endometrial biology and propose endometrial organoids as a robust and powerful model for studying ciliogenesis in vitro.


Asunto(s)
Cilios/fisiología , Endometrio/fisiología , Estrógenos/metabolismo , Organoides/metabolismo , Femenino , Regulación de la Expresión Génica/fisiología , Humanos , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal , Técnicas de Cultivo de Tejidos
17.
Stem Cell Reports ; 11(2): 537-551, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30078556

RESUMEN

Defective placentation is the underlying cause of various pregnancy complications, such as severe intrauterine growth restriction and preeclampsia. However, studies on human placental development are hampered by the lack of a self-renewing in vitro model that would recapitulate formation of trophoblast progenitors and differentiated subtypes, syncytiotrophoblast (STB) and invasive extravillous trophoblast (EVT), in a 3D orientation. Hence, we established long-term expanding organoid cultures from purified first-trimester cytotrophoblasts (CTBs). Molecular analyses revealed that the CTB organoid cultures (CTB-ORGs) express markers of trophoblast stemness and proliferation and are highly similar to primary CTBs at the level of global gene expression. Whereas CTB-ORGs spontaneously generated STBs, withdrawal of factors for self-renewal induced trophoblast outgrowth, expressing the EVT progenitor marker NOTCH1, and provoked formation of adjacent, distally located HLA-G+ EVTs. In summary, we established human CTB-ORGs that grow and differentiate under defined culture conditions, allowing future human placental disease modeling.


Asunto(s)
Diferenciación Celular , Autorrenovación de las Células , Organoides/citología , Placenta/citología , Trofoblastos/citología , Biomarcadores , Proliferación Celular , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Embarazo , Trofoblastos/metabolismo , Vía de Señalización Wnt
19.
Cell Stem Cell ; 15(3): 310-325, 2014 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-25042702

RESUMEN

Mesoderm is induced at the primitive streak (PS) and patterns subsequently into mesodermal subtypes and organ precursors. It is unclear whether mesoderm induction generates a multipotent PS progenitor or several distinct ones with restricted subtype potentials. We induced mesoderm in human pluripotent stem cells with ACTIVIN and BMP or with GSK3-ß inhibition. Both approaches induced BRACHYURY(+) mesoderm of distinct PS-like identities, which had differing patterning potential. ACTIVIN and BMP-induced mesoderm patterned into cardiac but not somitic subtypes. Conversely, PS precursors induced by GSK3-ß inhibition did not generate lateral plate and cardiac mesoderm and favored instead somitic differentiation. The mechanism of these cell fate decisions involved mutual repression of NANOG and CDX2. Although NANOG was required for cardiac specification but blocked somitic subtypes, CDX2 was required for somitic mesoderm but blocked cardiac differentiation. In sum, rather than forming a common PS progenitor, separate induction mechanisms distinguish human mesoderm subtypes.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Mesodermo/embriología , Mesodermo/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Activinas/metabolismo , Tipificación del Cuerpo , Proteínas Morfogenéticas Óseas/metabolismo , Factor de Transcripción CDX2 , Línea Celular , Linaje de la Célula , Proteínas Fetales/metabolismo , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Mesodermo/citología , Miocardio/citología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Proteína Homeótica Nanog , Línea Primitiva/citología , Secuencias Reguladoras de Ácidos Nucleicos/genética , Transducción de Señal , Proteínas de Dominio T Box/metabolismo
20.
Cell Stem Cell ; 10(6): 646-647, 2012 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-22704499

RESUMEN

Differentiating tissue stem cells can self-assemble into structures that strikingly resemble functional organ subunits. Translating this insight to regenerative medicine presents several challenges.


Asunto(s)
Materiales Biocompatibles , Medicina Regenerativa/métodos , Células Madre/citología , Ingeniería de Tejidos , Animales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA