Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Redox Biol ; 56: 102431, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35988446

RESUMEN

YAP1 and TAZ are transcriptional co-activator proteins that play fundamental roles in many biological processes, from cell proliferation and cell lineage fate determination to tumorigenesis. We previously demonstrated that Limb Expression 1 (LIX1) regulates YAP1 and TAZ activity and controls digestive mesenchymal progenitor proliferation. However, LIX1 mode of action remains elusive. Here, we found that endogenous LIX1 is localized in mitochondria and is anchored to the outer mitochondrial membrane through S-palmitoylation of cysteine 84, a residue conserved in all LIX1 orthologs. LIX1 downregulation altered the mitochondrial ultrastructure, resulting in a significantly decreased respiration and attenuated production of mitochondrial reactive oxygen species (mtROS). Mechanistically, LIX1 knock-down impaired the stability of the mitochondrial proteins PHB2 and OPA1 that are found in complexes with mitochondrial-specific phospholipids and are required for cristae organization. Supplementation with unsaturated fatty acids counteracted the effects of LIX1 knock-down on mitochondrial morphology and ultrastructure and restored YAP1/TAZ signaling. Collectively, our data demonstrate that LIX1 is a key regulator of cristae organization, modulating mtROS level and subsequently regulating the signaling cascades that control fate commitment of digestive mesenchyme-derived cells.


Asunto(s)
Cisteína , Mitocondrias , Cisteína/metabolismo , Mesodermo/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Fosfolípidos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
2.
Stem Cells Dev ; 30(2): 79-90, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33256572

RESUMEN

Forced coexpression of the transcription factors Oct3/4, Klf4, Sox2, and c-Myc reprograms somatic cells into pluripotent stem cells (PSCs). Such induced PSCs (iPSCs) can generate any cell type of the adult body or indefinitely proliferate without losing their potential. Accordingly, iPSCs can serve as an unlimited cell source for the development of various disease models and regenerative therapies for animals and humans. Although canine peripheral blood mononuclear cells (PBMCs) can be easily obtained, they have a very low iPSC reprogramming efficiency. In this study, we determined the reprogramming efficiency of canine PBMCs under several conditions involving three types of media supplemented with small-molecule compounds. We found that canine iPSCs (ciPSCs) could be efficiently generated from PBMCs using N2B27 medium supplemented with leukemia inhibitory factor (LIF), basic fibroblast growth factor (bFGF), and a small-molecule cocktail (Y-27632, PD0325901, CHIR99021, A-83-01, Forskolin, and l-ascorbic acid). We generated five ciPSC lines that could be maintained in StemFit® medium supplemented with LIF. The SeVdp(KOSM)302L vectors were appropriately silenced in four ciPSC lines. Of the two lines characterized, both were positive for alkaline phosphatase activity and expressed pluripotency markers, including the Oct3/4, Sox2, and Nanog transcripts, as well as the octamer-binding transcription factor (OCT) 3/4 and NANOG proteins, and the SSEA-1 carbohydrate antigen. The ciPSCs could form embryoid bodies and differentiate into the three germ layers, as indicated by marker gene and protein expression. Furthermore, one ciPSC line formed teratomas comprising several tissues from every germ layer. Our ciPSC lines maintained a normal karyotype even after multiple passages. Moreover, our new reprogramming method was able to generate ciPSCs from multiple donor PBMCs. In conclusion, we developed an easy and efficient strategy for the generation of footprint-free ciPSCs from PBMCs. We believe that this strategy can be useful for disease modeling and regenerative medicine in the veterinary field.


Asunto(s)
Diferenciación Celular/genética , Reprogramación Celular/genética , Expresión Génica/genética , Células Madre Pluripotentes Inducidas/metabolismo , Leucocitos Mononucleares/metabolismo , Animales , Células Cultivadas , Técnicas de Reprogramación Celular/métodos , Medios de Cultivo/química , Medios de Cultivo/farmacología , Perros , Ectodermo/citología , Ectodermo/metabolismo , Endodermo/citología , Endodermo/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/citología , Leucocitos Mononucleares/citología , Mesodermo/citología , Mesodermo/metabolismo , Ratones Endogámicos ICR , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Reproducibilidad de los Resultados , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
3.
Proc Natl Acad Sci U S A ; 116(47): 23636-23642, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31685615

RESUMEN

Sonic hedgehog (SHH) signaling plays a pivotal role in 2 different phases during brain development. Early SHH signaling derived from the prechordal plate (PrCP) triggers secondary Shh induction in the forebrain, which overlies the PrCP, and the induced SHH signaling, in turn, directs late neuronal differentiation of the forebrain. Consequently, Shh regulation in the PrCP is crucial for initiation of forebrain development. However, no enhancer that regulates prechordal Shh expression has yet been found. Here, we identified a prechordal enhancer, named SBE7, in the vicinity of a cluster of known forebrain enhancers for Shh This enhancer also directs Shh expression in the ventral midline of the forebrain, which receives the prechordal SHH signal. Thus, the identified enhancer acts not only for the initiation of Shh regulation in the PrCP but also for subsequent Shh induction in the forebrain. Indeed, removal of the enhancer from the mouse genome markedly down-regulated the expression of Shh in the rostral domains of the axial mesoderm and in the ventral midline of the forebrain and hypothalamus in the mouse embryo, and caused a craniofacial abnormality similar to human holoprosencephaly (HPE). These findings demonstrate that SHH signaling mediated by the newly identified enhancer is essential for development and growth of the ventral midline of the forebrain and hypothalamus. Understanding of the Shh regulation governed by this prechordal and brain enhancer provides an insight into the mechanism underlying craniofacial morphogenesis and the etiology of HPE.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/fisiología , Proteínas del Tejido Nervioso/fisiología , Prosencéfalo/embriología , Animales , Sistemas CRISPR-Cas , Proteínas del Ojo/fisiología , Técnicas de Inactivación de Genes , Genes Reporteros , Proteínas Hedgehog/biosíntesis , Proteínas Hedgehog/genética , Holoprosencefalia/genética , Proteínas de Homeodominio/fisiología , Hipotálamo/anomalías , Hipotálamo/embriología , Hipotálamo/metabolismo , Operón Lac , Mesencéfalo/embriología , Mesencéfalo/metabolismo , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Prosencéfalo/anomalías , Prosencéfalo/metabolismo , Transducción de Señal , Transgenes , Proteína Homeobox SIX3
4.
Birth Defects Res ; 111(19): 1520-1534, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31518072

RESUMEN

BACKGROUND: Periconceptional intake of supplemental folic acid can reduce the incidence of neural tube defects by as much as 70%, but the mechanisms by which folic acid supports cellular processes during neural tube closure are unknown. The mitochondrial 10-formyl-tetrahydrofolate synthetase MTHFD1L catalyzes production of formate, thus generating one-carbon units for cytoplasmic processes. Deletion of Mthfd1l causes embryonic lethality, developmental delay, and neural tube defects in mice. METHODS: To investigate the role of mitochondrial one-carbon metabolism during cranial neural tube closure, we have analyzed cellular morphology and function in neural tissues in Mthfd1l knockout embryos. RESULTS: The head mesenchyme showed significantly lower cellular density in Mthfd1l nullizygous embryos compared to wildtype embryos during the process of neural tube closure. Apoptosis and neural crest cell specification were not affected by deletion of Mthfd1l. Sections from the cranial region of Mthfd1l knockout embryos exhibited decreased cellular proliferation, but only after completion of neural tube closure. Supplementation of pregnant dams with formate improved mesenchymal density and corrected cell proliferation in the nullizygous embryos. CONCLUSIONS: Deletion of Mthfd1l causes decreased density in the cranial mesenchyme and this defect is improved with formate supplementation. This study reveals a mechanistic link between folate-dependent mitochondrially produced formate, head mesenchyme formation and neural tube defects.


Asunto(s)
Formiato-Tetrahidrofolato Ligasa/genética , Meteniltetrahidrofolato Ciclohidrolasa/genética , Metilenotetrahidrofolato Deshidrogenasa (NADP)/genética , Enzimas Multifuncionales/genética , Defectos del Tubo Neural/genética , Animales , Embrión de Mamíferos/metabolismo , Femenino , Ácido Fólico/genética , Ácido Fólico/metabolismo , Formiato-Tetrahidrofolato Ligasa/metabolismo , Formiatos/metabolismo , Masculino , Mesodermo/metabolismo , Meteniltetrahidrofolato Ciclohidrolasa/metabolismo , Metilenotetrahidrofolato Deshidrogenasa (NADP)/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Enzimas Multifuncionales/metabolismo , Cresta Neural/metabolismo , Defectos del Tubo Neural/metabolismo , Neurulación , Eliminación de Secuencia
5.
Cell Stem Cell ; 24(2): 318-327.e8, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30554961

RESUMEN

Human protein-coding genes are often accompanied by divergently transcribed non-coding RNAs whose functions, especially in cell fate decisions, are poorly understood. Using an hESC-based cardiac differentiation model, we define a class of divergent lncRNAs, termed yin yang lncRNAs (yylncRNAs), that mirror the cell-type-specific expression pattern of their protein-coding counterparts. yylncRNAs are preferentially encoded from the genomic loci of key developmental cell fate regulators. Most yylncRNAs are spliced polyadenylated transcripts showing comparable expression patterns in vivo in mouse and in human embryos. Signifying their developmental function, the key mesoderm specifier BRACHYURY (T) is accompanied by yylncT, which localizes to the active T locus during mesoderm commitment. yylncT binds the de novo DNA methyltransferase DNMT3B, and its transcript is required for activation of the T locus, with yylncT depletion specifically abolishing mesodermal commitment. Collectively, we report a lncRNA-mediated regulatory layer safeguarding embryonic cell fate transitions.


Asunto(s)
Linaje de la Célula/genética , Proteínas Fetales/metabolismo , Mesodermo/metabolismo , Células Madre Pluripotentes/metabolismo , ARN Largo no Codificante/genética , Proteínas de Dominio T Box/metabolismo , Transcripción Genética , Animales , Diferenciación Celular , Línea Celular , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Sitios Genéticos , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Ratones , ARN Largo no Codificante/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ADN Metiltransferasa 3B
6.
Sci Rep ; 6: 21629, 2016 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-26899926

RESUMEN

The mesenchymal state in cancer is usually associated with poor prognosis due to the metastatic predisposition and the hyper-activated metabolism. Exploiting cell glucose metabolism we propose a new method to detect mesenchymal-like cancer cells. We demonstrate that the uptake of glucose-coated magnetic nanoparticles (MNPs) by mesenchymal-like cells remains constant when the glucose in the medium is increased from low (5.5 mM) to high (25 mM) concentration, while the MNPs uptake by epithelial-like cells is significantly reduced. These findings reveal that the glucose-shell of MNPs plays a major role in recognition of cells with high-metabolic activity. By selectively blocking the glucose transporter 1 channels we showed its involvement in the internalization process of glucose-coated MNPs. Our results suggest that glucose-coated MNPs can be used for metabolic-based assays aimed at detecting cancer cells and that can be used to selectively target cancer cells taking advantage, for instance, of the magnetic-thermotherapy.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Transportador de Glucosa de Tipo 1/genética , Glucosa/administración & dosificación , Nanopartículas de Magnetita/administración & dosificación , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/patología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Femenino , Glucosa/química , Glucosa/metabolismo , Transportador de Glucosa de Tipo 1/antagonistas & inhibidores , Humanos , Hipertermia Inducida , Células MCF-7 , Nanopartículas de Magnetita/química , Mesodermo/metabolismo , Mesodermo/patología
7.
PLoS One ; 9(4): e93989, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24705708

RESUMEN

The Hox gene family encodes homeodomain-containing transcriptional regulators that confer positional information to axial and paraxial tissues in the developing embryo. The dynamic Hox gene expression pattern requires mechanisms that differentially control Hox transcription in a precise spatio-temporal fashion. This implies an integrated regulation of neighbouring Hox genes achieved through the sharing and the selective use of defined enhancer sequences. The Hoxa5 gene plays a crucial role in lung and gut organogenesis. To position Hoxa5 in the regulatory hierarchy that drives organ morphogenesis, we searched for cis-acting regulatory sequences and associated trans-acting factors required for Hoxa5 expression in the developing lung and gut. Using mouse transgenesis, we identified two DNA regions included in a 1.5-kb XbaI-XbaI fragment located in the Hoxa4-Hoxa5 intergenic domain and known to control Hoxa4 organ expression. The multifunctional YY1 transcription factor binds the two regulatory sequences in vitro and in vivo. Moreover, the mesenchymal deletion of the Yy1 gene function in mice results in a Hoxa5-like lung phenotype with decreased Hoxa5 and Hoxa4 gene expression. Thus, YY1 acts as a positive regulator of Hoxa5 expression in the developing lung and gut. Our data also support a role for YY1 in the coordinated expression of Hox genes for correct organogenesis.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Organogénesis/genética , Fosfoproteínas/genética , Factor de Transcripción YY1/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Orden Génico , Marcación de Gen , Sitios Genéticos , Proteínas de Homeodominio/química , Proteínas de Homeodominio/metabolismo , Pulmón/embriología , Pulmón/metabolismo , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Especificidad de Órganos/genética , Fenotipo , Fosfoproteínas/química , Regiones Promotoras Genéticas , Unión Proteica , Secuencias Reguladoras de Ácidos Nucleicos , Elementos de Respuesta , Factores de Transcripción
8.
Am J Physiol Lung Cell Mol Physiol ; 305(4): L301-11, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23812632

RESUMEN

Curcumin, a potent anti-inflammatory and antioxidant agent, modulates peroxisome proliferator-activated receptor-γ signaling, a key molecule in the etiology of bronchopulmonary dysplasia (BPD). We have previously shown curcumin's acute protection against neonatal hyperoxia-induced lung injury. However, its longer-term protection against BPD is not known. Hypothesizing that concurrent treatment with curcumin protects the developing lung against hyperoxia-induced lung injury long-term, we determined if curcumin protects against hyperoxic neonatal rat lung injury for the first 5 days of life, as determined at postnatal day (PND) 21. One-day-old rat pups were exposed to either 21 or 95% O2 for 5 days with or without curcumin treatment (5 mg/kg) administered intraperitoneally one time daily, following which the pups grew up to PND21 in room air. At PND21 lung development was determined, including gross and cellular structural and functional effects, and molecular mediators of inflammatory injury. To gain mechanistic insights, embryonic day 19 fetal rat lung fibroblasts were examined for markers of apoptosis and MAP kinase activation following in vitro exposure to hyperoxia for 24 h in the presence or absence of curcumin (5 µM). Curcumin effectively blocked hyperoxia-induced lung injury based on systematic analysis of markers for lung injury (apoptosis, Bcl-2/Bax, collagen III, fibronectin, vimentin, calponin, and elastin-related genes) and lung morphology (radial alveolar count and alveolar septal thickness). Mechanistically, curcumin prevented the hyperoxia-induced increases in cleaved caspase-3 and the phosphorylation of Erk1/2. Molecular effects of curcumin, both structural and cytoprotective, suggest that its actions against hyperoxia-induced lung injury are mediated via Erk1/2 activation and that it is a potential intervention against BPD.


Asunto(s)
Curcumina/uso terapéutico , Hiperoxia/tratamiento farmacológico , Hiperoxia/prevención & control , Pulmón/embriología , Pulmón/patología , Sustancias Protectoras/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Proliferación Celular/efectos de los fármacos , Curcumina/farmacología , Elastina/genética , Elastina/metabolismo , Activación Enzimática/efectos de los fármacos , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Fibronectinas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Hiperoxia/embriología , Hiperoxia/genética , Pulmón/efectos de los fármacos , Pulmón/enzimología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , Mesodermo/patología , Sustancias Protectoras/farmacología , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta/metabolismo , Triglicéridos/metabolismo
9.
PLoS One ; 8(3): e58828, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23554936

RESUMEN

Mouse embryos lacking the polycomb group gene member Yin-Yang1 (YY1) die during the peri-implantation stage. To assess the post-gastrulation role of YY1, a conditional knock-out (cKO) strategy was used to delete YY1 from the visceral endoderm of the yolk sac and the definitive endoderm of the embryo. cKO embryos display profound yolk sac defects at 9.5 days post coitum (dpc), including disrupted angiogenesis in mesoderm derivatives and altered epithelial characteristics in the visceral endoderm. Significant changes in both cell death and proliferation were confined to the YY1-expressing yolk sac mesoderm indicating that loss of YY1 in the visceral endoderm causes defects in the adjacent yolk sac mesoderm. Production of Vascular Endothelial Growth Factor A (VEGFA) by the visceral endoderm is essential for normal growth and development of the yolk sac vasculature. Reduced levels of VEGFA are observed in the cKO yolk sac, suggesting a cause for the angiogenesis defects. Ex vivo culture with exogenous VEGF not only rescued angiogenesis and apoptosis in the cKO yolk sac mesoderm, but also restored the epithelial defects observed in the cKO visceral endoderm. Intriguingly, blocking the activity of the mesoderm-localized VEGF receptor, FLK1, recapitulates both the mesoderm and visceral endoderm defects observed in the cKO yolk sac. Taken together, these results demonstrate that YY1 is responsible for maintaining VEGF in the developing visceral endoderm and that a VEGF-responsive paracrine signal, originating in the yolk sac mesoderm, is required to promote normal visceral endoderm development.


Asunto(s)
Endodermo/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor de Transcripción YY1/metabolismo , Saco Vitelino/metabolismo , Animales , Muerte Celular/genética , Proliferación Celular , Endodermo/embriología , Femenino , Expresión Génica , Técnicas de Inactivación de Genes , Masculino , Mesodermo/irrigación sanguínea , Mesodermo/embriología , Mesodermo/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/farmacología , Factor de Transcripción YY1/genética , Saco Vitelino/irrigación sanguínea , Saco Vitelino/efectos de los fármacos , Saco Vitelino/embriología
10.
Dev Cell ; 24(6): 600-11, 2013 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-23537631

RESUMEN

Hematopoietic stem cells (HSCs) are produced by a small cohort of hemogenic endothelial cells (ECs) during development through the formation of intra-aortic hematopoietic cell (HC) clusters. The Runx1 transcription factor plays a key role in the EC-to-HC and -HSC transition. We show that Runx1 expression in hemogenic ECs and the subsequent initiation of HC formation are tightly controlled by the subaortic mesenchyme, although the mesenchyme is not a source of HCs. Runx1 and Notch signaling are involved in this process, with Notch signaling decreasing with time in HCs. Inhibiting Notch signaling readily increases HC production in mouse and chicken embryos. In the mouse, however, this increase is transient. Collectively, we show complementary roles of hemogenic ECs and mesenchymal compartments in triggering aortic hematopoiesis. The subaortic mesenchyme induces Runx1 expression in hemogenic-primed ECs and collaborates with Notch dynamics to control aortic hematopoiesis.


Asunto(s)
Aorta/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Células Endoteliales/metabolismo , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Animales , Aorta/crecimiento & desarrollo , Proteínas de Unión al Calcio , Diferenciación Celular/genética , Movimiento Celular , Células Cultivadas , Pollos , Subunidad alfa 2 del Factor de Unión al Sitio Principal/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Hemangioblastos , Péptidos y Proteínas de Señalización Intercelular , Proteína Jagged-2 , Proteínas de la Membrana , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Codorniz , Receptores Notch/metabolismo , Proteínas Serrate-Jagged , Transducción de Señal/genética
11.
Dev Growth Differ ; 53(6): 780-92, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21711459

RESUMEN

Bone morphogenetic proteins (BMPs) play a crucial role in programmed cell death (PCD), a biological process required for the sculpturing of the embryonic limbs. However, it is unknown if BMP signaling directly promotes cell death, or if it induces a molecular cascade that culminates in cell death. Given that Smad8, which encodes one component of BMP signaling, is expressed during the regression of interdigital tissue and responds to BMPs, we presumed that it may be expressed in other cell death areas during chick limb development such as the anterior and posterior necrotic zones (ANZ and PNZ). The present study found that the Smad8 expression pattern in the anterior mesoderm of the hindlimb is very similar to that observed in limbs stained to detect cell death. Also, BMPs and retinoic acid, which act as apoptosis-promoting factors, induced expression of Smad8 before the onset of cell death, while sonic hedgehog protein, acting as a survival factor, inhibited Smad8 expression in the ANZ. However, although there was correlation between Smad8 expression patterns and PCD in the ANZ, phosphorylated forms of SMAD1/5/8 and TUNEL staining did not co-localize in dying cells. Interestingly, a short pulse of BMP was sufficient to trigger cell death. On the other hand, most dying cells were located in the avascular region, while many cells expressing Smad8 were located in the vascular region of the ANZ. These results suggest that BMPs mediated by SMAD signaling activate a molecular cascade that culminates in PCD.


Asunto(s)
Proteína Morfogenética Ósea 7/farmacología , Muerte Celular , Embrión de Pollo/efectos de los fármacos , Miembro Posterior/embriología , Proteína Smad8/metabolismo , Animales , Embrión de Pollo/citología , Embrión de Pollo/embriología , Clonación Molecular , ADN Complementario/genética , ADN Complementario/metabolismo , Desarrollo Embrionario , Técnica del Anticuerpo Fluorescente , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Proteínas Hedgehog/farmacología , Miembro Posterior/citología , Miembro Posterior/efectos de los fármacos , Humanos , Mesodermo/citología , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , Microscopía Confocal , Fosforilación , Proteínas Recombinantes/farmacología , Transducción de Señal , Proteína Smad8/genética , Factores de Tiempo , Tretinoina/administración & dosificación , Tretinoina/farmacología
12.
Development ; 138(10): 1913-23, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21471156

RESUMEN

The essential roles of SHH in anteroposterior (AP) and AER-FGF signalling in proximodistal (PD) limb bud development are well understood. In addition, these morphoregulatory signals are key components of the self-regulatory SHH/GREM1/AER-FGF feedback signalling system that regulates distal progression of limb bud development. This study uncovers an additional signalling module required for coordinated progression of limb bud axis development. Transcriptome analysis using Shh-deficient mouse limb buds revealed that the expression of proximal genes was distally extended from early stages onwards, which pointed to a more prominent involvement of SHH in PD limb axis development. In particular, retinoic acid (RA) target genes were upregulated proximally, while the expression of the RA-inactivating Cyp26b1 enzyme was downregulated distally, pointing to increased RA activity in Shh-deficient mouse limb buds. Further genetic and molecular analysis established that Cyp26b1 expression is regulated by AER-FGF signalling. During initiation of limb bud outgrowth, the activation of Cyp26b1 expression creates a distal 'RA-free' domain, as indicated by complementary downregulation of a transcriptional sensor of RA activity. Subsequently, Cyp26b1 expression increases as a consequence of SHH-dependent upregulation of AER-FGF signalling. To better understand the underlying signalling interactions, computational simulations of the spatiotemporal expression patterns and interactions were generated. These simulations predicted the existence of an antagonistic AER-FGF/CYP26B1/RA signalling module, which was verified experimentally. In summary, SHH promotes distal progression of limb development by enhancing CYP26B1-mediated RA clearance as part of a signalling network linking the SHH/GREM1/AER-FGF feedback loop to the newly identified AER-FGF/CYP26B1/RA module.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas Hedgehog/metabolismo , Esbozos de los Miembros/embriología , Esbozos de los Miembros/metabolismo , Tretinoina/metabolismo , Animales , Sistema Enzimático del Citocromo P-450/genética , Ectodermo/embriología , Ectodermo/metabolismo , Activación Enzimática , Retroalimentación Fisiológica , Femenino , Factores de Crecimiento de Fibroblastos/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/deficiencia , Proteínas Hedgehog/genética , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Ratones Noqueados , Ratones Mutantes , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Embarazo , Ácido Retinoico 4-Hidroxilasa , Transducción de Señal
13.
Proc Natl Acad Sci U S A ; 107(29): 12907-12, 2010 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-20615943

RESUMEN

Vertebrate embryo somite formation is temporally controlled by the cyclic expression of somitogenesis clock genes in the presomitic mesoderm (PSM). The somitogenesis clock is believed to be an intrinsic property of this tissue, operating independently of embryonic midline structures and the signaling molecules produced therein, namely Sonic hedgehog (Shh). This work revisits the notochord signaling contribution to temporal control of PSM segmentation by assessing the rate and number of somites formed and somitogenesis molecular clock gene expression oscillations upon notochord ablation. The absence of the notochord causes a delay in somite formation, accompanied by an increase in the period of molecular clock oscillations. Shh is the notochord-derived signal responsible for this effect, as these alterations are recapitulated by Shh signaling inhibitors and rescued by an external Shh supply. We have characterized chick smoothened expression pattern and have found that the PSM expresses both patched1 and smoothened Shh signal transducers. Upon notochord ablation, patched1, gli1, and fgf8 are down-regulated, whereas gli2 and gli3 are overexpressed. Strikingly, notochord-deprived PSM segmentation rate recovers over time, concomitant with raldh2 overexpression. Accordingly, exogenous RA supplement rescues notochord ablation effects on somite formation. A model is presented in which Shh and RA pathways converge to inhibit PSM Gli activity, ensuring timely somite formation. Altogether, our data provide evidence that a balance between different pathways ensures the robustness of timely somite formation and that notochord-derived Shh is a component of the molecular network regulating the pace of the somitogenesis clock.


Asunto(s)
Tipificación del Cuerpo , Proteínas Hedgehog/metabolismo , Somitos/metabolismo , Animales , Relojes Biológicos/efectos de los fármacos , Relojes Biológicos/genética , Tipificación del Cuerpo/efectos de los fármacos , Embrión de Pollo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Mesodermo/citología , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , Notocorda/citología , Notocorda/efectos de los fármacos , Notocorda/metabolismo , Transducción de Señal/efectos de los fármacos , Somitos/citología , Somitos/efectos de los fármacos , Factores de Tiempo , Tretinoina/farmacología
14.
Dev Biol ; 344(2): 869-80, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20534379

RESUMEN

Neural tube closure defects are among the most frequent congenital malformations in humans. Supplemental maternal intake of folic acid before and during pregnancy reduces their incidence significantly, but the mechanism underlying this preventive effect is unknown. As a number of genes that cause neural tube closure defects encode transcriptional regulators in mice, one possibility is that folic acid could induce the expression of transcription factors to compensate for the primary genetic defect. We report that folic acid is required in mouse embryos for the specific expression of the homeodomain gene Alx3 in the head mesenchyme, an important tissue for cranial neural tube closure. Alx3-deficient mice exhibit increased failure of cranial neural tube closure and increased cell death in the craniofacial region, two effects that are also observed in wild type embryos developing in the absence of folic acid. Folic acid cannot prevent these defects in Alx3-deficient embryos, indicating that one mechanism of folic acid action is through induced expression of Alx3. Thus, Alx3 emerges as a candidate gene for human neural tube defects and reveals the existence of induced transcription factor gene expression as a previously unknown mechanism by which folic acid prevents neural tube closure defects.


Asunto(s)
Ácido Fólico/metabolismo , Defectos del Tubo Neural/genética , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos , Femenino , Ácido Fólico/genética , Ácido Fólico/farmacología , Genotipo , Hematínicos , Mesodermo/metabolismo , Ratones , Defectos del Tubo Neural/prevención & control , Neurulación , Embarazo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
Blood ; 113(19): 4525-33, 2009 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-19196659

RESUMEN

The peculiar site of development of primary effusion lymphoma (PEL) highlights a specific role of body cavities in the pathogenesis of this neoplasia. We used a xenograft murine model of PEL to characterize the contribution of the host microenvironment to PEL growth. The activity of a murine (ie, host-specific) interferon-alpha(1) (IFN-alpha(1))-expressing lentiviral vector (mIFN-alpha(1)-LV) was compared with that of a human (h) IFN-alpha(2)b-LV. LVs efficiently delivered the transgene to PEL cells and conferred long-term transgene expression in vitro and in vivo. Treatment of PEL-injected severe combined immunodeficiency mice with hIFN-alpha(2)b-LV significantly prolonged mice survival and reduced ascites development. Interestingly, mIFN-alpha(1)-LV showed an antineoplastic activity comparable with that observed with hIFN-alpha(2)b-LV. As mIFN-alpha(1) retained species-restricted activity in vitro, it probably acted in vivo on the intracavitary murine milieu. mIFN-alpha(1)-treated murine mesothelial cells were found to express tumor necrosis factor-related apoptosis-inducing ligand and to significantly trigger apoptosis of cocultured PEL cells in a tumor necrosis factor-related apoptosis-inducing ligand-dependent manner. These data suggest that the interaction between lymphomatous and mesothelial cells lining the body cavities may play a key role in PEL growth control and also indicate that the specific targeting of microenvironment may impair PEL development.


Asunto(s)
Antineoplásicos/uso terapéutico , Vectores Genéticos , Interferón-alfa/uso terapéutico , Lentivirus/genética , Linfoma de Efusión Primaria/tratamiento farmacológico , Animales , Células Cultivadas , Citocinas/metabolismo , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Interferón alfa-2 , Riñón/citología , Riñón/efectos de los fármacos , Riñón/metabolismo , Linfoma de Efusión Primaria/genética , Linfoma de Efusión Primaria/patología , Mesodermo/citología , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , Ratones , Ratones SCID , Peritoneo/citología , Peritoneo/efectos de los fármacos , Peritoneo/metabolismo , Proteínas Recombinantes
16.
Biol Reprod ; 80(4): 781-7, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19109223

RESUMEN

Untimely rupture of the fetal membranes (FMs) is a major precipitant of preterm birth. Although the mechanism of FM weakening leading to rupture is not completely understood, proinflammatory cytokines, including tumor necrosis factor (TNF) and interleukin 1 beta (IL1B), have been shown to weaken FMs concomitant with the induction of reactive oxygen species, collagen remodeling, and prostaglandin release. We hypothesized that alpha-lipoic acid, a dietary antioxidant, may block the effect of inflammatory mediators and thereby inhibit FM weakening. Full-thickness FM fragments were incubated with control media or TNF, with or without alpha-lipoic acid pretreatment. Fetal membrane rupture strength and the release of matrix metalloproteinase 9 (MMP9) and prostaglandin E(2) (PGE(2)) from the full-thickness FM fragments were determined. The two constituent cell populations in amnion, the mechanically strongest FM component, were similarly examined. Amnion epithelial and mesenchymal cells were treated with TNF or IL1B, with or without alpha-lipoic acid pretreatment. MMP9 and PGE(2) were analyzed by ELISA, Western blot, and zymography. TNF decreased FM rupture strength 50% while increasing MMP9 and PGE(2) release. Lipoic acid inhibited these TNF-induced effects. Lipoic acid pretreatment also inhibited TNF- and IL1B-induced increases in MMP9 protein activity and release in amnion epithelial cells, as well as PGE(2) increases in both amnion epithelial and mesenchymal cells. In summary, lipoic acid pretreatment inhibited TNF-induced weakening of FM and cytokine-induced MMP9 and PGE(2) in both intact FM and amnion cells. We speculate that dietary supplementation with alpha-lipoic acid might prove clinically useful in prevention of preterm premature rupture of fetal membranes.


Asunto(s)
Membranas Extraembrionarias/efectos de los fármacos , Resistencia al Corte/efectos de los fármacos , Ácido Tióctico/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Amnios/citología , Amnios/efectos de los fármacos , Amnios/metabolismo , Células Cultivadas , Dinoprostona/metabolismo , Evaluación Preclínica de Medicamentos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Membranas Extraembrionarias/metabolismo , Membranas Extraembrionarias/fisiología , Femenino , Rotura Prematura de Membranas Fetales/prevención & control , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Mesodermo/metabolismo , Mesodermo/fisiología , Embarazo , Ácido Tióctico/uso terapéutico , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
17.
Gene Expr Patterns ; 8(7-8): 481-5, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18647663

RESUMEN

The adenohypophysis is derived from a structure called the Rathke's pouch, which is an invagination of the hypophyseal placode. Hedgehog (Hh) and retinoic acid (RA) signals as well as several transcription factors have been suggested to play a role in the development of the adenohypophysis. We have therefore examined the expression pattern of Sonic hedgehog (Shh), the hedgehog receptor Patched2 (Ptc2), the retinoic acid producing enzyme Retinaldehyde dehydrogenase3 (Raldh3) and four transcription factors, Pitx2, Isl1, Lim3 and Pax6 in chick embryos from head fold stage to embryonic day (E) 4.5. We show that already at the head fold stage, Ptc2 is expressed in prospective hypophyseal placodal cells and that Shh is expressed in the underlying mesoderm. Moreover, Shh continues to be expressed in tissues surrounding the prospective adenohypophysis, and Ptc2 is expressed in prospective hypophyseal cells. Lim3 and Pax6 are expressed from stage 10 in the prospective hypophyseal placode, whereas Pitx2 starts to be expressed before stage 10. Pitx2 is together with Pax6 expressed in the entire domain of the Rathke's pouch. Raldh3 is detected at the 20 somite stage and is together with Lim3 expressed in the anterior part of the Rathke's pouch. Isl1 is expressed in the most posterior part of the hypophyseal ectoderm in a complementary pattern to Raldh3 and Lim3.


Asunto(s)
Embrión de Pollo , Proteínas Hedgehog/metabolismo , Adenohipófisis/embriología , Adenohipófisis/metabolismo , Factores de Transcripción/metabolismo , Aldehído Oxidorreductasas/metabolismo , Animales , Proteínas del Ojo/metabolismo , Expresión Génica , Proteínas de Homeodominio/metabolismo , Mesodermo/metabolismo , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/metabolismo , Receptores Patched , Receptores de Superficie Celular/metabolismo , Proteínas Represoras/metabolismo
18.
Stem Cells ; 26(8): 2104-13, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18511600

RESUMEN

Liver injury activates quiescent hepatic stellate cells (Q-HSC) to proliferative myofibroblasts. Accumulation of myofibroblastic hepatic stellate cells (MF-HSC) sometimes causes cirrhosis and liver failure. However, MF-HSC also promote liver regeneration by producing growth factors for oval cells, bipotent progenitors of hepatocytes and cholangiocytes. Genes that are expressed by primary hepatic stellate cell (HSC) isolates overlap those expressed by oval cells, and hepatocytic and ductular cells emerge when HSC are cultured under certain conditions. We evaluated the hypothesis that HSC are a type of oval cell and, thus, capable of generating hepatocytes to regenerate injured livers. Because Q-HSC express glial fibrillary acidic protein (GFAP), we crossed mice in which GFAP promoter elements regulated Cre-recombinase with ROSA-loxP-stop-loxP-green fluorescent protein (GFP) mice to generate GFAP-Cre/GFP double-transgenic mice. These mice were fed methionine choline-deficient, ethionine-supplemented diets to activate and expand HSC and oval cell populations. GFP(+) progeny of GFAP-expressing precursors were characterized by immunohistochemistry. Basal expression of mesenchymal markers was negligible in GFAP(+)Q-HSC. When activated by liver injury or culture, HSC downregulated expression of GFAP but remained GFP(+); they became highly proliferative and began to coexpress markers of mesenchyme and oval cells. These transitional cells disappeared as GFP-expressing hepatocytes emerged, began to express albumin, and eventually repopulated large areas of the hepatic parenchyma. Ductular cells also expressed GFAP and GFP, but their proliferative activity did not increase in this model. These findings suggest that HSC are a type of oval cell that transitions through a mesenchymal phase before differentiating into hepatocytes during liver regeneration. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Células Epiteliales/citología , Hepatocitos/citología , Regeneración Hepática/fisiología , Células Madre/citología , Animales , Linaje de la Célula , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Inmunohistoquímica/métodos , Cirrosis Hepática/patología , Mesodermo/metabolismo , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas
19.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 27(6): 535-9, 2007 Jun.
Artículo en Chino | MEDLINE | ID: mdl-17633368

RESUMEN

OBJECTIVE: To study the effect of Kurarinone on renal tubular epithelial cell-mesenchyma (ECM) trans-differentiation in rats with renal interstitial fibrosis and to explore its possible mechanisms. METHODS: The rat model of renal interstitial fibrosis was established by unilateral ureteral obstruction (UUO). Sprague-Dawley male rats were randomly divided into 3 groups, the sham-operated group, the UUO group and the Kurarinone treated group (KTG). Rats in the KTG were intraperitoneally injected with Kurarinone 100 mg/kg daily after modeling. Five rats of each group were killed respectively at day 7, 14 and 21 after UUO. The serum levels of blood urea nitrogen (BUN), serum creatinine (SCr), total protein (TP) and albumin (ALB), 24-h urinary protein excretion in rats were measured. Pathological changes of renal tissue were observed by PAS and Masson stain. The expression of transforming growth factor beta1 (TGF-beta1), Smad3, alpha-smooth muscle actin (alpha-SMA) and collagen I (Col I) in kidney were determined with immunohistochemistry. And the expressions of TGF-beta1 and alpha-SMA mRNA in renal tissue were determined using reverse transcription polymerase chain reaction (RT-PCR). RESULTS: The expression of TGF-beta1, Smad3, alpha-SMA and Col I in the KTG was significantly decreased as compared with that in the UUO group respectively, and the degree of tubular damage and renal interstitial fibrosis was also ameliorated more obviously in the KTG. The TGF-beta1 and alpha-SMA mRNA expressions in KTG were significantly lower than those in the UUO group determined at the corresponding time points (P < 0.05). CONCLUSION: Kurarinone could down-regulate the expression of TGF-beta1 and Col I, inhibit EC-M trans-differentiation, suppress the activation and proliferation of myofibroblast. The probable pathway may be by way of down-regulating Smad3 expression to interfere its induction on intercellular signal transduction and consequently ameliorate renal interstitial fibrosis.


Asunto(s)
Transdiferenciación Celular/efectos de los fármacos , Flavonoides/farmacología , Nefritis Intersticial/fisiopatología , Animales , Colágeno Tipo I/biosíntesis , Colágeno Tipo I/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Fibrosis , Inmunohistoquímica , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/metabolismo , Túbulos Renales/patología , Masculino , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , Mesodermo/patología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Crecimiento Transformador beta1/biosíntesis , Factor de Crecimiento Transformador beta1/genética
20.
Gene Expr Patterns ; 6(7): 719-23, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16466970

RESUMEN

Interaction of Eph receptor tyrosine kinases with their membrane bound ephrin ligands initiates bidirectional signaling events that regulate cell migratory and adhesive behavior. Whole-mount in situ hybridization revealed overlapping expression of the Epha1 receptor and its high-affinity ligands ephrin A1 (Efna1) and ephrin A3 (Efna3) in the primitive streak and the posterior paraxial mesoderm during early mouse development. These results show complex and dynamic expression for all three genes with expression domains that are successively complementary, overlapping, and divergent.


Asunto(s)
Desarrollo Embrionario/genética , Efrina-A1/genética , Efrina-A3/genética , Gástrula/metabolismo , Mesodermo/metabolismo , Receptor EphA1/genética , Animales , Efrina-A1/metabolismo , Efrina-A3/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Ligandos , Ratones , Organogénesis , Embarazo , Receptor EphA1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA