Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Rev Mol Cell Biol ; 16(2): 110-23, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25560970

RESUMEN

Retinoic acid (RA) signalling has a central role during vertebrate development. RA synthesized in specific locations regulates transcription by interacting with nuclear RA receptors (RARs) bound to RA response elements (RAREs) near target genes. RA was first implicated in signalling on the basis of its teratogenic effects on limb development. Genetic studies later revealed that endogenous RA promotes forelimb initiation by repressing fibroblast growth factor 8 (Fgf8). Insights into RA function in the limb serve as a paradigm for understanding how RA regulates other developmental processes. In vivo studies have identified RAREs that control repression of Fgf8 during body axis extension or activation of homeobox (Hox) genes and other key regulators during neuronal differentiation and organogenesis.


Asunto(s)
Extremidades/crecimiento & desarrollo , Transducción de Señal/genética , Tretinoina/metabolismo , Animales , Tipificación del Cuerpo/genética , Factor 8 de Crecimiento de Fibroblastos/genética , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Vertebrados/genética , Vertebrados/crecimiento & desarrollo , Vertebrados/metabolismo
2.
Genes Dev ; 31(13): 1325-1338, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28794185

RESUMEN

Deciphering the fundamental mechanisms controlling cardiac specification is critical for our understanding of how heart formation is initiated during embryonic development and for applying stem cell biology to regenerative medicine and disease modeling. Using systematic and unbiased functional screening approaches, we discovered that the Id family of helix-loop-helix proteins is both necessary and sufficient to direct cardiac mesoderm formation in frog embryos and human embryonic stem cells. Mechanistically, Id proteins specify cardiac cell fate by repressing two inhibitors of cardiogenic mesoderm formation-Tcf3 and Foxa2-and activating inducers Evx1, Grrp1, and Mesp1. Most importantly, CRISPR/Cas9-mediated ablation of the entire Id (Id1-4) family in mouse embryos leads to failure of anterior cardiac progenitor specification and the development of heartless embryos. Thus, Id proteins play a central and evolutionarily conserved role during heart formation and provide a novel means to efficiently produce cardiovascular progenitors for regenerative medicine and drug discovery applications.


Asunto(s)
Linaje de la Célula/genética , Corazón/embriología , Proteínas Inhibidoras de la Diferenciación/genética , Proteínas Inhibidoras de la Diferenciación/metabolismo , Organogénesis/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/genética , Línea Celular , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión no Mamífero/citología , Embrión no Mamífero/embriología , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Edición Génica , Regulación del Desarrollo de la Expresión Génica/genética , Cardiopatías Congénitas/genética , Humanos , Mesodermo/citología , Mesodermo/fisiología , Ratones , Mutación , Semillas , Xenopus laevis/embriología
3.
Front Neuroendocrinol ; 71: 101099, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37647946

RESUMEN

It is well established that retinoic acid receptors (RARs) function as nuclear receptors that control gene expression in response to binding of the ligand retinoic acid (RA). However, some studies have proposed that RAR-alpha (RARa) controls synaptic plasticity via non-genomic effects outside the nucleus, i.e. effects on mRNA translation of GluA1, a sub-unit of the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor. In order to support this non-genomic mechanism, studies have reported RARa knockout mice or treatment with pharmacological levels of RA and RAR antagonists to propose that RARa is required to control normal synaptic plasticity. A major shortcoming of the non-genomic hypothesis is that there have been no mutational studies showing that RARa can bind the GluA1 mRNA to control GLUA1 protein levels in a non-genomic manner. Also, without a genetic study that removes the endogenous ligand RA, it is impossible to conclude that RARa and its ligand RA control synaptic plasticity through a non-genomic signaling mechanism.


Asunto(s)
Receptores de Ácido Retinoico , Tretinoina , Ratones , Animales , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Ligandos , Tretinoina/metabolismo , Tretinoina/farmacología , Receptor alfa de Ácido Retinoico , Plasticidad Neuronal/fisiología
4.
PLoS Biol ; 18(5): e3000719, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32421711

RESUMEN

Identification of target genes that mediate required functions downstream of transcription factors is hampered by the large number of genes whose expression changes when the factor is removed from a specific tissue and the numerous binding sites for the factor in the genome. Retinoic acid (RA) regulates transcription via RA receptors bound to RA response elements (RAREs) of which there are thousands in vertebrate genomes. Here, we combined chromatin immunoprecipitation sequencing (ChIP-seq) for epigenetic marks and RNA-seq on trunk tissue from wild-type and Aldh1a2-/- embryos lacking RA synthesis that exhibit body axis and forelimb defects. We identified a relatively small number of genes with altered expression when RA is missing that also have nearby RA-regulated deposition of histone H3 K27 acetylation (H3K27ac) (gene activation mark) or histone H3 K27 trimethylation (H3K27me3) (gene repression mark) associated with conserved RAREs, suggesting these genes function downstream of RA. RA-regulated epigenetic marks were identified near RA target genes already known to be required for body axis and limb formation, thus validating our approach; plus, many other candidate RA target genes were found. Nuclear receptor 2f1 (Nr2f1) and nuclear receptor 2f2 (Nr2f2) in addition to Meis homeobox 1 (Meis1) and Meis homeobox 2 (Meis2) gene family members were identified by our approach, and double knockouts of each family demonstrated previously unknown requirements for body axis and/or limb formation. A similar epigenetic approach can be used to determine the target genes for any transcriptional regulator for which a knockout is available.


Asunto(s)
Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Estudios de Asociación Genética/métodos , Código de Histonas , Tretinoina/metabolismo , Animales , Secuencia de Bases , Secuenciación de Inmunoprecipitación de Cromatina , Secuencia Conservada , Epigénesis Genética , Ratones , Familia de Multigenes , Elementos de Respuesta , Análisis de Secuencia de ARN , Factores de Transcripción/metabolismo
5.
Cell ; 134(6): 921-31, 2008 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-18805086

RESUMEN

Retinoic acid, a derivative of vitamin A, is an essential component of cell-cell signaling during vertebrate organogenesis. In early development, retinoic acid organizes the trunk by providing an instructive signal for posterior neuroectoderm and foregut endoderm and a permissive signal for trunk mesoderm differentiation. At later stages, retinoic acid contributes to the development of the eye and other organs. Recent studies suggest that retinoic acid may act primarily in a paracrine manner and provide insight into the cell-cell signaling networks that control differentiation of pluripotent cells.


Asunto(s)
Organogénesis , Transducción de Señal , Tretinoina/metabolismo , Animales , Diferenciación Celular , Humanos
6.
Development ; 146(13)2019 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-31273085

RESUMEN

Retinoic acid (RA), a metabolite of retinol (vitamin A), functions as a ligand for nuclear RA receptors (RARs) that regulate development of chordate animals. RA-RARs can activate or repress transcription of key developmental genes. Genetic studies in mouse and zebrafish embryos that are deficient in RA-generating enzymes or RARs have been instrumental in identifying RA functions, revealing that RA signaling regulates development of many organs and tissues, including the body axis, spinal cord, forelimbs, heart, eye and reproductive tract. An understanding of the normal functions of RA signaling during development will guide efforts for use of RA as a therapeutic agent to improve human health. Here, we provide an overview of RA signaling and highlight its key functions during development.


Asunto(s)
Genes del Desarrollo , Receptores de Ácido Retinoico/fisiología , Tretinoina/farmacología , Tretinoina/fisiología , Animales , Embrión de Mamíferos , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Genes del Desarrollo/efectos de los fármacos , Genes del Desarrollo/genética , Humanos , Ratones , Receptores de Ácido Retinoico/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Tretinoina/metabolismo , Pez Cebra
7.
Mol Cell ; 53(6): 1005-19, 2014 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-24530304

RESUMEN

Here, we generated a genome-scale shRNA library targeting long intergenic noncoding RNAs (lincRNAs) in the mouse. We performed an unbiased loss-of-function study in mouse embryonic stem cells (mESCs) and identified 20 lincRNAs involved in the maintenance of pluripotency. Among these, TUNA (Tcl1 Upstream Neuron-Associated lincRNA, or megamind) was required for pluripotency and formed a complex with three RNA-binding proteins (RBPs). The TUNA-RBP complex was detected at the promoters of Nanog, Sox2, and Fgf4, and knockdown of TUNA or the individual RBPs inhibited neural differentiation of mESCs. TUNA showed striking evolutionary conservation of both sequence- and CNS-restricted expression in vertebrates. Accordingly, knockdown of tuna in zebrafish caused impaired locomotor function, and TUNA expression in the brains of Huntington's disease patients was significantly associated with disease grade. Our results suggest that the lincRNA TUNA plays a vital role in pluripotency and neural differentiation of ESCs and is associated with neurological function of adult vertebrates.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Enfermedad de Huntington/genética , Neuronas/metabolismo , Células Madre Pluripotentes/metabolismo , ARN Largo no Codificante/genética , Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Evolución Biológica , Diferenciación Celular , Secuencia Conservada , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Factor 4 de Crecimiento de Fibroblastos/genética , Factor 4 de Crecimiento de Fibroblastos/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Ratones , Datos de Secuencia Molecular , Actividad Motora , Proteína Homeótica Nanog , Neuronas/citología , Células Madre Pluripotentes/citología , Regiones Promotoras Genéticas , ARN Largo no Codificante/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Homología de Secuencia de Aminoácido , Índice de Severidad de la Enfermedad , Transducción de Señal , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo
9.
Alcohol Alcohol ; 55(1): 11-19, 2020 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-31825074

RESUMEN

AIMS: It is still unclear which enzymes contribute to the adaptive enhancement of alcohol metabolism by chronic alcohol consumption (CAC). ADH1 (Class I) has the lowest Km for ethanol and the highest sensitivity for 4-methylpyrazole (4MP) among ADH isozymes, while ADH3 (Class III) has the highest Km and the lowest sensitivity. We investigated how these two major ADHs relate to the adaptive enhancement of alcohol metabolism. METHODS: Male mice with different ADH genotypes (WT, Adh1-/- and Adh3-/-) were subjected to CAC experiment using a 10% ethanol solution for 1 month. Alcohol elimination rate (AER) was measured after ethanol injection at a 4.0 g/kg dose. 4MP-sensitive and -insensitive AERs were measured by the simultaneous administration of 4MP at a dose of 0.5 mmol/kg in order to estimate ADH1 and non-ADH1 pathways. RESULTS: AER was enhanced by CAC in all ADH genotypes, especially more than twofold in Adh1-/- mice, with increasing ADH1 and/or ADH3 liver contents, but not CYP2E1 content. 4MP-sensitive AER was also increased by CAC in WT and Adh3-/- strains, which was greater in Adh3-/- than in WT mice. The sensitive AER was increased even in Adh1-/- mice probably due to the increase in ADH3, which is semi-sensitive for 4MP. 4MP-insensitive AER was also increased in WT and Adh1-/- by CAC, but not in Adh3-/- mice. CONCLUSION: ADH1 contributes to the enhancement of alcohol metabolism by CAC, particularly in the absence of ADH3. ADH3 also contributes to the enhancement as a non-ADH1 pathway, especially in the absence of ADH1.


Asunto(s)
Alcohol Deshidrogenasa/fisiología , Eliminación Renal/fisiología , Alcohol Deshidrogenasa/genética , Consumo de Bebidas Alcohólicas/metabolismo , Animales , Etanol/metabolismo , Fomepizol/farmacología , Genotipo , Masculino , Ratones , Ratones Endogámicos , Eliminación Renal/efectos de los fármacos
10.
Genes Dev ; 26(23): 2567-79, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23152446

RESUMEN

Tight control over the segregation of endoderm, mesoderm, and ectoderm is essential for normal embryonic development of all species, yet how neighboring embryonic blastomeres can contribute to different germ layers has never been fully explained. We postulated that microRNAs, which fine-tune many biological processes, might modulate the response of embryonic blastomeres to growth factors and other signals that govern germ layer fate. A systematic screen of a whole-genome microRNA library revealed that the let-7 and miR-18 families increase mesoderm at the expense of endoderm in mouse embryonic stem cells. Both families are expressed in ectoderm and mesoderm, but not endoderm, as these tissues become distinct during mouse and frog embryogenesis. Blocking let-7 function in vivo dramatically affected cell fate, diverting presumptive mesoderm and ectoderm into endoderm. siRNA knockdown of computationally predicted targets followed by mutational analyses revealed that let-7 and miR-18 down-regulate Acvr1b and Smad2, respectively, to attenuate Nodal responsiveness and bias blastomeres to ectoderm and mesoderm fates. These findings suggest a crucial role for the let-7 and miR-18 families in germ layer specification and reveal a remarkable conservation of function from amphibians to mammals.


Asunto(s)
Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Genoma/genética , Estratos Germinativos/embriología , MicroARNs/metabolismo , Animales , Células Cultivadas , Análisis Mutacional de ADN , Células Madre Embrionarias , Técnicas de Silenciamiento del Gen , Ratones , MicroARNs/genética , Xenopus laevis
11.
Dev Biol ; 441(1): 127-131, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29964026

RESUMEN

In mouse, retinoic acid (RA) is required for the early phase of body axis extension controlled by a population of neuromesodermal progenitors (NMPs) in the trunk called expanding-NMPs, but not for the later phase of body axis extension controlled by a population of NMPs in the tail called depleting-NMPs. Recent observations suggest that zebrafish utilize depleting-NMPs but not expanding-NMPs for body axis extension. In zebrafish, a role for RA in body axis extension was not supported by previous studies on aldh1a2 (raldh2) mutants lacking RA synthesis. Here, by treating zebrafish embryos with an RA synthesis inhibitor, we also found that body axis extension and somitogenesis was not perturbed, although loss of pectoral fin and cardiac edema were observed consistent with previous studies. The conclusion that zebrafish diverges from mouse in not requiring RA for body axis extension is consistent with zebrafish lacking early expanding-NMPs to generate the trunk. We suggest that RA control of body axis extension was added to higher vertebrates during evolution of expanding-NMPs.


Asunto(s)
Embrión de Mamíferos/embriología , Embrión no Mamífero/embriología , Mesodermo/embriología , Células-Madre Neurales/metabolismo , Tretinoina/metabolismo , Pez Cebra/embriología , Animales , Embrión de Mamíferos/citología , Embrión no Mamífero/citología , Mesodermo/citología , Ratones , Células-Madre Neurales/citología , Especificidad de la Especie
12.
Dev Biol ; 418(1): 204-215, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27506116

RESUMEN

Retinoic acid (RA) repression of Fgf8 is required for many different aspects of organogenesis, however relatively little is known about how endogenous RA controls gene repression as opposed to gene activation. Here, we show that nuclear receptor corepressors NCOR1 and NCOR2 (SMRT) redundantly mediate the ability of RA to repress Fgf8. Ncor1;Ncor2 double mutants generated by CRISPR/Cas9 gene editing exhibited a small somite and distended heart phenotype similar to that of RA-deficient Raldh2-/- embryos, associated with increased Fgf8 expression and FGF signaling in caudal progenitors and heart progenitors. Embryo chromatin immunoprecipitation studies revealed that NCOR1/2 but not coactivators are recruited to the Fgf8 RA response element (RARE) in an RA-dependent manner, whereas coactivators but not NCOR1/2 are recruited RA-dependently to a RARE near Rarb that is activated by RA. CRISPR/Cas9-mediated genomic deletion of the Fgf8 RARE in mouse embryos often resulted in a small somite defect with Fgf8 derepression caudally, but no defect was observed in heart development or heart Fgf8 expression. This suggests the existence of another DNA element whose function overlaps with the Fgf8 RARE to mediate Fgf8 repression by RA and NCOR1/2. Our studies support a model in which NCOR1/2 mediates direct RA-dependent repression of Fgf8 in caudal progenitors in order to control somitogenesis.


Asunto(s)
Factor 8 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Co-Represor 1 de Receptor Nuclear/genética , Co-Represor 2 de Receptor Nuclear/genética , Organogénesis/genética , Somitos/anomalías , Tretinoina/metabolismo , Aldehído Oxidorreductasas/genética , Animales , Secuencia de Bases , Proteínas Co-Represoras/genética , Desarrollo Embrionario , Edición Génica/métodos , Corazón/embriología , Ratones , Ratones Noqueados , Transducción de Señal , Somitos/embriología
14.
Development ; 141(15): 2972-7, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25053430

RESUMEN

Retinoic acid (RA) generated in the mesoderm of vertebrate embryos controls body axis extension by downregulating Fgf8 expression in cells exiting the caudal progenitor zone. RA activates transcription by binding to nuclear RA receptors (RARs) at RA response elements (RAREs), but it is unknown whether RA can directly repress transcription. Here, we analyzed a conserved RARE upstream of Fgf8 that binds RAR isoforms in mouse embryos. Transgenic embryos carrying Fgf8 fused to lacZ exhibited expression similar to caudal Fgf8, but deletion of the RARE resulted in ectopic trunk expression extending into somites and neuroectoderm. Epigenetic analysis using chromatin immunoprecipitation of trunk tissues from E8.25 wild-type and Raldh2(-/-) embryos lacking RA synthesis revealed RA-dependent recruitment of the repressive histone marker H3K27me3 and polycomb repressive complex 2 (PRC2) near the Fgf8 RARE. The co-regulator RERE, the loss of which results in ectopic Fgf8 expression and somite defects, was recruited near the RARb RARE by RA, but was released from the Fgf8 RARE by RA. Our findings demonstrate that RA directly represses Fgf8 through a RARE-mediated mechanism that promotes repressive chromatin, thus providing valuable insight into the mechanism of RA-FGF antagonism during progenitor cell differentiation.


Asunto(s)
Tipificación del Cuerpo/genética , Factor 8 de Crecimiento de Fibroblastos/genética , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Tretinoina/metabolismo , Animales , Secuencia de Bases , Diferenciación Celular , Cromatina/metabolismo , Epigénesis Genética , Histonas/metabolismo , Ligandos , Mesodermo/embriología , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Isoformas de Proteínas/metabolismo , Receptores de Ácido Retinoico/metabolismo , Homología de Secuencia de Ácido Nucleico , Transducción de Señal , Somitos/embriología , Células Madre/citología
15.
Development ; 141(19): 3772-81, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25209250

RESUMEN

Although many regulatory networks involved in defining definitive endoderm have been identified, the mechanisms through which these networks interact to pattern the endoderm are less well understood. To explore the mechanisms involved in midgut patterning, we dissected the transcriptional regulatory elements of nephrocan (Nepn), the earliest known midgut specific gene in mice. We observed that Nepn expression is dramatically reduced in Sox17(-/-) and Raldh2(-/-) embryos compared with wild-type embryos. We further show that Nepn is directly regulated by Sox17 and the retinoic acid (RA) receptor via two enhancer elements located upstream of the gene. Moreover, Nepn expression is modulated by Activin signaling, with high levels inhibiting and low levels enhancing RA-dependent expression. In Foxh1(-/-) embryos in which Nodal signaling is reduced, the Nepn expression domain is expanded into the anterior gut region, confirming that Nodal signaling can modulate its expression in vivo. Together, Sox17 is required for Nepn expression in the definitive endoderm, while RA signaling restricts expression to the midgut region. A balance of Nodal/Activin signaling regulates the anterior boundary of the midgut expression domain.


Asunto(s)
Tipificación del Cuerpo/fisiología , Endodermo/fisiología , Tracto Gastrointestinal/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Redes Reguladoras de Genes/fisiología , Glicoproteínas/metabolismo , Transducción de Señal/fisiología , Activinas/metabolismo , Aldehído Oxidorreductasas/metabolismo , Animales , Ensayo de Cambio de Movilidad Electroforética , Redes Reguladoras de Genes/genética , Vectores Genéticos/genética , Proteínas HMGB/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Luciferasas , Ratones , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Ácido Retinoico/metabolismo , Factores de Transcripción SOXF/metabolismo
16.
Exp Eye Res ; 154: 190-195, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27840061

RESUMEN

Retinoic acid (RA) is a biologically active metabolite of vitamin A (retinol) that serves as an important signaling molecule in orchestrating diverse developmental processes including multiple roles during ocular development. Loss-of-function studies using gene knockouts of RA-synthesizing enzymes encoded by Aldh1a1, Aldh1a2, and Aldh1a3 (also known as Raldh1, Raldh2, and Raldh3) have provided valuable insight into how RA controls eye morphogenesis including corneal development. However, it is unclear whether endogenous RA is required for maintenance and regeneration of adult cornea. Here, we investigated the role of Aldh1a genes in the adult cornea using a novel conditional Aldh1a1,2,3-flox/flox;Rosa26-CreERT2 loss-of-function mouse model to determine the biological function of RA. Our findings indicate that loss of RA synthesis results in corneal thinning characterized by reduced thickness of the stromal layer, impaired corneal epithelial cell proliferation, and increased apoptosis. Corneal thinning in Aldh1a-deficient mice was significantly rescued by RA administration, indicating an important role of endogenous RA signaling in adult corneal homeostasis and regeneration. Thus, Aldh1a1,2,3-flox/flox;Rosa26-CreERT2 mice provide a useful model for investigating the mechanistic role of RA signaling in adult corneal maintenance and could provide new insights into therapeutic approaches for controlling corneal repair to prevent vision loss.


Asunto(s)
Apoptosis , Epitelio Corneal/metabolismo , Regeneración/fisiología , Tretinoina/metabolismo , Animales , Proliferación Celular , Epitelio Corneal/patología , Ratones , Transducción de Señal
17.
Dev Dyn ; 244(6): 797-807, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25809880

RESUMEN

BACKGROUND: Vertebrate body axis extension occurs in a head-to-tail direction from a caudal progenitor zone that responds to interacting signals. Wnt/ß-catenin signaling is critical for generation of paraxial mesoderm, somite formation, and maintenance of the axial stem cell pool. Body axis extension requires Wnt8a in lower vertebrates, but in mammals Wnt3a is required, although the anterior trunk develops in the absence of Wnt3a. RESULTS: We examined mouse Wnt8a(-/-) and Wnt3a(-/-) single and double mutants to explore whether mammalian Wnt8a contributes to body axis extension and to determine whether a posterior growth function for Wnt8a is conserved throughout the vertebrate lineage. We find that caudal Wnt8a is expressed only during early somite stages and is required for normal development of the anterior trunk in the absence of Wnt3a. During this time, we show that Wnt8a and Wnt3a cooperate to maintain Fgf8 expression and prevent premature Sox2 up-regulation in the axial stem cell niche, critical for posterior growth. Similar to Fgf8, Wnt8a requires retinoic acid (RA) signaling to restrict its caudal expression boundary and possesses an upstream RA response element that binds RA receptors. CONCLUSIONS: These findings provide new insight into interaction of caudal Wnt-FGF-RA signals required for body axis extension.


Asunto(s)
Tipificación del Cuerpo/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Nicho de Células Madre/fisiología , Proteína Wnt3A/fisiología , Anomalías Múltiples/embriología , Anomalías Múltiples/genética , Oxidorreductasas de Alcohol/deficiencia , Oxidorreductasas de Alcohol/genética , Animales , Tipificación del Cuerpo/genética , Secuencia Conservada , Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Factor 8 de Crecimiento de Fibroblastos/genética , Gastrulación , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Noqueados , Fenotipo , Receptores de Ácido Retinoico/fisiología , Elementos de Respuesta/genética , Factores de Transcripción SOXB1/biosíntesis , Factores de Transcripción SOXB1/genética , Transducción de Señal/fisiología , Somitos/crecimiento & desarrollo , Somitos/metabolismo , Tretinoina/farmacología , Vertebrados/embriología , Proteínas Wnt , Proteína Wnt3A/deficiencia , Proteína Wnt3A/genética
18.
Semin Cell Dev Biol ; 24(10-12): 694-700, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23973941

RESUMEN

The vitamin A metabolite retinoic acid (RA) serves as a ligand for nuclear RA receptors that control differentiation of progenitor cells important for vertebrate development. Genetic studies in mouse embryos deficient for RA-generating enzymes have been invaluable for deciphering RA function. RA first begins to act during early organogenesis when RA generated in trunk mesoderm begins to function as a diffusible signal controlling progenitor cell differentiation. In neuroectoderm, RA functions as an instructive signal to stimulate neuronal differentiation of progenitor cells in the hindbrain and spinal cord. RA is not required for early neuronal differentiation of the forebrain, but at later stages RA stimulates neuronal differentiation in forebrain basal ganglia. RA also acts as a permissive signal for differentiation by repressing fibroblast growth factor (FGF) signaling in differentiated cells as they emerge from progenitor populations in the caudal progenitor zone and second heart field. In addition, RA signaling stimulates differentiation of spermatogonial germ cells and induces meiosis in male but not female gonads. A more complete understanding of the normal functions of RA signaling during development will guide efforts to use RA as a differentiation agent for therapeutic purposes.


Asunto(s)
Diferenciación Celular , Desarrollo Embrionario , Retinoides/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo , Animales , Células Germinativas/citología , Ratones
19.
Hepatology ; 60(3): 1044-53, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24668648

RESUMEN

UNLABELLED: The important roles of retinols and their metabolites have recently been emphasized in the interactions between hepatic stellate cells (HSCs) and natural killer (NK) cells. Nevertheless, the expression and role of retinol metabolizing enzyme in both cell types have yet to be clarified. Thus, we investigated the expression of retinol metabolizing enzyme and its role in liver fibrosis. Among several retinol metabolizing enzymes, only alcohol dehydrogenase (ADH) 3 expression was detected in isolated HSCs and NK cells, whereas hepatocytes express all of them. In vitro treatment with 4-methylpyrazole (4-MP), a broad ADH inhibitor, or depletion of the ADH3 gene down-regulated collagen and transforming growth factor-ß1 (TGF-ß1) gene expression, but did not affect α-smooth muscle actin gene expression in cultured HSCs. Additionally, in vitro, treatments with retinol suppressed NK cell activities, whereas inhibition of ADH3 enhanced interferon-γ (IFN-γ) production and cytotoxicity of NK cells against HSCs. In vivo, genetic depletion of the ADH3 gene ameliorated bile duct ligation- and carbon tetrachloride-induced liver fibrosis, in which a higher number of apoptotic HSCs and an enhanced activation of NK cells were detected. Freshly isolated HSCs from ADH3-deficient mice showed reduced expression of collagen and TGF-ß1, but enhanced expression of IFN-γ was detected in NK cells from these mice compared with those of control mice. Using reciprocal bone marrow transplantation of wild-type and ADH3-deficient mice, we demonstrated that ADH3 deficiency in both HSCs and NK cells contributed to the suppressed liver fibrosis. CONCLUSION: ADH3 plays important roles in promoting liver fibrosis by enhancing HSC activation and inhibiting NK cell activity, and could be used as a potential therapeutic target for the treatment of liver fibrosis.


Asunto(s)
Aldehído Oxidorreductasas/metabolismo , Células Estrelladas Hepáticas/fisiología , Células Asesinas Naturales/fisiología , Cirrosis Hepática/enzimología , Animales , Trasplante de Médula Ósea , Interferón gamma/metabolismo , Cirrosis Hepática/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL
20.
Dev Biol ; 381(1): 28-37, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23806210

RESUMEN

The enteric nervous system (ENS) forms from the neural crest-derived precursors that colonize the bowel before differentiating into a network of neurons and glia that control intestinal function. Retinoids are essential for normal ENS development, but the role of retinoic acid (RA) metabolism in development remains incompletely understood. Because RA is produced locally in the tissues where it acts by stimulating RAR and RXR receptors, RA signaling during development is absolutely dependent on the rate of RA synthesis and degradation. RA is produced by three different enzymes called retinaldehyde dehydrogenases (RALDH1, RALDH2 and RALDH3) that are all expressed in the developing bowel. To determine the relative importance of these enzymes for ENS development, we analyzed whole mount preparations of adult (8-12-week old) myenteric and submucosal plexus stained with NADPH diaphorase (neurons and neurites), anti-TuJ1 (neurons and neurites), anti-HuC/HuD (neurons), and anti-S100ß (glia) in an allelic series of mice with mutations in Raldh1, Raldh2, and Raldh3. We found that Raldh1-/-, Raldh2+/-, Raldh3+/- (R1(KO)R2(Het)R3(Het)) mutant mice had a reduced colon myenteric neuron density, reduced colon myenteric neuron to glia ratio, reduced colon submucosal neuron density, and increased colon myenteric fibers per neuron when compared to the wild type (WT; Raldh1WT, Raldh2WT, Raldh3WT) mice. These defects are unlikely to be due to defective ENS precursor migration since R1(KO)R2(Het)R3(KO) mice had increased enteric neuron progenitor migration into the distal colon compared to WT during development. RALDH mutant mice also have reduced contractility in the colon compared to WT mice. These data suggest that RALDH1, RALDH2 and RALDH3 each contribute to ENS development and function.


Asunto(s)
Aldehído Oxidorreductasas/fisiología , Colon/inervación , Sistema Nervioso Entérico/metabolismo , Isoenzimas/fisiología , Retinal-Deshidrogenasa/fisiología , Familia de Aldehído Deshidrogenasa 1 , Animales , Movimiento Celular , Colon/enzimología , Suplementos Dietéticos , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Mutación , Neuroglía/citología , Neuronas/metabolismo , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA