Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
PLoS Biol ; 21(6): e3002121, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37315073

RESUMEN

Pluripotency defines the unlimited potential of individual cells of vertebrate embryos, from which all adult somatic cells and germ cells are derived. Understanding how the programming of pluripotency evolved has been obscured in part by a lack of data from lower vertebrates; in model systems such as frogs and zebrafish, the function of the pluripotency genes NANOG and POU5F1 have diverged. Here, we investigated how the axolotl ortholog of NANOG programs pluripotency during development. Axolotl NANOG is absolutely required for gastrulation and germ-layer commitment. We show that in axolotl primitive ectoderm (animal caps; ACs) NANOG and NODAL activity, as well as the epigenetic modifying enzyme DPY30, are required for the mass deposition of H3K4me3 in pluripotent chromatin. We also demonstrate that all 3 protein activities are required for ACs to establish the competency to differentiate toward mesoderm. Our results suggest the ancient function of NANOG may be establishing the competence for lineage differentiation in early cells. These observations provide insights into embryonic development in the tetrapod ancestor from which terrestrial vertebrates evolved.


Asunto(s)
Proteínas de Homeodominio , Células Madre Pluripotentes , Animales , Proteínas de Homeodominio/metabolismo , Ambystoma mexicanum/genética , Ambystoma mexicanum/metabolismo , Pez Cebra/genética , Diferenciación Celular , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Regulación del Desarrollo de la Expresión Génica
2.
Development ; 149(6)2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35319748

RESUMEN

Developmental failures occurring shortly after blastocyst hatching from the zona pellucida constitute a major cause of pregnancy losses in both humans and farm ungulates. The developmental events occurring following hatching in ungulates include the proliferation and maturation of extra-embryonic membranes - trophoblast and hypoblast - and the formation of a flat embryonic disc, similar to that found in humans, which initiates gastrulation prior to implantation. Unfortunately, our understanding of these key processes for embryo survival is limited because current culture systems cannot sustain ungulate embryo development beyond hatching. Here, we report a culture system that recapitulates most developmental landmarks of gastrulating ovine embryos: trophoblast maturation, hypoblast migration, embryonic disc formation, disappearance of the Rauber's layer, epiblast polarization and mesoderm differentiation. Our system represents a highly valuable platform for exploring the cell differentiation, proliferation and migration processes governing gastrulation in a flat embryonic disc and for understanding pregnancy failures during the second week of gestation. This article has an associated 'The people behind the papers' interview.


Asunto(s)
Gastrulación , Estratos Germinativos , Animales , Blastocisto , Embrión de Mamíferos , Desarrollo Embrionario , Femenino , Humanos , Embarazo , Ovinos
3.
Development ; 148(23)2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34874452

RESUMEN

Despite four decades of effort, robust propagation of pluripotent stem cells from livestock animals remains challenging. The requirements for self-renewal are unclear and the relationship of cultured stem cells to pluripotent cells resident in the embryo uncertain. Here, we avoided using feeder cells or serum factors to provide a defined culture microenvironment. We show that the combination of activin A, fibroblast growth factor and the Wnt inhibitor XAV939 (AFX) supports establishment and continuous expansion of pluripotent stem cell lines from porcine, ovine and bovine embryos. Germ layer differentiation was evident in teratomas and readily induced in vitro. Global transcriptome analyses highlighted commonality in transcription factor expression across the three species, while global comparison with porcine embryo stages showed proximity to bilaminar disc epiblast. Clonal genetic manipulation and gene targeting were exemplified in porcine stem cells. We further demonstrated that genetically modified AFX stem cells gave rise to cloned porcine foetuses by nuclear transfer. In summary, for major livestock mammals, pluripotent stem cells related to the formative embryonic disc are reliably established using a common and defined signalling environment. This article has an associated 'The people behind the papers' interview.


Asunto(s)
Diferenciación Celular , Embrión de Mamíferos/metabolismo , Estratos Germinativos/metabolismo , Células Madre Pluripotentes/metabolismo , Animales , Bovinos , Embrión de Mamíferos/citología , Estratos Germinativos/citología , Ganado , Células Madre Pluripotentes/citología , Ovinos , Especificidad de la Especie , Porcinos
4.
Nature ; 546(7658): 416-420, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28607482

RESUMEN

Human primordial germ cells (hPGCs), the precursors of sperm and eggs, originate during weeks 2-3 of early post-implantation development. Using in vitro models of hPGC induction, recent studies have suggested that there are marked mechanistic differences in the specification of human and mouse PGCs. This may be due in part to the divergence in their pluripotency networks and early post-implantation development. As early human embryos are not accessible for direct study, we considered alternatives including porcine embryos that, as in humans, develop as bilaminar embryonic discs. Here we show that porcine PGCs originate from the posterior pre-primitive-streak competent epiblast by sequential upregulation of SOX17 and BLIMP1 in response to WNT and BMP signalling. We use this model together with human and monkey in vitro models simulating peri-gastrulation development to show the conserved principles of epiblast development for competency for primordial germ cell fate. This process is followed by initiation of the epigenetic program and regulated by a balanced SOX17-BLIMP1 gene dosage. Our combinatorial approach using human, porcine and monkey in vivo and in vitro models provides synthetic insights into early human development.


Asunto(s)
Diferenciación Celular , Desarrollo Embrionario , Células Germinativas/citología , Macaca fascicularis/embriología , Modelos Biológicos , Células Madre Pluripotentes/citología , Porcinos/embriología , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Linaje de la Célula , Cuerpos Embrioides/citología , Epigénesis Genética , Femenino , Gastrulación , Dosificación de Gen , Células Germinativas/metabolismo , Estratos Germinativos/citología , Humanos , Técnicas In Vitro , Masculino , Modelos Animales , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Línea Primitiva/citología , Proteínas Represoras/genética , Factores de Transcripción SOXF/genética , Vía de Señalización Wnt
5.
Reprod Fertil Dev ; 36(2): 81-92, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38064186

RESUMEN

Communication between the maternal endometrium and developing embryo/conceptus is critical to support successful pregnancy to term. Studying the peri-implantation period of pregnancy is critical as this is when most pregnancy loss occurs in cattle. Our current understanding of these interactions is limited, due to the lack of appropriate in vitro models to assess these interactions. The endometrium is a complex and heterogeneous tissue that is regulated in a transcriptional and translational manner throughout the oestrous cycle. While there are in vitro models to study endometrial function, they are static and 2D in nature or explant models and are limited in how well they recapitulate the in vivo endometrium. Recent developments in organoid systems, microfluidic approaches, extracellular matrix biology, and in silico approaches provide a new opportunity to develop in vitro systems that better model the in vivo scenario. This will allow us to investigate in a more high-throughput manner the fundamental molecular interactions that are required for successful pregnancy in cattle.


Asunto(s)
Implantación del Embrión , Endometrio , Embarazo , Femenino , Bovinos , Animales , Embrión de Mamíferos
6.
Reproduction ; 162(1): F59-F68, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34096507

RESUMEN

The birth and adult development of 'Dolly' the sheep, the first mammal produced by the transfer of a terminally differentiated cell nucleus into an egg, provided unequivocal evidence of nuclear equivalence among somatic cells. This ground-breaking experiment challenged a long-standing dogma of irreversible cellular differentiation that prevailed for over a century and enabled the development of methodologies for reversal of differentiation of somatic cells, also known as nuclear reprogramming. Thanks to this new paradigm, novel alternatives for regenerative medicine in humans, improved animal breeding in domestic animals and approaches to species conservation through reproductive methodologies have emerged. Combined with the incorporation of new tools for genetic modification, these novel techniques promise to (i) transform and accelerate our understanding of genetic diseases and the development of targeted therapies through creation of tailored animal models, (ii) provide safe animal cells, tissues and organs for xenotransplantation, (iii) contribute to the preservation of endangered species, and (iv) improve global food security whilst reducing the environmental impact of animal production. This review discusses recent advances that build on the conceptual legacy of nuclear transfer and - when combined with gene editing - will have transformative potential for medicine, biodiversity and sustainable agriculture. We conclude that the potential of these technologies depends on further fundamental and translational research directed at improving the efficiency and safety of these methods.


Asunto(s)
Animales Modificados Genéticamente/genética , Núcleo Celular/genética , Reprogramación Celular , Clonación de Organismos/veterinaria , Edición Génica , Ganado/genética , Técnicas de Transferencia Nuclear/veterinaria , Animales , Animales Modificados Genéticamente/crecimiento & desarrollo , Aniversarios y Eventos Especiales , Clonación de Organismos/métodos , Clonación de Organismos/tendencias , Ganado/crecimiento & desarrollo
7.
Development ; 149(21)2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36259525

Asunto(s)
Ovario , Edición , Femenino , Humanos
8.
BMC Dev Biol ; 19(1): 13, 2019 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-31272387

RESUMEN

BACKGROUND: The segregation of the hypoblast and the emergence of the pluripotent epiblast mark the final stages of blastocyst formation in mammalian embryos. In bovine embryos the formation of the hypoblast has been partially studied, and evidence shows that MEK signalling plays a limited role in the segregation of this lineage. Here we explored the role of different signalling pathways during lineage segregation in the bovine embryo using immunofluorescence analysis of NANOG and SOX17 as readouts of epiblast and hypoblast, respectively. RESULTS: We show that SOX17 starts to be expressed in 16-32-cell stage embryos, whereas NANOG is first detected from 8-cell stage. SOX17 is first co-expressed with NANOG, but these markers become mutually exclusive by the late blastocyst stage. By assessing the expression kinetics of NANOG/SOX17 we show that inhibition of MEK signalling can eliminate SOX17 expression in bovine blastocysts, without altering NANOG expression. Modulation of WNT, PKC and LIF did not affect NANOG expression in the epiblast when used in combination with the ERK inhibitor. CONCLUSIONS: This study shows that SOX17 can be used as a reliable early marker of hypoblast in the bovine, and based on its expression profile we show that the hypoblast segregates in day 7 blastocysts. Furthermore, SOX17 expression is abolished using 1 µM of PD0325901, without affecting the NANOG population in the epiblast. Modulation of WNT, PKC and LIF are not sufficient to support enhanced NANOG expression in the epiblast when combined with ERK inhibitor, indicating that additional signalling pathways should be examined to determine their potential roles in epiblast expansion.


Asunto(s)
Blastocisto/citología , Embrión de Mamíferos/embriología , Estratos Germinativos/embriología , Proteína Homeótica Nanog/metabolismo , Factores de Transcripción SOXF/metabolismo , Animales , Benzamidas/farmacología , Bovinos , Difenilamina/análogos & derivados , Difenilamina/farmacología , Estratos Germinativos/citología , Factor Inhibidor de Leucemia/biosíntesis , Proteína Homeótica Nanog/genética , Proteína Quinasa C/biosíntesis , Factores de Transcripción SOXF/genética , Transducción de Señal/fisiología , Proteína Wnt1/biosíntesis
9.
Development ; 142(16): 2730-9, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26286941

RESUMEN

Embryos of many animal models express germ line determinants that suppress transcription and mediate early germ line commitment, which occurs before the somatic cell lineages are established. However, not all animals segregate their germ line in this manner. The 'last cell standing' model describes primordial germ cell (PGC) development in axolotls, in which PGCs are maintained by an extracellular signalling niche, and germ line commitment occurs after gastrulation. Here, we propose that this 'stochastic' mode of PGC specification is conserved in vertebrates, including non-rodent mammals. We postulate that early germ line segregation liberates genetic regulatory networks for somatic development to evolve, and that it therefore emerged repeatedly in the animal kingdom in response to natural selection.


Asunto(s)
Evolución Biológica , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Células Germinativas/fisiología , Modelos Biológicos , Vertebrados/embriología , Animales , Especificidad de la Especie
10.
Reprod Fertil Dev ; 30(1): 73-84, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29539305

RESUMEN

Mammalian embryo development is characterised by regulative mechanisms of lineage segregation and cell specification. A combination of carefully orchestrated gene expression networks, signalling pathways and epigenetic marks defines specific developmental stages that can now be resolved at the single-cell level. These new ways to depict developmental processes have the potential to provide answers to unresolved questions on how lineage allocation and cell fate decisions are made during embryogenesis. Over the past few years, a flurry of studies reporting detailed single-cell transcription profiles in early embryos has complemented observations acquired using live cell imaging following gene editing techniques to manipulate specific genes. The adoption of this newly available toolkit is reshaping how researchers are designing experiments and how they view animal development. This review presents an overview of the current knowledge on lineage segregation and cell specification in mammals, and discusses some of the outstanding questions that current technological advances can help scientists address, like never before.


Asunto(s)
Blastocisto/metabolismo , Diferenciación Celular/genética , Desarrollo Embrionario/genética , Epigénesis Genética/fisiología , Animales , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica , Humanos
11.
Adv Exp Med Biol ; 953: 383-440, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27975276

RESUMEN

Two unique characteristics of the germ line are the ability to persist from generation to generation and to retain full developmental potential while differentiating into gametes. How the germ line is specified that allows it to retain these characteristics within the context of a developing embryo remains unknown and is one focus of current research. Germ cell specification proceeds through one of two basic mechanisms: cell autonomous or inductive. Here, we discuss how germ plasm driven germ cell specification (cell autonomous) occurs in both zebrafish and the frog Xenopus. We describe the segregation of germ cells during embryonic development of solitary and colonial ascidians to provide an evolutionary context to both mechanisms. We conclude with a discussion of the inductive mechanism as exemplified by both the mouse and axolotl model systems. Regardless of mechanism, several general themes can be recognized including the essential role of repression and posttranscriptional regulation of gene expression.


Asunto(s)
Diferenciación Celular/genética , Desarrollo Embrionario/genética , Redes Reguladoras de Genes/genética , Células Germinativas/crecimiento & desarrollo , Animales , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/metabolismo , Ratones , Xenopus/crecimiento & desarrollo , Xenopus laevis/crecimiento & desarrollo , Pez Cebra/crecimiento & desarrollo
12.
Dev Biol ; 387(1): 15-27, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24445281

RESUMEN

The crosstalk between the epiblast and the trophoblast is critical in supporting the early stages of conceptus development. FGF4 and BMP4 are inductive signals that participate in the communication between the epiblast and the extraembryonic ectoderm (ExE) of the developing mouse embryo. Importantly, however, it is unknown whether a similar crosstalk operates in species that lack a discernible ExE and develop a mammotypical embryonic disc (ED). Here we investigated the crosstalk between the epiblast and the trophectoderm (TE) during pig embryo elongation. FGF4 ligand and FGFR2 were detected primarily on the plasma membrane of TE cells of peri-elongation embryos. The binding of this growth factor to its receptor triggered a signal transduction response evidenced by an increase in phosphorylated MAPK/ERK. Particular enrichment was detected in the periphery of the ED in early ovoid embryos, indicating that active FGF signalling was operating during this stage. Gene expression analysis shows that CDX2 and ELF5, two genes expressed in the mouse ExE, are only co-expressed in the Rauber's layer, but not in the pig mural TE. Interestingly, these genes were detected in the nascent mesoderm of early gastrulating embryos. Analysis of BMP4 expression by in situ hybridisation shows that this growth factor is produced by nascent mesoderm cells. A functional test in differentiating epiblast shows that CDX2 and ELF5 are activated in response to BMP4. Furthermore, the effects of BMP4 were also demonstrated in the neighbouring TE cells, as demonstrated by an increase in phosphorylated SMAD1/5/8. These results show that BMP4 produced in the extraembryonic mesoderm is directly influencing the SMAD response in the TE of elongating embryos. These results demonstrate that paracrine signals from the embryo, represented by FGF4 and BMP4, induce a response in the TE prior to the extensive elongation. The study also confirms that expression of CDX2 and ELF5 is not conserved in the mural TE, indicating that although the signals that coordinate conceptus growth are similar between rodents and pigs, the gene regulatory network of the trophoblast lineage is not conserved in these species.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Proteínas de Unión al ADN/metabolismo , Factor 4 de Crecimiento de Fibroblastos/metabolismo , Proteínas de Homeodominio/metabolismo , Porcinos/embriología , Factores de Transcripción/metabolismo , Animales , Factor de Transcripción CDX2 , Diferenciación Celular/genética , Membrana Celular/metabolismo , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Gastrulación , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Estratos Germinativos/metabolismo , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Comunicación Paracrina/genética , Fosforilación , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Proteína Smad8/metabolismo , Transcripción Genética , Trofoblastos/metabolismo
13.
Biol Reprod ; 92(4): 103, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25788662

RESUMEN

Oocytes treated with the protein synthesis inhibitor cycloheximide (CHX) arrest at the germinal vesicle (GV) stage and undergo accelerated GV breakdown (GVBD) after CHX is removed. However, little is known about the underlying mechanism of accelerated meiotic maturation. Here, we investigated this mechanism and found that oocytes released from CHX arrest have higher amounts of cyclin B1 (CCNB1) and phosphorylated mitogen-activated protein kinase (pMAPK) proteins. Increased levels of these factors were not associated with mRNA polyadenylation or increased transcription rates of CCNB1 and MOS (Moloney murine sarcoma viral oncogene homolog) during CHX arrest. We found that treatment of CHX-arrested oocytes with the actin filament-stabilizing agent Jasplakinolide (Jasp) delayed GVBD following release from CHX arrest and that this was correlated with reduced maturation-promoting factor (MPF) activity. These results suggest that CCNB1 mRNAs released from actin filaments during CHX arrest increase CCNB1 transcripts available for translation after release from CHX arrest, leading to the precocious activation of MPF and accelerated meiotic progression.


Asunto(s)
Actinas/metabolismo , Cicloheximida/farmacología , Meiosis/efectos de los fármacos , Oocitos/efectos de los fármacos , Inhibidores de la Síntesis de la Proteína/farmacología , Animales , Ciclo Celular/efectos de los fármacos , Ciclina B1/metabolismo , Depsipéptidos/farmacología , Femenino , Factor Promotor de Maduración/farmacología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Virus del Sarcoma Murino de Moloney/genética , Técnicas de Transferencia Nuclear , Polimerizacion , Embarazo , Ovinos
14.
Stem Cell Res Ther ; 15(1): 128, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38693576

RESUMEN

BACKGROUND: Testicular germ cell tumours (TGCTs) represent a clinical challenge; they are most prevalent in young individuals and are triggered by molecular mechanisms that are not fully understood. The origin of TGCTs can be traced back to primordial germ cells that fail to mature during embryonic development. These cells express high levels of pluripotency factors, including the transcription factor NANOG which is highly expressed in TGCTs. Gain or amplification of the NANOG locus is common in advanced tumours, suggesting a key role for this master regulator of pluripotency in TGCT stemness and malignancy. METHODS: In this study, we analysed the expression of microRNAs (miRNAs) that are regulated by NANOG in TGCTs via integrated bioinformatic analyses of data from The Cancer Genome Atlas and NANOG chromatin immunoprecipitation in human embryonic stem cells. Through gain-of-function experiments, MIR9-2 was further investigated as a novel tumour suppressor regulated by NANOG. After transfection with MIR9-2 mimics, TGCT cells were analysed for cell proliferation, invasion, sensitivity to cisplatin, and gene expression signatures by RNA sequencing. RESULTS: For the first time, we identified 86 miRNAs regulated by NANOG in TGCTs. Among these, 37 miRNAs were differentially expressed in NANOG-high tumours, and they clustered TGCTs according to their subtypes. Binding of NANOG within 2 kb upstream of the MIR9-2 locus was associated with a negative regulation. Low expression of MIR9-2 was associated with tumour progression and MIR9-2-5p was found to play a role in the control of tumour stemness. A gain of function of MIR9-2-5p was associated with reduced proliferation, invasion, and sensitivity to cisplatin in both embryonal carcinoma and seminoma tumours. MIR9-2-5p expression in TGCT cells significantly reduced the expression of genes regulating pluripotency and cell division, consistent with its functional effect on reducing cancer stemness. CONCLUSIONS: This study provides new molecular insights into the role of NANOG as a key determinant of pluripotency in TGCTs through the regulation of MIR9-2-5p, a novel epigenetic modulator of cancer stemness. Our data also highlight the potential negative feedback mediated by MIR9-2-5p on NANOG expression, which could be exploited as a therapeutic strategy for the treatment of TGCTs.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , MicroARNs , Proteína Homeótica Nanog , Neoplasias de Células Germinales y Embrionarias , Neoplasias Testiculares , Humanos , Proteína Homeótica Nanog/metabolismo , Proteína Homeótica Nanog/genética , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias de Células Germinales y Embrionarias/genética , Neoplasias de Células Germinales y Embrionarias/metabolismo , Neoplasias de Células Germinales y Embrionarias/patología , Neoplasias Testiculares/patología , Neoplasias Testiculares/metabolismo , Neoplasias Testiculares/genética , Masculino , Línea Celular Tumoral , Proliferación Celular/genética , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Cisplatino/farmacología
15.
Nat Commun ; 15(1): 5210, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38890321

RESUMEN

Cell-fate decisions during mammalian gastrulation are poorly understood outside of rodent embryos. The embryonic disc of pig embryos mirrors humans, making them a useful proxy for studying gastrulation. Here we present a single-cell transcriptomic atlas of pig gastrulation, revealing cell-fate emergence dynamics, as well as conserved and divergent gene programs governing early porcine, primate, and murine development. We highlight heterochronicity in extraembryonic cell-types, despite the broad conservation of cell-type-specific transcriptional programs. We apply these findings in combination with functional investigations, to outline conserved spatial, molecular, and temporal events during definitive endoderm specification. We find early FOXA2 + /TBXT- embryonic disc cells directly form definitive endoderm, contrasting later-emerging FOXA2/TBXT+ node/notochord progenitors. Unlike mesoderm, none of these progenitors undergo epithelial-to-mesenchymal transition. Endoderm/Node fate hinges on balanced WNT and hypoblast-derived NODAL, which is extinguished upon endodermal differentiation. These findings emphasise the interplay between temporal and topological signalling in fate determination during gastrulation.


Asunto(s)
Embrión de Mamíferos , Endodermo , Gastrulación , Regulación del Desarrollo de la Expresión Génica , Análisis de la Célula Individual , Animales , Endodermo/citología , Endodermo/metabolismo , Endodermo/embriología , Porcinos , Ratones , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Diferenciación Celular , Mesodermo/citología , Mesodermo/embriología , Mesodermo/metabolismo , Transcriptoma , Factor Nuclear 3-beta del Hepatocito/metabolismo , Factor Nuclear 3-beta del Hepatocito/genética , Linaje de la Célula , Proteínas de Dominio T Box/metabolismo , Proteínas de Dominio T Box/genética , Transición Epitelial-Mesenquimal/genética
16.
Development ; 137(18): 2973-80, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20736286

RESUMEN

Cells in the pluripotent ground state can give rise to somatic cells and germ cells, and the acquisition of pluripotency is dependent on the expression of Nanog. Pluripotency is conserved in the primitive ectoderm of embryos from mammals and urodele amphibians, and here we report the isolation of a Nanog ortholog from axolotls (axNanog). axNanog does not contain a tryptophan repeat domain and is expressed as a monomer in the axolotl animal cap. The monomeric form is sufficient to regulate pluripotency in mouse embryonic stem cells, but axNanog dimers are required to rescue LIF-independent self-renewal. Our results show that protein interactions mediated by Nanog dimerization promote proliferation. More importantly, they demonstrate that the mechanisms governing pluripotency are conserved from urodele amphibians to mammals.


Asunto(s)
Células Madre Embrionarias/metabolismo , Proteínas de Homeodominio/metabolismo , Células Madre Pluripotentes/metabolismo , Ambystoma mexicanum , Anfibios , Animales , Línea Celular , Proliferación Celular , Proteínas de Homeodominio/genética , Humanos , Mamíferos , Ratones , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Unión Proteica
17.
BMC Biol ; 10: 90, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23153189

RESUMEN

BACKGROUND: This work describes the first genome-wide analysis of the transcriptional landscape of the pig. A new porcine Affymetrix expression array was designed in order to provide comprehensive coverage of the known pig transcriptome. The new array was used to generate a genome-wide expression atlas of pig tissues derived from 62 tissue/cell types. These data were subjected to network correlation analysis and clustering. RESULTS: The analysis presented here provides a detailed functional clustering of the pig transcriptome where transcripts are grouped according to their expression pattern, so one can infer the function of an uncharacterized gene from the company it keeps and the locations in which it is expressed. We describe the overall transcriptional signatures present in the tissue atlas, where possible assigning those signatures to specific cell populations or pathways. In particular, we discuss the expression signatures associated with the gastrointestinal tract, an organ that was sampled at 15 sites along its length and whose biology in the pig is similar to human. We identify sets of genes that define specialized cellular compartments and region-specific digestive functions. Finally, we performed a network analysis of the transcription factors expressed in the gastrointestinal tract and demonstrate how they sub-divide into functional groups that may control cellular gastrointestinal development. CONCLUSIONS: As an important livestock animal with a physiology that is more similar than mouse to man, we provide a major new resource for understanding gene expression with respect to the known physiology of mammalian tissues and cells. The data and analyses are available on the websites http://biogps.org and http://www.macrophages.com/pig-atlas.


Asunto(s)
Bases de Datos Genéticas , Regulación de la Expresión Génica/fisiología , Genoma , Porcinos/genética , Animales , Análisis por Conglomerados , Perfilación de la Expresión Génica , Especificidad de Órganos , Transcriptoma
18.
Emerg Top Life Sci ; 7(4): 397-408, 2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-37933589

RESUMEN

Gastrulation represents a pivotal phase of development and aberrations during this period can have major consequences, from minor anatomical deviations to severe congenital defects. Animal models are used to study gastrulation, however, there is considerable morphological and molecular diversity of gastrula across mammalian species. Here, we provide an overview of the latest research on interspecies developmental control across mammals. This includes single-cell atlases of several mammalian gastrula which have enabled comparisons of the temporal and molecular dynamics of differentiation. These studies highlight conserved cell differentiation regulators and both absolute and relative differences in differentiation dynamics between species. Recent advances in in vitro culture techniques have facilitated the derivation, maintenance and differentiation of cell lines from a range of species and the creation of multi-species models of gastrulation. Gastruloids are three-dimensional aggregates capable of self-organising and recapitulating aspects of gastrulation. Such models enable species comparisons outside the confines of the embryo. We highlight recent in vitro evidence that differentiation processes such as somitogenesis and neuronal maturation scale with known in vivo differences in developmental tempo across species. This scaling is likely due to intrinsic differences in cell biochemistry. We also highlight several studies which provide examples of cell differentiation dynamics being influenced by extrinsic factors, including culture conditions, chimeric co-culture, and xenotransplantation. These collective studies underscore the complexity of gastrulation across species, highlighting the necessity of additional datasets and studies to decipher the intricate balance between intrinsic cellular programs and extrinsic signals in shaping embryogenesis.


Asunto(s)
Gástrula , Gastrulación , Animales , Diferenciación Celular/fisiología , Embrión de Mamíferos/metabolismo , Gástrula/metabolismo , Mamíferos
19.
BMC Dev Biol ; 11: 11, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21352525

RESUMEN

BACKGROUND: Epigenetic reprogramming is critical for genome regulation during germ line development. Genome-wide demethylation in mouse primordial germ cells (PGC) is a unique reprogramming event essential for erasing epigenetic memory and preventing the transmission of epimutations to the next generation. In addition to DNA demethylation, PGC are subject to a major reprogramming of histone marks, and many of these changes are concurrent with a cell cycle arrest in the G2 phase. There is limited information on how well conserved these events are in mammals. Here we report on the dynamic reprogramming of DNA methylation at CpGs of imprinted loci and DNA repeats, and the global changes in H3K27me3 and H3K9me2 in the developing germ line of the domestic pig. RESULTS: Our results show loss of DNA methylation in PGC colonizing the genital ridges. Analysis of IGF2-H19 regulatory region showed a gradual demethylation between E22-E42. In contrast, DMR2 of IGF2R was already demethylated in male PGC by E22. In females, IGF2R demethylation was delayed until E29-31, and was de novo methylated by E42. DNA repeats were gradually demethylated from E25 to E29-31, and became de novo methylated by E42. Analysis of histone marks showed strong H3K27me3 staining in migratory PGC between E15 and E21. In contrast, H3K9me2 signal was low in PGC by E15 and completely erased by E21. Cell cycle analysis of gonadal PGC (E22-31) showed a typical pattern of cycling cells, however, migrating PGC (E17) showed an increased proportion of cells in G2. CONCLUSIONS: Our study demonstrates that epigenetic reprogramming occurs in pig migratory and gonadal PGC, and establishes the window of time for the occurrence of these events. Reprogramming of histone H3K9me2 and H3K27me3 detected between E15-E21 precedes the dynamic DNA demethylation at imprinted loci and DNA repeats between E22-E42. Our findings demonstrate that major epigenetic reprogramming in the pig germ line follows the overall dynamics shown in mice, suggesting that epigenetic reprogramming of germ cells is conserved in mammals. A better understanding of the sequential reprogramming of PGC in the pig will facilitate the derivation of embryonic germ cells in this species.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Células Germinativas/metabolismo , Histona Demetilasas/metabolismo , Oxidorreductasas N-Desmetilantes/metabolismo , Porcinos/embriología , Animales , Ciclo Celular , Islas de CpG , Femenino , Citometría de Flujo , Impresión Genómica , Histonas/metabolismo , Masculino , Familia de Multigenes , Mutación , Factor 3 de Transcripción de Unión a Octámeros/genética , Reacción en Cadena de la Polimerasa , Receptor IGF Tipo 2/genética , Homología de Secuencia , Elementos de Nucleótido Esparcido Corto , Porcinos/genética
20.
Mol Cancer ; 10(1): 7, 2011 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-21232089

RESUMEN

BACKGROUND: Breast cancer is a disease characterised by both genetic and epigenetic alterations. Epigenetic silencing of tumour suppressor genes is an early event in breast carcinogenesis and reversion of gene silencing by epigenetic reprogramming can provide clues to the mechanisms responsible for tumour initiation and progression. In this study we apply the reprogramming capacity of oocytes to cancer cells in order to study breast oncogenesis. RESULTS: We show that breast cancer cells can be directly reprogrammed by amphibian oocyte extracts. The reprogramming effect, after six hours of treatment, in the absence of DNA replication, includes DNA demethylation and removal of repressive histone marks at the promoters of tumour suppressor genes; also, expression of the silenced genes is re-activated in response to treatment. This activity is specific to oocytes as it is not elicited by extracts from ovulated eggs, and is present at very limited levels in extracts from mouse embryonic stem cells. Epigenetic reprogramming in oocyte extracts results in reduction of cancer cell growth under anchorage independent conditions and a reduction in tumour growth in mouse xenografts. CONCLUSIONS: This study presents a new method to investigate tumour reversion by epigenetic reprogramming. After testing extracts from different sources, we found that axolotl oocyte extracts possess superior reprogramming ability, which reverses epigenetic silencing of tumour suppressor genes and tumorigenicity of breast cancer cells in a mouse xenograft model. Therefore this system can be extremely valuable for dissecting the mechanisms involved in tumour suppressor gene silencing and identifying molecular activities capable of arresting tumour growth. These applications can ultimately shed light on the contribution of epigenetic alterations in breast cancer and advance the development of epigenetic therapies.


Asunto(s)
Ambystoma mexicanum , Neoplasias de la Mama/genética , Extractos Celulares/farmacología , Oocitos/química , Xenopus , Animales , Neoplasias de la Mama/patología , Permeabilidad de la Membrana Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensamble y Desensamble de Cromatina , Digitonina/farmacología , Células Madre Embrionarias/química , Epigénesis Genética , Femenino , Perfilación de la Expresión Génica , Genes Supresores de Tumor , Histonas/metabolismo , Humanos , Metilación , Ratones , Proteínas de Microfilamentos/genética , Trasplante de Neoplasias , Regiones Promotoras Genéticas , Activación Transcripcional , Trasplante Heterólogo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA