Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Endocrinol ; 244(2): 395-413, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31829965

RESUMEN

The embryonic Müllerian ducts give rise to the female reproductive tract (fallopian tubes, uterus and upper vagina in humans, the oviducts in birds). Embryonic Müllerian ducts initially develop in both sexes, but later regress in males under the influence of anti-Müllerian hormone. While the molecular and endocrine control of duct regression in males have been well studied, early development of the ducts in both sexes is less well understood. Here, we describe a novel role for the adhesion G protein-coupled receptor, GPR56, in development of the Müllerian ducts in the chicken embryo. GPR56 is expressed in the ducts of both sexes from early stages. The mRNA is present during the elongation phase of duct formation, and it is restricted to the inner Müllerian duct epithelium. The putative ligand, Collagen III, is abundantly expressed in the Müllerian duct at the same developmental stages. Knockdown of GPR56 expression using in ovo electroporation results in variably truncated ducts, with a loss of expression of both epithelial and mesenchymal markers of duct development. Over-expression of GPR56 in vitro results in enhanced cell proliferation and cell migration. These results show that GPR56 plays an essential role in avian Müllerian duct development through the regulation of duct elongation.


Asunto(s)
Proteínas Aviares/metabolismo , Pollos/metabolismo , Conductos Paramesonéfricos/embriología , Conductos Paramesonéfricos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Proteínas Aviares/genética , Proliferación Celular , Embrión de Pollo , Pollos/genética , Pollos/crecimiento & desarrollo , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Receptores Acoplados a Proteínas G/genética
2.
Endocrinology ; 159(10): 3492-3502, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30124802

RESUMEN

Birds have a ZZ male and ZW female sex chromosome system. The relative roles of genetics and hormones in regulating avian sexual development have been revealed by studies on gynandromorphs. Gynandromorphs are rare bilateral sex chimeras, male on one side of the body and female on the other. We examined a naturally occurring gynandromorphic chicken that was externally male on the right side of the body and female on the left. The bird was diploid but with a mix of ZZ and ZW cells that correlated with the asymmetric sexual phenotype. The male side was 96% ZZ, and the female side was 77% ZZ and 23% ZW. The gonads of this bird at sexual maturity were largely testicular. The right gonad was a testis, with SOX9+ Sertoli cells, DMRT1+ germ cells, and active spermatogenesis. The left gonad was primarily testicular, but with some peripheral aromatase-expressing follicles. The bird had low levels of serum estradiol and high levels of testosterone, as expected for a male. Despite the low percentage of ZW cells on that side, the left side had female sex-linked feathering, smaller muscle mass, smaller leg and spur, and smaller wattle than the male side. This indicates that these sexually dimorphic structures must be at least partly independent of sex steroid effects. Even a small percentage of ZW cells appears sufficient to support female sexual differentiation. Given the lack of chromosome-wide dosage compensation in birds, various sexually dimorphic features may arise due to Z-gene dosage differences between the sexes.


Asunto(s)
Trastornos del Desarrollo Sexual/metabolismo , Sistema Endocrino/metabolismo , Gónadas/metabolismo , Análisis para Determinación del Sexo/métodos , Animales , Proteínas Aviares/genética , Proteínas Aviares/metabolismo , Pollos , Trastornos del Desarrollo Sexual/genética , Femenino , Genotipo , Gónadas/citología , Cariotipificación , Masculino , Fenotipo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Cromosomas Sexuales/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
Int J Dev Biol ; 62(1-2-3): 153-166, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29616724

RESUMEN

Our understanding of avian sex determination and gonadal development is derived primarily from the studies in the chicken. Analysis of gynandromorphic chickens and experimental chimeras indicate that sexual phenotype is at least partly cell autonomous in the chicken, with sexually dimorphic gene expression occurring in different tissue and different stages. Gonadal sex differentiation is just one of the many manifestations of sexual phenotype. As in other birds, the chicken has a ZZ male: ZW female sex chromosome system, in which the male is the homogametic sex. Most evidence favours a Z chromosome dosage mechanism underling chicken sex determination, with little evidence of a role for the W chromosome. Indeed, the W appears to harbour a small number of genes that are un-related to sexual development, but have been retained because they are dosage sensitive factors. As global Z dosage compensation is absent in birds, Z-linked genes may direct sexual development in different tissues (males having on average 1.5 to 2 times the expression level of females). In the embryonic gonads, the Z-linked DMRT1 gene plays a key role in testis development. Beyond the gonads, other combinations of Z-linked genes may govern sexual development, together with a role for sex steroid hormones. Gonadal DMRT1 is thought to activate other players in testis development, namely SOX9 and AMH, and the recently identified HEMGN gene. DMRT1 also represses ovarian pathway genes, such as FOXL2 and CYP19A1. A lower level of DMRT1 expression in the female gonads is compatible with activation of the ovarian pathway. Some outstanding questions include how the key testis and ovary genes, DMRT1 and FOXL2, are regulated. In addition, confirmation of the central role of these genes awaits genome editing approaches.


Asunto(s)
Embrión de Pollo , Cromosomas Sexuales , Procesos de Determinación del Sexo , Diferenciación Sexual/genética , Animales , Aromatasa/metabolismo , Diferenciación Celular , Pollos , Compensación de Dosificación (Genética) , Femenino , Dosificación de Gen , Regulación del Desarrollo de la Expresión Génica , Genoma , Gónadas/embriología , Masculino , Ovario/embriología , Factor de Transcripción SOX9/metabolismo , Testículo/embriología
4.
Endocrinology ; 158(9): 2970-2987, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28911174

RESUMEN

The exact genetic mechanism regulating avian gonadal sex differentiation has not been completely resolved. The most likely scenario involves a dosage mechanism, whereby the Z-linked DMRT1 gene triggers testis development. However, the possibility still exists that the female-specific W chromosome may harbor an ovarian determining factor. In this study, we provide evidence that the universal gene regulating gonadal sex differentiation in birds is Z-linked DMRT1 and not a W-linked (ovarian) factor. Three candidate W-linked ovarian determinants are HINTW, female-expressed transcript 1 (FET1), and female-associated factor (FAF). To test the association of these genes with ovarian differentiation in the chicken, we examined their expression following experimentally induced female-to-male sex reversal using the aromatase inhibitor fadrozole (FAD). Administration of FAD on day 3 of embryogenesis induced a significant loss of aromatase enzyme activity in female gonads and masculinization. However, expression levels of HINTW, FAF, and FET1 were unaltered after experimental masculinization. Furthermore, comparative analysis showed that FAF and FET1 expression could not be detected in zebra finch gonads. Additionally, an antibody raised against the predicted HINTW protein failed to detect it endogenously. These data do not support a universal role for these genes or for the W sex chromosome in ovarian development in birds. We found that DMRT1 (but not the recently identified Z-linked HEMGN gene) is male upregulated in embryonic zebra finch and emu gonads, as in the chicken. As chicken, zebra finch, and emu exemplify the major evolutionary clades of birds, we propose that Z-linked DMRT1, and not the W sex chromosome, regulates gonadal sex differentiation in birds.


Asunto(s)
Aves , Cromosomas Sexuales/genética , Procesos de Determinación del Sexo/genética , Diferenciación Sexual/genética , Factores de Transcripción/fisiología , Animales , Aves/embriología , Aves/genética , Embrión de Pollo , Pollos , Desarrollo Embrionario , Femenino , Feminización/embriología , Feminización/genética , Pinzones/embriología , Pinzones/genética , Regulación del Desarrollo de la Expresión Génica , Gónadas , Masculino
5.
Methods Mol Biol ; 1650: 177-190, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28809021

RESUMEN

One of the advantages of the avian embryo as an experimental model is its in ovo development and hence accessibility for genetic manipulation. Electroporation has been used extensively in the past to study gene function in chicken and quail embryos . Readily accessible tissues such as the neural tube, somites, and limb bud, in particular, have been targeted. However, more inaccessible tissues, such as the embryonic urogenital system , have proven more challenging to study. Here, we describe the use of in ovo electroporation of TOL2 vectors or RCASBP avian viral vectors for the rapid functional analysis of genes involved in avian sex determination and urogenital development . In the context of the developing urogenital system , these vectors have inherent advantages and disadvantages, which will be considered here. Either vector can both be used for mis-expressing a gene and for targeting endogenous gene knockdown via expression of short hairpin RNAs (shRNAs). Both of these vectors integrate into the genome and are hence spread throughout developing tissues. Going forward, electroporation could be combined with CRISPR/Cas9 technology for targeted genome editing in the avian urogenital system .


Asunto(s)
Embrión de Pollo , Pollos/genética , Electroporación/métodos , Técnicas de Transferencia de Gen , Vectores Genéticos , Sistema Urogenital/fisiología , Animales , Elementos Transponibles de ADN , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen
6.
Elife ; 52016 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-27218451

RESUMEN

How cells in the embryo coordinate epithelial plasticity with cell fate decision in a fast changing cellular environment is largely unknown. In chick embryos, skeletal muscle formation is initiated by migrating Delta1-expressing neural crest cells that trigger NOTCH signaling and myogenesis in selected epithelial somite progenitor cells, which rapidly translocate into the nascent muscle to differentiate. Here, we uncovered at the heart of this response a signaling module encompassing NOTCH, GSK-3ß, SNAI1 and ß-catenin. Independent of its transcriptional function, NOTCH profoundly inhibits GSK-3ß activity. As a result SNAI1 is stabilized, triggering an epithelial to mesenchymal transition. This allows the recruitment of ß-catenin from the membrane, which acts as a transcriptional co-factor to activate myogenesis, independently of WNT ligand. Our results intimately associate the initiation of myogenesis to a change in cell adhesion and may reveal a general principle for coupling cell fate changes to EMT in many developmental and pathological processes.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Células Musculares/metabolismo , Desarrollo de Músculos/genética , Músculo Esquelético/metabolismo , Receptores Notch/genética , beta Catenina/genética , Animales , Adhesión Celular , Diferenciación Celular , Membrana Celular/metabolismo , Embrión de Pollo , Citoplasma/metabolismo , Regulación del Desarrollo de la Expresión Génica , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Células Musculares/citología , Músculo Esquelético/citología , Músculo Esquelético/crecimiento & desarrollo , Cresta Neural/citología , Cresta Neural/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo , Somitos/citología , Somitos/metabolismo , beta Catenina/metabolismo
7.
Dev Biol ; 407(1): 68-74, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26277216

RESUMEN

Gene-targeted knockout technologies are invaluable tools for understanding the functions of genes in vivo. CRISPR/Cas9 system of RNA-guided genome editing is revolutionizing genetics research in a wide spectrum of organisms. Here, we combined CRISPR with in vivo electroporation in the chicken embryo to efficiently target the transcription factor PAX7 in tissues of the developing embryo. This approach generated mosaic genetic mutations within a wild-type cellular background. This series of proof-of-principle experiments indicate that in vivo CRISPR-mediated cell genome engineering is an effective method to achieve gene loss-of-function in the tissues of the chicken embryo and it completes the growing genetic toolbox to study the molecular mechanisms regulating development in this important animal model.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/fisiología , Ingeniería Genética , Animales , Secuencia de Bases , Embrión de Pollo , Pollos , Electroporación , Genoma , Datos de Secuencia Molecular , Factor de Transcripción PAX7/fisiología
8.
Results Probl Cell Differ ; 56: 99-122, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25344668

RESUMEN

This review will focus on the use of the chicken and quail as model systems to analyze myogenesis and as such will emphasize the experimental approaches that are strongest in these systems-the amenability of the avian embryo to manipulation and in ovo observation. During somite differentiation, a wide spectrum of developmental processes occur such as cellular differentiation, migration, and fusion. Cell lineage studies combined with recent advancements in cell imaging allow these biological phenomena to be readily observed and hypotheses tested extremely rapidly-a strength that is restricted to the avian system. A clear weakness of the chicken in the past has been genetic approaches to modulate gene function. Recent advances in the electroporation of expression vectors, siRNA constructs, and use of tissue specific reporters have opened the door to increasingly sophisticated experiments that address questions of interest not only to the somite/muscle field in particular but also fundamental to biology in general. Importantly, an ever-growing body of evidence indicates that somite differentiation in birds is indistinguishable to that of mammals; therefore, these avian studies complement the complex genetic models of the mouse.


Asunto(s)
Diferenciación Celular/genética , Modelos Biológicos , Desarrollo de Músculos/genética , Somitos/crecimiento & desarrollo , Animales , Linaje de la Célula/genética , Embrión de Pollo , Regulación del Desarrollo de la Expresión Génica , Ratones , Codorniz/crecimiento & desarrollo
9.
Stem Cell Reports ; 1(1): 53-65, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24052942

RESUMEN

We investigated the role of canonical WNT signaling in mesoderm and hematopoietic development from human embryonic stem cells (hESCs) using a recombinant human protein-based differentiation medium (APEL). In contrast to prior studies using less defined culture conditions, we found that WNT3A alone was a poor inducer of mesoderm. However, WNT3A synergized with BMP4 to accelerate mesoderm formation, increase embryoid body size, and increase the number of hematopoietic blast colonies. Interestingly, inclusion of WNT3A or a GSK3 inhibitor in methylcellulose colony-forming assays at 4 days of differentiation abrogated blast colony formation but supported the generation of mesospheres that expressed genes associated with mesenchymal lineages. Mesospheres differentiated into cells with characteristics of bone, fat, and smooth muscle. These studies identify distinct effects for WNT3A, supporting the formation of hematopoietic or mesenchymal lineages from human embryonic stem cells, depending upon differentiation stage at the time of exposure.


Asunto(s)
Cuerpos Embrioides/citología , Hematopoyesis , Células Madre Mesenquimatosas/citología , Proteína Wnt3A/metabolismo , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Linaje de la Célula , Células Cultivadas , Cuerpos Embrioides/metabolismo , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Humanos , Células Madre Mesenquimatosas/metabolismo , Mesodermo/citología , Mesodermo/embriología , Mesodermo/metabolismo , Vía de Señalización Wnt , Proteína Wnt3A/antagonistas & inhibidores , Proteína Wnt3A/genética
10.
Stem Cell Res ; 10(1): 103-17, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23164599

RESUMEN

The limited availability of human vascular endothelial cells (ECs) hampers research into EC function whilst the lack of precisely defined culture conditions for this cell type presents problems for addressing basic questions surrounding EC physiology. We aimed to generate endothelial progenitors from human pluripotent stem cells to facilitate the study of human EC physiology, using a defined serum-free protocol. Human embryonic stem cells (hESC-ECs) differentiated under serum-free conditions generated CD34(+)KDR(+) endothelial progenitor cells after 6days that could be further expanded in the presence of vascular endothelial growth factor (VEGF). The resultant EC population expressed CD31 and TIE2/TEK, took up acetylated low-density lipoprotein (LDL) and up-regulated expression of ICAM-1, PAI-1 and ET-1 following treatment with TNFα. Immunofluorescence studies indicated that a key mediator of vascular tone, endothelial nitric oxide synthase (eNOS), was localised to a perinuclear compartment of hESC-ECs, in contrast with the pan-cellular distribution of this enzyme within human umbilical vein ECs (HUVECs). Further investigation revealed that that the serum-associated lipids, lysophosphatidic acid (LPA) and platelet activating factor (PAF), were the key molecules that affected eNOS localisation in hESC-ECs cultures. These studies illustrate the feasibility of EC generation from hESCs and the utility of these cells for investigating environmental cues that impact on EC phenotype. We have demonstrated a hitherto unrecognized role for LPA and PAF in the regulation of eNOS subcellular localization.


Asunto(s)
Medios de Cultivo/farmacología , Células Madre Embrionarias/efectos de los fármacos , Células Endoteliales/citología , Lisofosfolípidos/farmacología , Óxido Nítrico Sintasa de Tipo III/análisis , Factor de Activación Plaquetaria/farmacología , Antígenos CD34/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Colágeno/química , Combinación de Medicamentos , Células Madre Embrionarias/citología , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Laminina/química , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteoglicanos/química , Factor de Necrosis Tumoral alfa/farmacología
11.
Stem Cell Res Ther ; 3(4): 31, 2012 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-22897941

RESUMEN

Ischaemia-related diseases such as peripheral artery disease and coronary heart disease constitute a major issue in medicine as they affect millions of individuals each year and represent a considerable economic burden to healthcare systems. If the underlying ischaemia is not sufficiently resolved it can lead to tissue damage, with subsequent cell death. Treating such diseases remains difficult and several strategies have been used to stimulate the growth of blood vessels and promote regeneration of ischaemic tissues, such as the use of recombinant proteins and gene therapy. Although these approaches remain promising, they have limitations and results from clinical trials using these methods have had limited success. Recently, there has been growing interest in the therapeutic potential of using a cell-based approach to treat vasodegenerative disorders. In vascular medicine, various stem cells and adult progenitors have been highlighted as having a vasoreparative role in ischaemic tissues. This review will examine the clinical potential of several stem and progenitor cells that may be utilised to regenerate defunct or damaged vasculature and restore blood flow to the ischaemic tissue. In particular, we focus on the therapeutic potential of endothelial progenitor cells as an exciting new option for the treatment of ischaemic diseases.


Asunto(s)
Isquemia/terapia , Trasplante de Células Madre , Células Madre/citología , Células Madre Adultas/citología , Tratamiento Basado en Trasplante de Células y Tejidos , Endotelio Vascular/citología , Humanos , Neovascularización Fisiológica , Células Madre Pluripotentes/citología
12.
Blood ; 119(26): 6243-54, 2012 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-22611158

RESUMEN

Transcriptional profiling of differentiating human embryonic stem cells (hESCs) revealed that MIXL1-positive mesodermal precursors were enriched for transcripts encoding the G-protein-coupled APELIN receptor (APLNR). APLNR-positive cells, identified by binding of the fluoresceinated peptide ligand, APELIN (APLN), or an anti-APLNR mAb, were found in both posterior mesoderm and anterior mesendoderm populations and were enriched in hemangioblast colony-forming cells (Bl-CFC). The addition of APLN peptide to the media enhanced the growth of embryoid bodies (EBs), increased the expression of hematoendothelial genes in differentiating hESCs, and increased the frequency of Bl-CFCs by up to 10-fold. Furthermore, APLN peptide also synergized with VEGF to promote the growth of hESC-derived endothelial cells. These studies identified APLN as a novel growth factor for hESC-derived hematopoietic and endothelial cells.


Asunto(s)
Células Madre Embrionarias/efectos de los fármacos , Hematopoyesis/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Apelina , Receptores de Apelina , Células Cultivadas , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/fisiología , Endodermo/efectos de los fármacos , Endodermo/metabolismo , Endodermo/fisiología , Perfilación de la Expresión Génica , Hemangioblastos/efectos de los fármacos , Hemangioblastos/metabolismo , Hemangioblastos/fisiología , Hematopoyesis/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/fisiología , Mesodermo/citología , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , Mesodermo/fisiología , Análisis por Micromatrices , Modelos Biológicos , Unión Proteica/efectos de los fármacos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
13.
Stem Cell Res ; 8(2): 165-79, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22265737

RESUMEN

The Mixl1 homeodomain protein plays a key role in mesendoderm patterning during embryogenesis, but its target genes remain to be identified. We compared gene expression in differentiating heterozygous Mixl1(GFP/w) and homozygous null Mixl1(GFP/Hygro) mouse embryonic stem cells to identify potential downstream transcriptional targets of Mixl1. Candidate Mixl1 regulated genes whose expression was reduced in GFP+ cells isolated from differentiating Mixl1(GFP/Hygro) embryoid bodies included Pdgfrα and Flk1. Mixl1 bound to ATTA sequences located in the Pdgfrα and Flk1 promoters and chromatin immunoprecipitation assays confirmed Mixl1 occupancy of these promoters in vivo. Furthermore, Mixl1 transactivated the Pdgfrα and Flk1 promoters through ATTA sequences in a DNA binding dependent manner. These data support the hypothesis that Mixl1 directly regulates Pdgfrα and Flk1 gene expression and strengthens the position of Mixl1 as a key regulator of mesendoderm development during mammalian gastrulation.


Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Proteínas de Homeodominio/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular , Endodermo/citología , Endodermo/metabolismo , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/química , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas/genética , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Activación Transcripcional/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
14.
Stem Cell Res ; 8(2): 259-73, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22265745

RESUMEN

The recent introduction of technologies capable of reprogramming human somatic cells into induced pluripotent stem (iPS) cells offers a unique opportunity to study many aspects of neurodegenerative diseases in vitro that could ultimately lead to novel drug development and testing. Here, we report for the first time that human dermal fibroblasts from a patient with relapsing-remitting Multiple Sclerosis (MS) were reprogrammed to pluripotency by retroviral transduction using defined factors (OCT4, SOX2, KLF4, and c-MYC). The MSiPS cell lines resembled human embryonic stem (hES) cell-like colonies in morphology and gene expression and exhibited silencing of the retroviral transgenes after four passages. MSiPS cells formed embryoid bodies that expressed markers of all three germ layers by immunostaining and Reverse Transcriptase (RT)-PCR. The injection of undifferentiated iPS cell colonies into immunodeficient mice formed teratomas, thereby demonstrating pluripotency. The MSiPS cells were successfully differentiated into mature astrocytes, oligodendrocytes and neurons with normal karyotypes. Although MSiPS-derived neurons displayed some differences in their electrophysiological characteristics as compared to the control cell line, they exhibit properties of functional neurons, with robust resting membrane potentials, large fast tetrodotoxin-sensitive action potentials and voltage-gated sodium currents. This study provides for the first time proof of concept that disease cell lines derived from skin cells obtained from an MS patient can be generated and successfully differentiated into mature neural lineages. This represents an important step in a novel approach for the study of MS pathophysiology and potential drug discovery.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas/patología , Esclerosis Múltiple/patología , Esclerosis Múltiple/fisiopatología , Neuronas/patología , Animales , Linaje de la Célula , Fenómenos Electrofisiológicos , Fibroblastos/patología , Humanos , Factor 4 Similar a Kruppel , Ratones , Ratones SCID , Repeticiones de Microsatélite/genética , Factor 3 de Transcripción de Unión a Octámeros/genética , Oligodendroglía/patología , Células Madre Pluripotentes/patología , Regiones Promotoras Genéticas/genética , Retroviridae/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Piel/patología , Transducción Genética
15.
Nat Methods ; 8(12): 1037-40, 2011 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-22020065

RESUMEN

NKX2-5 is expressed in the heart throughout life. We targeted eGFP sequences to the NKX2-5 locus of human embryonic stem cells (hESCs); NKX2-5(eGFP/w) hESCs facilitate quantification of cardiac differentiation, purification of hESC-derived committed cardiac progenitor cells (hESC-CPCs) and cardiomyocytes (hESC-CMs) and the standardization of differentiation protocols. We used NKX2-5 eGFP(+) cells to identify VCAM1 and SIRPA as cell-surface markers expressed in cardiac lineages.


Asunto(s)
Separación Celular/métodos , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/metabolismo , Mioblastos Cardíacos/citología , Miocitos Cardíacos/citología , Factores de Transcripción/metabolismo , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Biomarcadores/análisis , Diferenciación Celular , Perfilación de la Expresión Génica , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/genética , Humanos , Mioblastos Cardíacos/metabolismo , Miocitos Cardíacos/metabolismo , Reacción en Cadena de la Polimerasa , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Factores de Transcripción/genética , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
16.
Int J Dev Biol ; 54(8-9): 1383-8, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20563991

RESUMEN

Slain1 was originally identified as a novel stem cell-associated gene in transcriptional profiling experiments comparing mouse and human embryonic stem cells (ESCs) and their immediate differentiated progeny. In order to obtain further insight into the potential function of Slain1, we examined the expression of beta-galactosidase in a gene-trap mouse line in which a beta-geo reporter gene was inserted into the second intron of Slain1. In early stage embryos (E7.5), the Slain1-betageo fusion protein was expressed within the entire epiblast, but by E9.5 became restricted to the developing nervous system and gastrointestinal tract. In later stage embryos (E11.5 - E13.5), expression was predominantly within the developing nervous system. Lower level expression was also observed in the developing limb buds, in the condensing mesenchyme, along the apical epidermal ridge and, at later stages, within the digital zones. These observations suggest that Slain1 may play a role in the development of the nervous system, as well as in the morphogenesis of several embryonic structures.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Sistema Nervioso/metabolismo , Proteínas/genética , beta-Galactosidasa/genética , Animales , Secuencia de Bases , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Tracto Gastrointestinal/embriología , Tracto Gastrointestinal/metabolismo , Perfilación de la Expresión Génica , Histocitoquímica , Humanos , Esbozos de los Miembros/embriología , Esbozos de los Miembros/metabolismo , Masculino , Ratones , Datos de Secuencia Molecular , Sistema Nervioso/embriología , Proteínas/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Ácido Nucleico , Factores de Tiempo , beta-Galactosidasa/metabolismo
17.
Stem Cells ; 27(2): 363-74, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19038793

RESUMEN

The Mixl1 gene encodes a homeodomain transcription factor that is required for normal mesoderm and endoderm development in the mouse. We have examined the consequences of enforced Mixl1 expression during mouse embryonic stem cell (ESC) differentiation. We show that three independently derived ESC lines constitutively expressing Mixl1 (Mixl1(C) ESCs) differentiate into embryoid bodies (EBs) containing a higher proportion of E-cadherin (E-Cad)(+) cells. Our analysis also shows that this differentiation occurs at the expense of hematopoietic mesoderm differentiation, with Mixl1(C) ESCs expressing only low levels of Flk1 and failing to develop hemoglobinized cells. Immunohistochemistry and immunofluorescence studies revealed that Mixl1(C) EBs have extensive areas containing cells with an epithelial morphology that express E-Cad, FoxA2, and Sox17, consistent with enhanced endoderm formation. Luciferase reporter transfection experiments indicate that Mixl1 can transactivate the Gsc, Sox17, and E-Cad promoters, supporting the hypothesis that Mixl1 has a direct role in definitive endoderm formation. Taken together, these studies suggest that high levels of Mixl1 preferentially allocate cells to the endoderm during ESC differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Endodermo/metabolismo , Proteínas de Homeodominio/fisiología , Mesodermo/citología , Mesodermo/metabolismo , Animales , Células 3T3 BALB , Western Blotting , Cadherinas/genética , Cadherinas/metabolismo , Diferenciación Celular/genética , Línea Celular , Ensayo de Cambio de Movilidad Electroforética , Endodermo/citología , Citometría de Flujo , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 3-beta del Hepatocito/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Ratones , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/metabolismo
18.
Dev Biol ; 293(1): 90-103, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16546155

RESUMEN

We analyzed the transcriptional profiles of differentiating mouse embryonic stem cells (mESCs) and show that embryoid bodies (EBs) sequentially expressed genes associated with the epiblast, primitive streak, mesoderm and endoderm of the developing embryo, validating ESCs as a model system for identifying cohorts of genes marking specific stages of embryogenesis. By comparing the transcriptional profiles of undifferentiated ESCs to those of their differentiated progeny, we identified 503 mESC and 983 hESC genes selectively expressed in undifferentiated ES cells. Over 75% of the mESC genes were expressed in hESC and vice versa, attesting to the underlying similarity of mESCs and hESCs. The expression of a cohort of 68 genes decreased greater than 2-fold during differentiation in both mESCs and hESCs. As well as containing many validated ESC genes such as Oct4 [Pou5f1], Nanog and Nodal, this cohort included an uncharacterised gene (FLJ30046), which we designated SLAIN1/Slain1. Slain1 was expressed at the stem cell and epiblast stages of ESC differentiation and in the epiblast, nervous system, tailbud and somites of the developing mouse embryo. SLAIN1 and its more widely expressed homologue SLAIN2 comprise a new family of structurally unique genes conserved throughout vertebrate evolution.


Asunto(s)
Diferenciación Celular/genética , Perfilación de la Expresión Génica , Proteínas/genética , Células Madre/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Datos de Secuencia Molecular , Proteínas/metabolismo , Células Madre/citología
19.
J Immunol ; 170(2): 805-15, 2003 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-12517944

RESUMEN

Granzyme B (grB) is a serine proteinase released by cytotoxic lymphocytes (CLs) to kill abnormal cells. GrB-mediated apoptotic pathways are conserved in nucleated cells; hence, CLs require mechanisms to protect against ectopic or misdirected grB. The nucleocytoplasmic serpin, proteinase inhibitor 9 (PI-9), is a potent inhibitor of grB that protects cells from grB-mediated apoptosis in model systems. Here we show that PI-9 is present in CD4(+) cells, CD8(+) T cells, NK cells, and at lower levels in B cells and myeloid cells. PI-9 is up-regulated in response to grB production and degranulation, and associates with grB-containing granules in activated CTLs and NK cells. Intracellular complexes of PI-9 and grB are evident in NK cells, and overexpression of PI-9 enhances CTL potency, suggesting that cytoplasmic grB, which may threaten CL viability, is rapidly inactivated by PI-9. Because dendritic cells (DCs) acquire characteristics similar to those of target cells to activate naive CD8(+) T cells and therefore may also require protection against grB, we investigated the expression of PI-9 in DCs. PI-9 is evident in thymic DCs (CD3(-), CD4(+), CD8(-), CD45(+)), tonsillar DCs, and DC subsets purified from peripheral blood (CD16(+) monocytes and CD123(+) plasmacytoid DCs). Furthermore, PI-9 is expressed in monocyte-derived DCs and is up-regulated upon TNF-alpha-induced maturation of monocyte-derived DCs. In conclusion, the presence and subcellular localization of PI-9 in leukocytes and DCs are consistent with a protective role against ectopic or misdirected grB during an immune response.


Asunto(s)
Células Presentadoras de Antígenos/citología , Degranulación de la Célula/inmunología , Citotoxicidad Inmunológica , Serina Endopeptidasas/metabolismo , Inhibidores de Serina Proteinasa/biosíntesis , Serpinas/biosíntesis , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Citotóxicos/inmunología , Regulación hacia Arriba/inmunología , Adyuvantes Inmunológicos/biosíntesis , Adyuvantes Inmunológicos/sangre , Adyuvantes Inmunológicos/genética , Adyuvantes Inmunológicos/fisiología , Células Presentadoras de Antígenos/metabolismo , Diferenciación Celular/inmunología , Línea Celular , Células Cultivadas , Citotoxicidad Inmunológica/genética , Células Dendríticas/clasificación , Células Dendríticas/metabolismo , Granzimas , Humanos , Líquido Intracelular/enzimología , Líquido Intracelular/metabolismo , Leucocitos/metabolismo , Serina Endopeptidasas/biosíntesis , Inhibidores de Serina Proteinasa/sangre , Inhibidores de Serina Proteinasa/genética , Inhibidores de Serina Proteinasa/fisiología , Serpinas/sangre , Serpinas/genética , Serpinas/fisiología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Citotóxicos/enzimología , Regulación hacia Arriba/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA