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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 10(11): 1193-9, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19783990

RESUMEN

The location of embryonic lymph node development is determined by the initial clustering of lymphoid tissue-inducer (LTi) cells. Here we demonstrate that both the chemokine CXCL13 and the chemokine CCL21 attracted LTi cells at embryonic days 12.5-14.5 and that initial clustering depended exclusively on CXCL13. Retinoic acid (RA) induced early CXCL13 expression in stromal organizer cells independently of lymphotoxin signaling. Notably, neurons adjacent to the lymph node anlagen expressed enzymes essential for RA synthesis. Furthermore, stimulation of parasymphathetic neural output in adults led to RA receptor (RAR)-dependent induction of CXCL13 in the gut. Therefore, our data show that the initiation of lymph node development is controlled by RA-mediated expression of CXCL13 and suggest that RA may be provided by adjacent neurons.


Asunto(s)
Quimiocina CXCL13/metabolismo , Ganglios Linfáticos/embriología , Neuronas/metabolismo , Tretinoina/metabolismo , Aldehído Deshidrogenasa/metabolismo , Familia de Aldehído Deshidrogenasa 1 , Animales , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Quimiocina CCL21/metabolismo , Embrión de Mamíferos/embriología , Femenino , Isoenzimas/metabolismo , Tejido Linfoide/embriología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Retinal-Deshidrogenasa , Células del Estroma/metabolismo , Estimulación del Nervio Vago
2.
Eur J Oral Sci ; 125(1): 8-17, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28084688

RESUMEN

Latent-transforming growth factor beta-binding protein 3 (LTBP-3) is important for craniofacial morphogenesis and hard tissue mineralization, as it is essential for activation of transforming growth factor-ß (TGF-ß). To investigate the role of LTBP-3 in tooth formation we performed micro-computed tomography (micro-CT), histology, and scanning electron microscopy analyses of adult Ltbp3-/- mice. The Ltbp3-/- mutants presented with unique craniofacial malformations and reductions in enamel formation that began at the matrix formation stage. Organization of maturation-stage ameloblasts was severely disrupted. The lateral side of the incisor was affected most. Reduced enamel mineralization, modification of the enamel prism pattern, and enamel nodules were observed throughout the incisors, as revealed by scanning electron microscopy. Molar roots had internal irregular bulbous-like formations. The cementum thickness was reduced, and microscopic dentinal tubules showed minor nanostructural changes. Thus, LTBP-3 is required for ameloblast differentiation and for the formation of decussating enamel prisms, to prevent enamel nodule formation, and for proper root morphogenesis. Also, and consistent with the role of TGF-ß signaling during mineralization, almost all craniofacial bone components were affected in Ltbp3-/- mice, especially those involving the upper jaw and snout. This mouse model demonstrates phenotypic overlap with Verloes Bourguignon syndrome, also caused by mutation of LTBP3, which is hallmarked by craniofacial anomalies and amelogenesis imperfecta phenotypes.


Asunto(s)
Amelogénesis/genética , Esmalte Dental/anomalías , Proteínas de Unión a TGF-beta Latente/genética , Ameloblastos/metabolismo , Amelogénesis Imperfecta/genética , Animales , Esmalte Dental/ultraestructura , Genotipo , Masculino , Ratones , Ratones Mutantes , Microscopía Electrónica de Rastreo , Mutación , Osteocondrodisplasias/genética , Fenotipo , Calcificación de Dientes/genética , Factor de Crecimiento Transformador beta/genética , Microtomografía por Rayos X
3.
Dev Dyn ; 245(3): 388-401, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26442704

RESUMEN

BACKGROUND: Retinoic acid (RA), the bioactive derivative of vitamin A, is essential for vertebrate heart development. Both excess and reduced RA signaling lead to cardiovascular malformations affecting the outflow tract (OFT). To address the cellular mechanisms underlying the effects of RA signaling during OFT morphogenesis, we used transient maternal RA supplementation to rescue the early lethality resulting from inactivation of the murine retinaldehyde dehydrogenase 2 (Raldh2) gene. RESULTS: By embryonic day 13.5, all rescued Raldh2(-/-) hearts exhibit severe, reproducible OFT septation defects, although wild-type and Raldh2(+/-) littermates have normal hearts. Cardiac neural crest cells (cNCC) were present in OFT cushions of Raldh2(-/-) mutant embryos but ectopically located in the periphery of the endocardial cushions, rather than immediately underlying the endocardium. Excess mesenchyme was generated by Raldh2(-/-) mutant endocardium, which displaced cNCC derivatives from their subendocardial, medial position. CONCLUSIONS: RA signaling affects not only cNCC numbers but also their position relative to endocardial mesenchyme during the septation process. Our study shows that inappropriate coordination between the different cell types of the OFT perturbs its morphogenesis and leads to a severe congenital heart defect, persistent truncus arteriosus.


Asunto(s)
Aldehído Oxidorreductasas/metabolismo , Linaje de la Célula/fisiología , Corazón/embriología , Organogénesis/fisiología , Transducción de Señal/fisiología , Tretinoina/metabolismo , Aldehído Oxidorreductasas/genética , Animales , Ratones , Ratones Noqueados
4.
Nat Rev Genet ; 9(7): 541-53, 2008 07.
Artículo en Inglés | MEDLINE | ID: mdl-18542081

RESUMEN

Retinoic acid (RA) has complex and pleiotropic functions during vertebrate development. Recent work in several species has increased our understanding of the roles of RA as a signalling molecule. These functions rely on a tight control of RA distribution within embryonic tissues through the combined action of synthesizing and metabolizing enzymes, possibly leading to diffusion gradients. Also important is the switching of nuclear receptors from a transcriptionally repressing state to an activating state. In addition, cross-talk with other key embryonic signals, especially fibroblast growth factors (FGFs) and sonic hedgehog (SHH), is being uncovered. Some of these functions could be maintained throughout the life of an organism to regulate cell-lineage decisions and/or the differentiation of stem cell populations, highlighting possibilities for regenerative medicine.


Asunto(s)
Tretinoina/metabolismo , Vertebrados/embriología , Vertebrados/metabolismo , Animales , Diferenciación Celular , Humanos , Células Madre/citología
5.
Prenat Diagn ; 34(13): 1312-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25088217

RESUMEN

OBJECTIVE: Increased nuchal translucency originates from disturbed lymphatic development. Abnormal neural crest cell (NCC) migration may be involved in lymphatic development. Because both neuronal and lymphatic development share retinoic acid (RA) as a common factor, this study investigated the involvement of NCCs and RA in specific steps in lymphatic endothelial cell (LEC) differentiation and nuchal edema, which is the morphological equivalent of increased nuchal translucency. METHODS: Mouse embryos in which all NCCs were fluorescently labeled (Wnt1-Cre;Rosa26(eYfp) ), reporter embryos for in vivo RA activity (DR5-luciferase) and embryos with absent (Raldh2(-/-) ) or in utero inhibition of RA signaling (BMS493) were investigated. Immunofluorescence using markers for blood vessels, lymphatic endothelium and neurons was applied. Flow cytometry was performed to measure specific LEC populations. RESULTS: Cranial nerves were consistently close to the jugular lymph sac (JLS), in which NCCs were identified. In the absence of RA synthesis, enlarged JLS and nuchal edema were observed. Inhibiting RA signaling in utero resulted in a significantly higher amount of precursor-LECs at the expense of mature LECs and caused nuchal edema. CONCLUSIONS: Neural crest cells are involved in lymphatic development. RA is required for differentiation into mature LECs. Blocking RA signaling in mouse embryos results in abnormal lymphatic development and nuchal edema.


Asunto(s)
Vasos Linfáticos/embriología , Cresta Neural/fisiología , Tretinoina/metabolismo , Animales , Diferenciación Celular , Células Endoteliales/citología , Femenino , Vasos Linfáticos/citología , Vasos Linfáticos/metabolismo , Ratones , Medida de Translucencia Nucal , Embarazo
6.
Proc Natl Acad Sci U S A ; 108(40): 16687-92, 2011 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-21930923

RESUMEN

Retinoic acid (RA), an active vitamin A metabolite, is a key signaling molecule in vertebrate embryos. Morphogenetic RA gradients are thought to be set up by tissue-specific actions of retinaldehyde dehydrogenases (RALDHs) and catabolizing enzymes. According to the species, two enzymatic pathways (ß-carotene cleavage and retinol oxidation) generate retinaldehyde, the substrate of RALDHs. Placental species depend on maternal retinol transferred to the embryo. The retinol-to-retinaldehyde conversion was thought to be achieved by several redundant enzymes; however, a random mutagenesis screen identified retinol dehydrogenase 10 [Rdh10(Trex) allele; Sandell LL, et al. (2007) Genes Dev 21:1113-1124] as responsible for a homozygous lethal phenotype with features of RA deficiency. We report here the production and characterization of unique murine Rdh10 loss-of-function alleles generated by gene targeting. We show that although Rdh10(-/-) mutants die at an earlier stage than Rdh10(Trex) mutants, their molecular patterning defects do not reflect a complete state of RA deficiency. Furthermore, we were able to correct most developmental abnormalities by administering retinaldehyde to pregnant mothers, thereby obtaining viable Rdh10(-/-) mutants. This demonstrates the rescue of an embryonic lethal phenotype by simple maternal administration of the missing retinoid compound. These results underscore the importance of maternal retinoids in preventing congenital birth defects, and lead to a revised model of the importance of RDH10 and RALDHs in controlling embryonic RA distribution.


Asunto(s)
Oxidorreductasas de Alcohol/metabolismo , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/fisiología , Región Branquial/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Rombencéfalo/embriología , Transducción de Señal/fisiología , Oxidorreductasas de Alcohol/genética , Animales , Tipificación del Cuerpo/efectos de los fármacos , Región Branquial/metabolismo , Galactósidos , Regulación del Desarrollo de la Expresión Génica/genética , Marcación de Gen , Técnicas Histológicas , Hibridación in Situ , Indoles , Ratones , Ratones Noqueados , Modelos Biológicos , Retinaldehído/metabolismo , Retinaldehído/farmacología , Rombencéfalo/metabolismo , Tretinoina/metabolismo , Vitamina A/metabolismo
7.
Nat Genet ; 37(10): 1082-9, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16186816

RESUMEN

Removal of toxic substances from the blood depends on patent connections between the kidney, ureters and bladder that are established when the ureter is transposed from its original insertion site in the male genital tract to the bladder. This transposition is thought to occur as the trigone forms from the common nephric duct and incorporates into the bladder. Here we re-examine this model in the context of normal and abnormal development. We show that the common nephric duct does not differentiate into the trigone but instead undergoes apoptosis, a crucial step for ureter transposition controlled by vitamin A-induced signals from the primitive bladder. Ureter abnormalities occur in 1-2% of the human population and can cause obstruction and end-stage renal disease. These studies provide an explanation for ureter defects underlying some forms of obstruction in humans and redefine the current model of ureter maturation.


Asunto(s)
Apoptosis , Nefronas/embriología , Uréter/embriología , Vejiga Urinaria/embriología , Vitamina A/fisiología , Animales , Proteínas de Homeodominio/genética , Ratones , Ratones Transgénicos , Nefronas/citología , Organogénesis/genética , Transducción de Señal
8.
Sci Rep ; 14(1): 445, 2024 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-38172607

RESUMEN

Kohlschütter-Tönz syndrome (KTS) is a rare autosomal recessive disorder characterized by severe intellectual disability, early-onset epileptic seizures, and amelogenesis imperfecta. Here, we present a novel Rogdi mutant mouse deleting exons 6-11- a mutation found in KTS patients disabling ROGDI function. This Rogdi-/- mutant model recapitulates most KTS symptoms. Mutants displayed pentylenetetrazol-induced seizures, confirming epilepsy susceptibility. Spontaneous locomotion and circadian activity tests demonstrate Rogdi mutant hyperactivity mirroring patient spasticity. Object recognition impairment indicates memory deficits. Rogdi-/- mutant enamel was markedly less mature. Scanning electron microscopy confirmed its hypomineralized/hypomature crystallization, as well as its low mineral content. Transcriptomic RNA sequencing of postnatal day 5 lower incisors showed downregulated enamel matrix proteins Enam, Amelx, and Ambn. Enamel crystallization appears highly pH-dependent, cycling between an acidic and neutral pH during enamel maturation. Rogdi-/- teeth exhibit no signs of cyclic dental acidification. Additionally, expression changes in Wdr72, Slc9a3r2, and Atp6v0c were identified as potential contributors to these tooth acidification abnormalities. These proteins interact through the acidifying V-ATPase complex. Here, we present the Rogdi-/- mutant as a novel model to partially decipher KTS pathophysiology. Rogdi-/- mutant defects in acidification might explain the unusual combination of enamel and rare neurological disease symptoms.


Asunto(s)
Amelogénesis Imperfecta , Demencia , Epilepsia , Anomalías Dentarias , Humanos , Animales , Ratones , Amelogénesis Imperfecta/genética , Convulsiones , Mutación , Proteínas de la Membrana/genética , Proteínas Nucleares/genética
9.
Development ; 137(2): 283-92, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20040494

RESUMEN

In humans and mice, mutations in the Ret gene result in Hirschsprung's disease and renal defects. In the embryonic kidney, binding of Ret to its ligand, Gdnf, induces a program of epithelial cell remodeling that controls primary branch formation and branching morphogenesis within the kidney. Our previous studies showed that transcription factors belonging to the retinoic acid (RA) receptor family are crucial for controlling Ret expression in the ureteric bud; however, the mechanism by which retinoid-signaling acts has remained unclear. In the current study, we show that expression of a dominant-negative RA receptor in mouse ureteric bud cells abolishes Ret expression and Ret-dependent functions including ureteric bud formation and branching morphogenesis, indicating that RA-receptor signaling in ureteric bud cells is crucial for renal development. Conversely, we find that RA-receptor signaling in ureteric bud cells depends mainly on RA generated in nearby stromal cells by retinaldehyde dehydrogenase 2, an enzyme required for most fetal RA synthesis. Together, these studies suggest that renal development depends on paracrine RA signaling between stromal mesenchyme and ureteric bud cells that regulates Ret expression both during ureteric bud formation and within the developing collecting duct system.


Asunto(s)
Riñón/embriología , Retinoides/metabolismo , Transducción de Señal , Aldehído Oxidorreductasas/genética , Aldehído Oxidorreductasas/fisiología , Animales , Células Cultivadas , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos , Inmunoquímica , Hibridación in Situ , Masculino , Ratones , Morfogénesis/genética , Morfogénesis/fisiología , Técnicas de Cultivo de Órganos , Retinal-Deshidrogenasa/genética , Retinal-Deshidrogenasa/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Proc Natl Acad Sci U S A ; 107(20): 9234-9, 2010 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-20439714

RESUMEN

Retinoic acid (RA) has several established functions during cardiac development, including actions in the fetal epicardium required for myocardial growth. An open question is if retinoid effects are limited to growth factor stimulation pathway(s) or if additional actions on uncommitted progenitor/stem populations might drive cardiac differentiation. Here we report the dual effects of RA deficiency on cardiac growth factor signaling and progenitor/stem biology using the mouse retinaldehyde dehydrogenase 2 (Raldh2) knockout model. Although early heart defects in Raldh2(-/-) embryos result from second-heart-field abnormalities, it is unclear whether this role is transient or whether RA has sustained effects on cardiac progenitors. To address this, we used transient maternal RA supplementation to overcome early Raldh2(-/-) lethality. By embryonic day 11.5-14.5, Raldh2(-/-) hearts exhibited reduced venticular compact layer outgrowth and altered coronary vessel development. Although reductions in Fgf2 and target pERK levels occurred, no alterations in Wnt/beta-catenin expression were observed. Cell proliferation is increased in compact zone myocardium, whereas cardiomyocyte differentiation is reduced, alterations that suggest progenitor defects. We report that the fetal heart contains a reservoir of stem/progenitor cells, which can be isolated by their ability to efflux a fluorescent dye and that retinoid signaling acts on this fetal cardiac side population (SP). Raldh2(-/-) hearts display increased SP cell numbers, with selective increases in expression of cardiac progenitor cell markers and reduced differentiation marker levels. Hence, although lack of RA signaling increases cardiac SP numbers, simultaneous reductions in Fgf signaling reduce cardiomyocyte differentiation, possibly accounting for long-term defects in myocardial growth.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Corazón/embriología , Transducción de Señal/efectos de los fármacos , Células Madre/efectos de los fármacos , Tretinoina/farmacología , Aldehído Oxidorreductasas/genética , Animales , Diferenciación Celular/fisiología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Citometría de Flujo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología , Células Madre/fisiología , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
11.
Nat Genet ; 31(1): 84-8, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11953746

RESUMEN

Retinoic acid, the active derivative of vitamin A (retinol), is a hormonal signaling molecule that acts in developing and adult tissues. The Cyp26a1 (cytochrome p450, 26) protein metabolizes retinoic acid into more polar hydroxylated and oxidized derivatives. Whether some of these derivatives are biologically active metabolites has been debated. Cyp26a1(-/-) mouse fetuses have lethal morphogenetic phenotypes mimicking those generated by excess retinoic acid administration, indicating that human CYP26A1 may be essential in controlling retinoic acid levels during development. This hypothesis suggests that the Cyp26a1(-/-) phenotype could be rescued under conditions in which embryonic retinoic acid levels are decreased. We show that Cyp26a1(-/-) mice are phenotypically rescued by heterozygous disruption of Aldh1a2 (also known as Raldh2), which encodes a retinaldehyde dehydrogenase responsible for the synthesis of retinoic acid during early embryonic development. Aldh1a2 haploinsufficiency prevents the appearance of spina bifida and rescues the development of posterior structures (sacral/caudal vertebrae, hindgut, urogenital tract), while partly preventing cervical vertebral transformations and hindbrain pattern alterations in Cyp26a1(-/-) mice. Thus, some of these double-mutant mice can reach adulthood. This study is the first report of a mutation acting as a dominant suppressor of a lethal morphogenetic mutation in mammals. We provide genetic evidence that ALDH1A2 and CYP26A1 activities concurrently establish local embryonic retinoic acid levels that must be finely tuned to allow posterior organ development and to prevent spina bifida.


Asunto(s)
Aldehído Oxidorreductasas/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Desarrollo Embrionario y Fetal/fisiología , Oxigenasas de Función Mixta/metabolismo , Retinoides/metabolismo , Tretinoina/metabolismo , Aldehído Oxidorreductasas/deficiencia , Aldehído Oxidorreductasas/genética , Animales , Sistema Enzimático del Citocromo P-450/deficiencia , Sistema Enzimático del Citocromo P-450/genética , Desarrollo Embrionario y Fetal/genética , Genes Reporteros , Heterocigoto , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Oxigenasas de Función Mixta/deficiencia , Oxigenasas de Función Mixta/genética , Oxidación-Reducción , Fenotipo , Retinal-Deshidrogenasa , Ácido Retinoico 4-Hidroxilasa , Transducción de Señal , Disrafia Espinal/genética , Disrafia Espinal/prevención & control
12.
Dev Biol ; 353(2): 266-74, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21385575

RESUMEN

Much of the heart, including the atria, right ventricle and outflow tract (OFT) is derived from a progenitor cell population termed the second heart field (SHF) that contributes progressively to the embryonic heart during cardiac looping. Several studies have revealed anterior-posterior patterning of the SHF, since the anterior region (anterior heart field) contributes to right ventricular and OFT myocardium whereas the posterior region gives rise to the atria. We have previously shown that Retinoic Acid (RA) signal participates to this patterning. We now show that Hoxb1, Hoxa1, and Hoxa3, as downstream RA targets, are expressed in distinct sub-domains within the SHF. Our genetic lineage tracing analysis revealed that Hoxb1, Hoxa1 and Hoxa3-expressing cardiac progenitor cells contribute to both atria and the inferior wall of the OFT, which subsequently gives rise to myocardium at the base of pulmonary trunk. By contrast to Hoxb1(Cre), the contribution of Hoxa1-enhIII-Cre and Hoxa3(Cre)-labeled cells is restricted to the distal regions of the OFT suggesting that proximo-distal patterning of the OFT is related to SHF sub-domains characterized by combinatorial Hox genes expression. Manipulation of RA signaling pathways showed that RA is required for the correct deployment of Hox-expressing SHF cells. This report provides new insights into the regulatory gene network in SHF cells contributing to the atria and sub-pulmonary myocardium.


Asunto(s)
Corazón Fetal/embriología , Corazón Fetal/metabolismo , Genes Homeobox , Aldehído Oxidorreductasas/deficiencia , Aldehído Oxidorreductasas/genética , Animales , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/fisiología , Linaje de la Célula , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Femenino , Corazón Fetal/citología , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Hibridación in Situ , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Cardiovasculares , Embarazo , Transducción de Señal , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Tretinoina/metabolismo
13.
Circ Res ; 106(4): 686-94, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-20110535

RESUMEN

RATIONALE: Loss of Tbx1 and decrease of retinoic acid (RA) synthesis result in DiGeorge/velocardiofacial syndrome (DGS/VCFS)-like phenotypes in mouse models, including defects in septation of the outflow tract of the heart and anomalies of pharyngeal arch-derived structures including arteries of the head and neck, laryngeal-tracheal cartilage, and thymus/parathyroid. Wild-type levels of T-box transcription factor (Tbx)1 and RA signaling are required for normal pharyngeal arch artery development. Recent studies have shown that reduction of RA or loss of Tbx1 alters the contribution of second heart field (SHF) progenitor cells to the elongating heart tube. OBJECTIVE: Here we tested whether Tbx1 and the RA signaling pathway interact during the deployment of the SHF and formation of the mature aortic arch. METHODS AND RESULTS: Molecular markers of the SHF, neural crest and smooth muscle cells, were analyzed in Raldh2;Tbx1 compound heterozygous mutants. Our results revealed that the SHF and outflow tract develop normally in Raldh2(+/-);Tbx1(+/-) embryos. However, we found that decreased levels of RA accelerate the recovery from arterial growth delay observed in Tbx1(+/-) mutant embryos. This compensation coincides with the differentiation of smooth muscle cells in the 4th pharyngeal arch arteries, and is associated with severity of neural crest cell migration defects observed in these mutants. CONCLUSIONS: Our data suggest that differences in levels of embryonic RA may contribute to the variability in great artery anomalies observed in DGS/VCFS patients.


Asunto(s)
Aorta Torácica/metabolismo , Región Branquial/metabolismo , Síndrome de DiGeorge/metabolismo , Músculo Liso Vascular/metabolismo , Transducción de Señal , Tretinoina/metabolismo , Aldehído Oxidorreductasas/genética , Aldehído Oxidorreductasas/metabolismo , Animales , Aorta Torácica/anomalías , Región Branquial/anomalías , Diferenciación Celular , Cruzamientos Genéticos , Síndrome de DiGeorge/embriología , Síndrome de DiGeorge/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo , Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genotipo , Edad Gestacional , Corazón/embriología , Heterocigoto , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Músculo Liso Vascular/anomalías , Mutación , Cresta Neural/anomalías , Cresta Neural/metabolismo , Fenotipo , Transducción de Señal/genética , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Técnicas de Cultivo de Tejidos
14.
Circ Res ; 106(7): 1212-20, 2010 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-20185795

RESUMEN

RATIONALE: The cardiac venous pole is a common focus of congenital malformations and atrial arrhythmias, yet little is known about the cellular and molecular mechanisms that regulate its development. The systemic venous return myocardium (sinus node and sinus horns) forms only late in cardiogenesis from a pool of pericardial mesenchymal precursor cells. OBJECTIVE: To analyze the cellular and molecular mechanisms directing the formation of the fetal sinus horns. METHODS AND RESULTS: We analyzed embryos deficient for the Wt1 (Wilms tumor 1) gene and observed a failure to form myocardialized sinus horns. Instead, the cardinal veins become embedded laterally in the pleuropericardial membranes that remain tethered to the lateral body wall by the persisting subcoelomic mesenchyme, a finding that correlates with decreased apoptosis in this region. We show by expression analysis and lineage tracing studies that Wt1 is expressed in the subcoelomic mesenchyme surrounding the cardinal veins, but that this Wt1-positive mesenchyme does not contribute cells to the sinus horn myocardium. Expression of the Raldh2 (aldehyde dehydrogenase family 1, subfamily A2) gene was lost from this mesenchyme in Wt1(-/-) embryos. Phenotypic analysis of Raldh2 mutant mice rescued from early cardiac defects by retinoic acid food supply revealed defects of the venous pole and pericardium highly similar to those of Wt1(-/-) mice. CONCLUSIONS: Pericardium and sinus horn formation are coupled and depend on the expansion and correct temporal release of pleuropericardial membranes from the underlying subcoelomic mesenchyme. Wt1 and downstream Raldh2/retinoic acid signaling are crucial regulators of this process. Thus, our results provide novel insight into the genetic and cellular pathways regulating the posterior extension of the mammalian heart and the formation of its coelomic lining.


Asunto(s)
Seno Coronario/metabolismo , Mesodermo/metabolismo , Pericardio/metabolismo , Pleura/metabolismo , Transducción de Señal , Nodo Sinoatrial/metabolismo , Tretinoina/metabolismo , Proteínas WT1/metabolismo , Aldehído Oxidorreductasas/genética , Aldehído Oxidorreductasas/metabolismo , Animales , Apoptosis , Linaje de la Célula , Seno Coronario/embriología , Muerte Fetal , Regulación del Desarrollo de la Expresión Génica , Genotipo , Edad Gestacional , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación , Pericardio/embriología , Fenotipo , Pleura/embriología , Transducción de Señal/genética , Nodo Sinoatrial/embriología , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Proteínas WT1/deficiencia , Proteínas WT1/genética
15.
Proc Natl Acad Sci U S A ; 105(8): 2913-8, 2008 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-18287057

RESUMEN

Retinoic acid (RA), the active derivative of vitamin A, has been implicated in various steps of cardiovascular development. The retinaldehyde dehydrogenase 2 (RALDH2) enzyme catalyzes the second oxidative step in RA biosynthesis and its loss of function creates a severe embryonic RA deficiency. Raldh2(-/-) knockout embryos fail to undergo heart looping and have impaired atrial and sinus venosus development. To understand the mechanism(s) producing these changes, we examined the contribution of the second heart field (SHF) to pharyngeal mesoderm, atria, and outflow tract in Raldh2(-/-) embryos. RA deficiency alters SHF gene expression in two ways. First, Raldh2(-/-) embryos exhibited a posterior expansion of anterior markers of the SHF, including Tbx1, Fgf8, and the Mlc1v-nlacZ-24/Fgf10 reporter transgene as well as of Islet1. This occurred at early somite stages, when cardiac defects became irreversible in an avian vitamin A-deficiency model, indicating that endogenous RA is required to restrict the SHF posteriorly. Explant studies showed that this expanded progenitor population cannot differentiate properly. Second, RA up-regulated cardiac Bmp expression levels at the looping stage. The contribution of the SHF to both inflow and outflow poles was perturbed under RA deficiency, creating a disorganization of the heart tube. We also investigated genetic cross-talk between Nkx2.5 and RA signaling by generating double mutant mice. Strikingly, Nkx2.5 deficiency was able to rescue molecular defects in the posterior region of the Raldh2(-/-) mutant heart, in a gene dosage-dependent manner.


Asunto(s)
Aldehído Oxidorreductasas/genética , Regulación del Desarrollo de la Expresión Génica/genética , Corazón/embriología , Tretinoina/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Dosificación de Gen , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Noqueados , Transducción de Señal/genética
16.
Dev Dyn ; 239(12): 3260-74, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21046629

RESUMEN

Retinoic acid (RA), a vitamin A derivative, is synthesized by specific cell populations and acts as a diffusible embryonic signal activating ligand-inducible transcription factors, the RA receptors (RARs). RA-activatable transgenic systems have revealed many discrete, transient sites of RA action during development. However, there has been no attempt to permanently label the RA-activated cell lineages during mouse ontogenesis. We describe the characterization of a RA-activatable Cre transgene, which through crosses with a conditional reporter strain (the ROSA26R lacZ reporter), leads to a stable labeling of the cell populations experiencing RA signaling during embryogenesis. RA response-element (RARE)-driven Cre activity mimics at early stages the known activity of the corresponding RARE-lacZ transgene (Rossant et al.,1991). Stable labeling of the Cre-excised cell populations allows to trace the distribution of the RA-activated cell lineages at later stages. These are described in relationship with current models of RA activity in various developmental systems, including the embryonic caudal region, limb buds, hindbrain, sensory organs, and heart.


Asunto(s)
Linaje de la Célula/fisiología , Embrión de Mamíferos/citología , Tretinoina/metabolismo , Animales , Embrión de Mamíferos/metabolismo , Corazón/embriología , Esbozos de los Miembros/embriología , Ratones , Prosencéfalo/embriología , Rombencéfalo/embriología
17.
Am J Med Genet A ; 152A(12): 2947-61, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21108385

RESUMEN

Retinoic acid (RA) is a biologically active derivative of vitamin A that is indispensable for inner ear development. The normal function of RA is achieved only at optimal homeostatic concentrations, with an excess or deficiency in RA leading to inner ear dysmorphogenesis. We present an overview of the role of RA in the developing mammalian inner ear, discussing both how and when RA may act to critically control a program of inner ear development. Molecular mechanisms of otic teratogenicity involving two members of the fibroblast growth factor family, FGF3 and FGF10, and their downstream targets, Dlx5 and Dlx6, are examined under conditions of both RA excess and deficiency. We term the effect of too little or too much RA on FGF/Dlx signaling a Goldilocks phenomenon. We demonstrate that in each case (RA excess, RA deficiency), RA can directly affect FGF3/FGF10 signaling within the otic epithelium, leading to downregulated expression of these essential signaling molecules, which in turn, leads to diminution in Dlx5/Dlx6 expression. Non-cell autonomous affects of the otic epithelium subsequently occur, altering transforming growth factor-beta (TGFß) expression in the neighboring periotic mesenchyme and serving as a putative explanation for RA-mediated otic capsule defects. We conclude that RA coordinates inner ear morphogenesis by controlling an FGF/Dlx signaling cascade, whose perturbation by deviations in local retinoid concentrations can lead to inner ear dysmorphogenesis.


Asunto(s)
Oído Interno/embriología , Organogénesis/genética , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Tretinoina/metabolismo , Animales , Comunicación Celular/genética , Oído Interno/anomalías , Embrión de Mamíferos , Epitelio/metabolismo , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/deficiencia , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Mesodermo/metabolismo , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/farmacología , Tretinoina/farmacología
18.
Med Sci (Paris) ; 36(3): 225-230, 2020 Mar.
Artículo en Francés | MEDLINE | ID: mdl-32228840

RESUMEN

The oral cavity is one of the main route for environmental contaminations associated to many chronic diseases (cancers, fertility and behavior disorders for example) via alimentation, medications and respiration. These environmental factors including, among others, endocrine disruptors and excessive fluoride can disrupt dental development and thus generate irreversible enamel defects. These defects are then treated with materials that may release molecules capable of generating these defects, leading to a vicious circle, particularly in pregnant women and young children. The present paper aims to review the state of knowledge, questions and controversies on common environmental factors in contact with the oral cavity. It also reviews their mechanisms of action and the mediators involved in enamel pathologies associated with environmental conditions. Dental tissues can not only be targeted by environmental factors but can also serve as early and easily accessible markers of exposure to these agents. Understanding and characterizing the environmental impact in the oral cavity will help to prevent multiple diseases, oral and distant, whose link with oral homeostasis is just being explored.


TITLE: La sphère orale, cible et marqueur de l'exposition environnementale - I. Défauts du développement dentaire. ABSTRACT: La cavité buccale est l'une des voies majeures des contaminations environnementales connues pour être impliquées dans de nombreuses pathologies chroniques (cancers, troubles de la fertilité et du comportement) via l'alimentation, les médications ou même la respiration. Ces facteurs environnementaux incluant, entre autres, des perturbateurs endocriniens et le fluor en excès, peuvent perturber le développement dentaire et ainsi générer des défauts irréversibles de l'émail. Ces défauts sont alors traités avec des matériaux dont certains libèrent des molécules capables à leur tour de générer ces défauts, conduisant à un cercle vicieux, notamment chez la femme enceinte et le jeune enfant. Cette synthèse fait le point sur l'état des connaissances, les questions et controverses sur les facteurs environnementaux courants susceptibles d'entrer en contact avec la sphère orale, leurs mécanismes d'actions et les médiateurs impliqués dans les pathologies de l'émail associées aux conditions environnementales.


Asunto(s)
Biomarcadores/análisis , Enfermedades del Desarrollo Óseo/inducido químicamente , Exposición a Riesgos Ambientales/análisis , Boca/fisiología , Enfermedades Estomatognáticas/inducido químicamente , Administración Oral , Enfermedades del Desarrollo Óseo/epidemiología , Niño , Preescolar , Hipoplasia del Esmalte Dental/inducido químicamente , Hipoplasia del Esmalte Dental/epidemiología , Dieta , Vías de Administración de Medicamentos , Disruptores Endocrinos/toxicidad , Contaminantes Ambientales/toxicidad , Femenino , Fluoruros/efectos adversos , Humanos , Boca/efectos de los fármacos , Boca/patología , Embarazo , Enfermedades Estomatognáticas/epidemiología
19.
Sci Rep ; 10(1): 14817, 2020 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-32908163

RESUMEN

Secreted extracellular matrix components which regulate craniofacial development could be reactivated and play roles in adult wound healing. We report a patient with a loss-of-function of the secreted matricellular protein SMOC2 (SPARC related modular calcium binding 2) presenting severe oligodontia, microdontia, tooth root deficiencies, alveolar bone hypoplasia, and a range of skeletal malformations. Turning to a mouse model, Smoc2-GFP reporter expression indicates SMOC2 dynamically marks a range of dental and bone progenitors. While germline Smoc2 homozygous mutants are viable, tooth number anomalies, reduced tooth size, altered enamel prism patterning, and spontaneous age-induced periodontal bone and root loss are observed in this mouse model. Whole-genome RNA-sequencing analysis of embryonic day (E) 14.5 cap stage molars revealed reductions in early expressed enamel matrix components (Odontogenic ameloblast-associated protein) and dentin dysplasia targets (Dentin matrix acidic phosphoprotein 1). We tested if like other matricellular proteins SMOC2 was required for regenerative repair. We found that the Smoc2-GFP reporter was reactivated in adjacent periodontal tissues 4 days after tooth avulsion injury. Following maxillary tooth injury, Smoc2-/- mutants had increased osteoclast activity and bone resorption surrounding the extracted molar. Interestingly, a 10-day treatment with the cyclooxygenase 2 (COX2) inhibitor ibuprofen (30 mg/kg body weight) blocked tooth injury-induced bone loss in Smoc2-/- mutants, reducing matrix metalloprotease (Mmp)9. Collectively, our results indicate that endogenous SMOC2 blocks injury-induced jaw bone osteonecrosis and offsets age-induced periodontal decay.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Animales , Resorción Ósea/genética , Resorción Ósea/metabolismo , Proteínas de Unión al Calcio/genética , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Microscopía Electrónica de Rastreo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Diente/metabolismo
20.
Nat Commun ; 10(1): 4137, 2019 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-31515492

RESUMEN

Developmental cell death plays an important role in the construction of functional neural circuits. In vertebrates, the canonical view proposes a selection of the surviving neurons through stochastic competition for target-derived neurotrophic signals, implying an equal potential for neurons to compete. Here we show an alternative cell fitness selection of neurons that is defined by a specific neuronal heterogeneity code. Proprioceptive sensory neurons that will undergo cell death and those that will survive exhibit different molecular signatures that are regulated by retinoic acid and transcription factors, and are independent of the target and neurotrophins. These molecular features are genetically encoded, representing two distinct subgroups of neurons with contrasted functional maturation states and survival outcome. Thus, in this model, a heterogeneous code of intrinsic cell fitness in neighboring neurons provides differential competitive advantage resulting in the selection of cells with higher capacity to survive and functionally integrate into neural networks.


Asunto(s)
Modelos Biológicos , Células Receptoras Sensoriales/citología , Animales , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Embrión de Pollo , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Ratones Endogámicos C57BL , Propiocepción/efectos de los fármacos , Receptor trkC/metabolismo , Células Receptoras Sensoriales/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Tretinoina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA