Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 87
Filtrer
1.
Prog Neurobiol ; 212: 102246, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-35151792

RÉSUMÉ

Retinoic acid is a powerful regulator of brain development, however its postnatal functions only start to be elucidated. We show that retinoic acid receptor beta (RARß), is involved in neuroprotection of striatopallidal medium spiny neurons (spMSNs), the cell type affected in different neuropsychiatric disorders and particularly prone to degenerate in Huntington disease (HD). Accordingly, the number of spMSNs was reduced in the striatum of adult Rarß-/- mice, which may result from mitochondrial dysfunction and neurodegeneration. Mitochondria morphology was abnormal in mutant mice whereas in cultured striatal Rarß-/- neurons mitochondria displayed exacerbated depolarization, and fragmentation followed by cell death in response to glutamate or thapsigargin-induced calcium increase. In vivo, Rarß-/- spMSNs were also more vulnerable to the mitochondrial toxin 3-nitropropionic acid (3NP), known to induce HD symptoms in human and rodents. In contrary, an RARß agonist, AC261066, decreased glutamate-induced toxicity in primary striatal neurons in vitro, and diminished mitochondrial dysfunction, spMSN cell death and motor deficits induced in wild type mice by 3NP. We demonstrate that the striatopallidal pathway is compromised in Rarß-/- mice and associated with HD-like motor abnormalities. Importantly, similar motor abnormalities and selective reduction of spMSNs were induced by striatal or spMSN-specific inactivation of RARß, further supporting a neuroprotective role of RARß in postnatal striatum.


Sujet(s)
Maladie de Huntington , Neurones , Animaux , Acide glutamique/métabolisme , Souris , Mitochondries/métabolisme , Neurones/métabolisme , Récepteurs à l'acide rétinoïque
2.
Elife ; 102021 10 08.
Article de Anglais | MEDLINE | ID: mdl-34623260

RÉSUMÉ

Retinoic acid (RA) is an essential signaling molecule for cardiac development and plays a protective role in the heart after myocardial infarction (MI). In both cases, the effect of RA signaling on cardiomyocytes, the principle cell type of the heart, has been reported to be indirect. Here we have developed an inducible murine transgenic RA-reporter line using CreERT2 technology that permits lineage tracing of RA-responsive cells and faithfully recapitulates endogenous RA activity in multiple organs during embryonic development. Strikingly, we have observed a direct RA response in cardiomyocytes during mid-late gestation and after MI. Ablation of RA signaling through deletion of the Aldh1a1/a2/a3 genes encoding RA-synthesizing enzymes leads to increased cardiomyocyte apoptosis in adults subjected to MI. RNA sequencing analysis reveals Tgm2 and Ace1, two genes with well-established links to cardiac repair, as potential targets of RA signaling in primary cardiomyocytes, thereby providing novel links between the RA pathway and heart disease.


Sujet(s)
Infarctus du myocarde/complications , Myocytes cardiaques/anatomopathologie , Trétinoïne/métabolisme , Aldehyde dehydrogenase/génétique , Aldehyde dehydrogenase/métabolisme , Animaux , Apoptose , Développement embryonnaire , Femelle , Mâle , Souris , Souris transgéniques , Transduction du signal
3.
PLoS Biol ; 18(11): e3000902, 2020 11.
Article de Anglais | MEDLINE | ID: mdl-33201874

RÉSUMÉ

Coordinated development of muscles, tendons, and their attachment sites ensures emergence of functional musculoskeletal units that are adapted to diverse anatomical demands among different species. How these different tissues are patterned and functionally assembled during embryogenesis is poorly understood. Here, we investigated the morphogenesis of extraocular muscles (EOMs), an evolutionary conserved cranial muscle group that is crucial for the coordinated movement of the eyeballs and for visual acuity. By means of lineage analysis, we redefined the cellular origins of periocular connective tissues interacting with the EOMs, which do not arise exclusively from neural crest mesenchyme as previously thought. Using 3D imaging approaches, we established an integrative blueprint for the EOM functional unit. By doing so, we identified a developmental time window in which individual EOMs emerge from a unique muscle anlage and establish insertions in the sclera, which sets these muscles apart from classical muscle-to-bone type of insertions. Further, we demonstrate that the eyeballs are a source of diffusible all-trans retinoic acid (ATRA) that allow their targeting by the EOMs in a temporal and dose-dependent manner. Using genetically modified mice and inhibitor treatments, we find that endogenous local variations in the concentration of retinoids contribute to the establishment of tendon condensations and attachment sites that precede the initiation of muscle patterning. Collectively, our results highlight how global and site-specific programs are deployed for the assembly of muscle functional units with precise definition of muscle shapes and topographical wiring of their tendon attachments.


Sujet(s)
Muscles oculomoteurs/embryologie , Muscles oculomoteurs/croissance et développement , Trétinoïne/métabolisme , Animaux , Tissu conjonctif/physiologie , Développement embryonnaire , Oeil , Imagerie tridimensionnelle/méthodes , Souris/embryologie , Souris de lignée C57BL , Souris de lignée DBA , Morphogenèse , Transduction du signal , Tendons/physiologie , Trétinoïne/physiologie
4.
Sci Rep ; 10(1): 14817, 2020 09 09.
Article de Anglais | MEDLINE | ID: mdl-32908163

RÉSUMÉ

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.


Sujet(s)
Protéines de liaison au calcium/métabolisme , Animaux , Résorption osseuse/génétique , Résorption osseuse/métabolisme , Protéines de liaison au calcium/génétique , Cyclooxygenase 2/génétique , Cyclooxygenase 2/métabolisme , Régulation de l'expression des gènes au cours du développement , Humains , Mâle , Matrix metalloproteinase 9/génétique , Matrix metalloproteinase 9/métabolisme , Souris , Microscopie électronique à balayage , ARN messager/génétique , ARN messager/métabolisme , Réaction de polymérisation en chaine en temps réel , Dent/métabolisme
5.
Sci Rep ; 7(1): 13671, 2017 10 20.
Article de Anglais | MEDLINE | ID: mdl-29057906

RÉSUMÉ

Embryonal carcinoma (EC) cells are pluripotent stem cells extensively used for studies of cell differentiation. Although retinoic acid (RA) is a powerful inducer of neurogenesis in EC cells, it is not clear what specific neuronal subtypes are generated and whether different RAR isotypes may contribute to such neuronal diversification. Here we show that RA treatment during EC embryoid body formation is a highly robust protocol for generation of striatal-like GABAergic neurons which display molecular characteristics of striatopallidal medium spiny neurons (MSNs), including expression of functional dopamine D2 receptor. By using RARα, ß and γ selective agonists we show that RARγ is the functionally dominant RAR in mediating RA control of early molecular determinants of MSNs leading to formation of striatopallidal-like neurons. In contrast, activation of RARα is less efficient in generation of this class of neurons, but is essential for differentiation of functional dopaminergic neurons, which may correspond to a subpopulation of inhibitory dopaminergic neurons expressing glutamic acid decarboxylase in vivo.


Sujet(s)
Cellules souches de carcinome embryonnaire/métabolisme , Neurogenèse/physiologie , Neurones/métabolisme , Récepteurs à l'acide rétinoïque/métabolisme , Animaux , Encéphale/métabolisme , Lignée cellulaire tumorale , Dopamine/métabolisme , Souris , Récepteur D2 de la dopamine/métabolisme , Acide gamma-amino-butyrique/métabolisme
6.
Dev Biol ; 430(1): 129-141, 2017 10 01.
Article de Anglais | MEDLINE | ID: mdl-28790015

RÉSUMÉ

A tight regulation of neuron production is required to generate a functional cerebral cortex and is achieved by a proper balance between proliferation and differentiation of progenitor cells. Though the vitamin A (retinol) active derivative retinoic acid (RA) has been implicated as one of the signals acting during mammalian forebrain neurogenesis, its function at the onset of neurogenesis as well as during establishment of cortical layers and neuronal subtypes remains elusive. One limitation is that murine mutants for genes encoding key enzymes involved in RA synthesis die during early embryonic development. We analysed corticogenesis in Rdh10 null mutants, in which an RA deficiency is generated as the intracellular retinol to retinaldehyde conversion is abolished. When analysed at the latest stage before lethality occurs (embryonic day [E]13.5), the mutants show smaller telencephalic vesicles and the thickness of their cortical plate is strongly reduced. The first progenitors formed in the cortical plate are radial glial (RG) cells which generate neurons either directly, or through an indirect mechanism involving the production of intermediate neuronal progenitors (INPs) which then give rise to neurons. We show that in absence of RA, the RG progenitors proliferate less and prematurely produce neurons, leading to their depletion at E11.5. Furthermore, we could demonstrate that lack of RA impairs the generation of INPs at E13.5 and affects the cell cycle exit of progenitor cells during corticogenesis, altogether leading to a deficit in projection neurons and to microcephaly.


Sujet(s)
Cortex cérébral/embryologie , Neurogenèse/effets des médicaments et des substances chimiques , Trétinoïne/pharmacologie , Alcohol oxidoreductases/métabolisme , Animaux , Cycle cellulaire/effets des médicaments et des substances chimiques , Cortex cérébral/effets des médicaments et des substances chimiques , Cycline D2/métabolisme , Cellules épendymogliales/effets des médicaments et des substances chimiques , Cellules épendymogliales/métabolisme , Délétion de gène , Souris knockout , Microcéphalie/anatomopathologie , Modèles biologiques , Cellules souches/effets des médicaments et des substances chimiques , Cellules souches/métabolisme
7.
Eur J Oral Sci ; 125(1): 8-17, 2017 02.
Article de Anglais | MEDLINE | ID: mdl-28084688

RÉSUMÉ

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.


Sujet(s)
Amélogenèse/génétique , Émail dentaire/malformations , Protéines de liaison au TGF-bêta latent/génétique , Améloblastes/métabolisme , Amélogenèse imparfaite/génétique , Animaux , Émail dentaire/ultrastructure , Génotype , Mâle , Souris , Souches mutantes de souris , Microscopie électronique à balayage , Mutation , Ostéochondrodysplasies/génétique , Phénotype , Calcification dentaire/génétique , Facteur de croissance transformant bêta/génétique , Microtomographie aux rayons X
8.
Mol Neurobiol ; 54(5): 3859-3878, 2017 07.
Article de Anglais | MEDLINE | ID: mdl-27405468

RÉSUMÉ

Retinoic acid (RA) signaling through retinoic acid receptors (RARs), known for its multiple developmental functions, emerged more recently as an important regulator of adult brain physiology. How RAR-mediated regulation is achieved is poorly known, partly due to the paucity of information on critical target genes in the brain. Also, it is not clear how reduced RA signaling may contribute to pathophysiology of diverse neuropsychiatric disorders. We report the first genome-wide analysis of RAR transcriptional targets in the brain. Using chromatin immunoprecipitation followed by high-throughput sequencing and transcriptomic analysis of RARß-null mutant mice, we identified genomic targets of RARß in the striatum. Characterization of RARß transcriptional targets in the mouse striatum points to mechanisms through which RAR may control brain functions and display neuroprotective activity. Namely, our data indicate with statistical significance (FDR 0.1) a strong contribution of RARß in controlling neurotransmission, energy metabolism, and transcription, with a particular involvement of G-protein coupled receptor (p = 5.0e-5), cAMP (p = 4.5e-4), and calcium signaling (p = 3.4e-3). Many identified RARß target genes related to these pathways have been implicated in Alzheimer's, Parkinson's, and Huntington's disease (HD), raising the possibility that compromised RA signaling in the striatum may be a mechanistic link explaining the similar affective and cognitive symptoms in these diseases. The RARß transcriptional targets were particularly enriched for transcripts affected in HD. Using the R6/2 transgenic mouse model of HD, we show that partial sequestration of RARß in huntingtin protein aggregates may account for reduced RA signaling reported in HD.


Sujet(s)
Étude d'association pangénomique , Maladie de Huntington/génétique , Maladies neurodégénératives/génétique , Récepteurs à l'acide rétinoïque/métabolisme , Transduction du signal , Transcription génétique , Trétinoïne/métabolisme , Animaux , Sites de fixation , ADN/métabolisme , Protéine huntingtine/métabolisme , Maladie de Huntington/anatomopathologie , Souris de lignée C57BL , Souris knockout , Néostriatum/métabolisme , Néostriatum/anatomopathologie , Maladies neurodégénératives/anatomopathologie , Agrégats de protéines , Liaison aux protéines , ARN messager/génétique , ARN messager/métabolisme , Reproductibilité des résultats , Éléments de réponse/génétique
9.
Biol Open ; 6(2): 148-160, 2017 Feb 15.
Article de Anglais | MEDLINE | ID: mdl-28011626

RÉSUMÉ

Retinoic acid (RA) is a diffusible molecule involved in early forebrain patterning. Its later production in the meninges by the retinaldehyde dehydrogenase RALDH2 coincides with the time of cortical neuron generation. A function of RA in this process has not been adressed directly as Raldh2-/- mouse mutants are embryonic lethal. Here, we used a conditional genetic strategy to inactivate Raldh2 just prior to onset of its expression in the developing meninges. This inactivation does not affect the formation of the cortical progenitor populations, their rate of division, or timing of differentiation. However, migration of late-born cortical neurons is delayed, with neurons stalling in the intermediate zone and exhibiting an abnormal multipolar morphology. This suggests that RA controls the multipolar-to-bipolar transition that occurs in the intermediate zone and allows neurons to start locomotion in the cortical plate. Our work also shows a role for RA in cortical lamination, as deep layers are expanded and a subset of layer IV neurons are not formed in the Raldh2-ablated mutants. These data demonstrate that meninges are a source of extrinsic signals important for cortical development.

10.
Exp Eye Res ; 154: 190-195, 2017 01.
Article de Anglais | MEDLINE | ID: mdl-27840061

RÉSUMÉ

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.


Sujet(s)
Apoptose , Épithélium antérieur de la cornée/métabolisme , Régénération/physiologie , Trétinoïne/métabolisme , Animaux , Prolifération cellulaire , Épithélium antérieur de la cornée/anatomopathologie , Souris , Transduction du signal
11.
Sci Rep ; 6: 25541, 2016 05 06.
Article de Anglais | MEDLINE | ID: mdl-27150455

RÉSUMÉ

The epicardium plays an essential role in coronary artery formation and myocardial development. However, signals controlling the developing epicardium and epicardial-mesenchymal transition (EMT) in the normal and diseased adult heart are studied less rigorously. Here we investigated the role of angiogenic hormone, prokineticin-2 and its receptor PKR1 in the epicardium of developing and adult heart. Genetic ablation of PKR1 in epicardium leads to partial embryonic and postnatal lethality with abnormal heart development. Cardiac developmental defects are manifested in the adult stage as ischemic cardiomyopathy with systolic dysfunction. We discovered that PKR1 regulates epicardial-mesenchymal transition (EMT) for epicardial-derived progenitor cell (EPDC), formation. This event affects at least three consequential steps during heart development: (i) EPDC and cardiomyocyte proliferation involved in thickening of an outer compact ventricular chamber wall, (ii) rhythmicity, (iii) formation of coronary circulation. In isolated embryonic EPDCs, overexpression or activation of PKR1 alters cell morphology and EMT markers via activating Akt signaling. Lack of PKR1 signal in epicardium leads to defective heart development and underlies the origin of congenital heart disease in adult mice. Our mice provide genetic models for congenital dysfunction of the heart and should facilitate studies of both pathogenesis and therapy of cardiac disorders in humans.


Sujet(s)
Transition épithélio-mésenchymateuse , Hormones gastrointestinales/métabolisme , Coeur/embryologie , Neuropeptides/métabolisme , Péricarde/embryologie , Récepteurs couplés aux protéines G/métabolisme , Transduction du signal , Animaux , Souris , Récepteurs couplés aux protéines G/génétique
12.
FASEB J ; 30(8): 2733-40, 2016 08.
Article de Anglais | MEDLINE | ID: mdl-27084889

RÉSUMÉ

Identification of factors regulating renal development is important to understand the pathogenesis of congenital kidney diseases. Little is known about the molecular mechanism of renal development and functions triggered by the angiogenic hormone prokineticin-2 and its receptor, PKR1. Utilizing the Gata5 (G5)-Cre and Wilms tumor 1 (Wt1)(GFP)cre transgenic lines, we generated mutant mice with targeted PKR1 gene disruptions in nephron progenitors. These mutant mice exhibited partial embryonic and postnatal lethality. Kidney developmental defects in PKR(G5-/-) mice are manifested in the adult stage as renal atrophy with glomerular defects, nephropathy, and uremia. PKR1(Wt1-/-) embryos exhibit hypoplastic kidneys with premature glomeruli and necrotic nephrons as a result of impaired proliferation and increased apoptosis in Wt1(+) renal mesenchymal cells. PKR1 regulates renal mesenchymal-epithelial transition (MET) that is involved in formation of renal progenitors, regulating glomerulogenesis toward forming nephrons during kidney development. In the isolated embryonic Wt1(+) renal cells, overexpression or activation of PKR1 promotes MET defined by the transition from elongated cell to octagonal cell morphology, and alteration of the expression of MET markers via activating NFATc3 signaling. Together, these results establish PKR1 via NFATc3 as a crucial modifier of MET processing to the development of nephron. Our study should facilitate new therapeutic opportunities in human renal disorders.-Arora, H., Boulberdaa, M., Qureshi, R., Bitirim, V., Messadeq, N., Dolle, P., Nebigil, C. G. Prokineticin receptor 1 is required for mesenchymal-epithelial transition in kidney development.


Sujet(s)
Transition épithélio-mésenchymateuse/physiologie , Régulation de l'expression des gènes au cours du développement/physiologie , Récepteurs couplés aux protéines G/métabolisme , Animaux , Apoptose , Prolifération cellulaire , Embryon de mammifère/métabolisme , Développement embryonnaire , Transition épithélio-mésenchymateuse/génétique , Souris , Souris knockout , Mutation , Néovascularisation physiologique , Récepteurs couplés aux protéines G/génétique
13.
Nat Commun ; 7: 11063, 2016 Mar 30.
Article de Anglais | MEDLINE | ID: mdl-27026076

RÉSUMÉ

TAF4 (TATA-binding protein-associated factor 4) and its paralogue TAF4b are components of the TFIID core module. We inactivated the murine Taf4a gene to address Taf4 function during embryogenesis. Here we show that Taf4a(-/-) embryos survive until E9.5 where primary germ layers and many embryonic structures are identified showing Taf4 is dispensable for their specification. In contrast, Taf4 is required for correct patterning of the trunk and anterior structures, ventral morphogenesis and proper heart positioning. Overlapping expression of Taf4a and Taf4b during embryogenesis suggests their redundancy at early stages. In agreement with this, Taf4a(-/-) embryonic stem cells (ESCs) are viable and comprise Taf4b-containing TFIID. Nevertheless, Taf4a(-/-) ESCs do not complete differentiation into glutamatergic neurons and cardiomyocytes in vitro due to impaired preinitiation complex formation at the promoters of critical differentiation genes. We define an essential role of a core TFIID TAF in differentiation events during mammalian embryogenesis.


Sujet(s)
Différenciation cellulaire , Développement embryonnaire , Cellules souches embryonnaires de souris/métabolisme , Sous-unités de protéines/métabolisme , Facteur de transcription TFIID/métabolisme , Animaux , Marqueurs biologiques/métabolisme , Plan d'organisation du corps/effets des médicaments et des substances chimiques , Plan d'organisation du corps/génétique , Malformations cardiovasculaires/embryologie , Malformations cardiovasculaires/génétique , Malformations cardiovasculaires/anatomopathologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/génétique , Survie cellulaire/effets des médicaments et des substances chimiques , Perte de l'embryon/génétique , Perte de l'embryon/anatomopathologie , Développement embryonnaire/effets des médicaments et des substances chimiques , Développement embryonnaire/génétique , Femelle , Régulation de l'expression des gènes au cours du développement/effets des médicaments et des substances chimiques , Cellules germinales/effets des médicaments et des substances chimiques , Cellules germinales/métabolisme , Souris , Souris de lignée C57BL , Cellules souches embryonnaires de souris/effets des médicaments et des substances chimiques , Mutation , Contraction myocardique/effets des médicaments et des substances chimiques , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Crête neurale/effets des médicaments et des substances chimiques , Crête neurale/anatomopathologie , Neurogenèse/effets des médicaments et des substances chimiques , Neurogenèse/génétique , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Phénotype , Grossesse , Sous-unités de protéines/génétique , Facteur de transcription TFIID/déficit , Facteur de transcription TFIID/génétique , Trétinoïne/pharmacologie
14.
Front Physiol ; 7: 673, 2016.
Article de Anglais | MEDLINE | ID: mdl-28111553

RÉSUMÉ

Abnormalities of enamel matrix proteins deposition, mineralization, or degradation during tooth development are responsible for a spectrum of either genetic diseases termed Amelogenesis imperfecta or acquired enamel defects. To assess if environmental/nutritional factors can exacerbate enamel defects, we investigated the role of the active form of vitamin A, retinoic acid (RA). Robust expression of RA-degrading enzymes Cyp26b1 and Cyp26c1 in developing murine teeth suggested RA excess would reduce tooth hard tissue mineralization, adversely affecting enamel. We employed a protocol where RA was supplied to pregnant mice as a food supplement, at a concentration estimated to result in moderate elevations in serum RA levels. This supplementation led to severe enamel defects in adult mice born from pregnant dams, with most severe alterations observed for treatments from embryonic day (E)12.5 to E16.5. We identified the enamel matrix proteins enamelin (Enam), ameloblastin (Ambn), and odontogenic ameloblast-associated protein (Odam) as target genes affected by excess RA, exhibiting mRNA reductions of over 20-fold in lower incisors at E16.5. RA treatments also affected bone formation, reducing mineralization. Accordingly, craniofacial ossification was drastically reduced after 2 days of treatment (E14.5). Massive RNA-sequencing (RNA-seq) was performed on E14.5 and E16.5 lower incisors. Reductions in Runx2 (a key transcriptional regulator of bone and enamel differentiation) and its targets were observed at E14.5 in RA-exposed embryos. RNA-seq analysis further indicated that bone growth factors, extracellular matrix, and calcium homeostasis were perturbed. Genes mutated in human AI (ENAM, AMBN, AMELX, AMTN, KLK4) were reduced in expression at E16.5. Our observations support a model in which elevated RA signaling at fetal stages affects dental cell lineages. Thereafter enamel protein production is impaired, leading to permanent enamel alterations.

15.
J Neurosci ; 35(43): 14467-75, 2015 Oct 28.
Article de Anglais | MEDLINE | ID: mdl-26511239

RÉSUMÉ

The mammalian striatum controls sensorimotor and psychoaffective functions through coordinated activities of its two striatonigral and striatopallidal output pathways. Here we show that retinoic acid receptor ß (RARß) controls development of a subpopulation of GABAergic, Gad65-positive striatonigral projection neurons. In Rarb(-/-) knock-out mice, concomitant reduction of Gad65, dopamine receptor D1 (Drd1), and substance P expression at different phases of prenatal development was associated with reduced number of Drd1-positive cells at birth, in contrast to normal numbers of striatopallidal projection neurons expressing dopamine receptor D2. Fate mapping using BrdU pulse-chase experiments revealed that such deficits may originate from compromised proliferation of late-born striosomal neurons and lead to decreased number of Drd1-positive cells retaining BrdU in postnatal day (P) 0 Rarb(-/-) striatum. Reduced expression of Fgf3 in the subventricular zone of the lateral ganglionic eminence (LGE) at embryonic day 13.5 may underlie such deficits by inducing premature differentiation of neuronal progenitors, as illustrated by reduced expression of the proneural gene Ascl1 (Mash1) and increased expression of Meis1, a marker of postmitotic LGE neurons. In agreement with a critical role of FGF3 in this control, reduced number of Ascl1-expressing neural progenitors, and a concomitant increase of Meis1-expressing cells, were observed in primary cell cultures of Rarb(-/-) LGE. This defect was normalized by addition of fibroblast growth factor (FGF). Such data point to role of Meis1 in striatal development, also supported by reduced neuronal differentiation in the LGE of Meis1(-/-) embryos. Our data unveil a novel mechanism of development of striatonigral projection neurons involving retinoic acid and FGF, two signals required for positioning the boundaries of Meis1-expressing cells.


Sujet(s)
Corps strié/physiologie , Facteurs de croissance fibroblastique/physiologie , Protéines à homéodomaine/physiologie , Protéines tumorales/physiologie , Neurones/physiologie , Récepteurs à l'acide rétinoïque/physiologie , Substantia nigra/physiologie , Animaux , Antimétabolites/pharmacologie , Broxuridine/pharmacologie , Corps strié/cytologie , Corps strié/embryologie , Femelle , Facteur de croissance fibroblastique de type 3/métabolisme , Glutamate decarboxylase/métabolisme , Souris , Souris de lignée C57BL , Souris knockout , Protéine du site-1 d'intégration des virus myéloïdes écotropiques , Neurogenèse/génétique , Neurogenèse/physiologie , Grossesse , Culture de cellules primaires , Récepteur dopamine D1/métabolisme , Substantia nigra/cytologie , Substantia nigra/embryologie
16.
Hum Mol Genet ; 24(11): 3038-49, 2015 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-25669657

RÉSUMÉ

Inherited dental malformations constitute a clinically and genetically heterogeneous group of disorders. Here, we report on four families, three of them consanguineous, with an identical phenotype, characterized by significant short stature with brachyolmia and hypoplastic amelogenesis imperfecta (AI) with almost absent enamel. This phenotype was first described in 1996 by Verloes et al. as an autosomal recessive form of brachyolmia associated with AI. Whole-exome sequencing resulted in the identification of recessive hypomorphic mutations including deletion, nonsense and splice mutations, in the LTBP3 gene, which is involved in the TGF-beta signaling pathway. We further investigated gene expression during mouse development and tooth formation. Differentiated ameloblasts synthesizing enamel matrix proteins and odontoblasts expressed the gene. Study of an available knockout mouse model showed that the mutant mice displayed very thin to absent enamel in both incisors and molars, hereby recapitulating the AI phenotype in the human disorder.


Sujet(s)
Amélogenèse imparfaite/génétique , Protéines de liaison au TGF-bêta latent/génétique , Ostéochondrodysplasies/génétique , Adolescent , Amélogenèse imparfaite/imagerie diagnostique , Animaux , Séquence nucléotidique , Enfant , Consanguinité , Analyse de mutations d'ADN , Femelle , Mutation avec décalage du cadre de lecture , Études d'associations génétiques , Humains , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Mutation faux-sens , Ostéochondrodysplasies/imagerie diagnostique , Pedigree , Radiographie , Délétion de séquence
17.
PLoS One ; 10(2): e0118024, 2015.
Article de Anglais | MEDLINE | ID: mdl-25706271

RÉSUMÉ

An in situ hybridization (ISH) study was performed on 2000 murine genes representing around 10% of the protein-coding genes present in the mouse genome using data generated by the EURExpress consortium. This study was carried out in 25 tissues of late gestation embryos (E14.5), with a special emphasis on the developing ear and on five distinct developing sensory organs, including the cochlea, the vestibular receptors, the sensory retina, the olfactory organ, and the vibrissae follicles. The results obtained from an analysis of more than 11,000 micrographs have been integrated in a newly developed knowledgebase, called ImAnno. In addition to managing the multilevel micrograph annotations performed by human experts, ImAnno provides public access to various integrated databases and tools. Thus, it facilitates the analysis of complex ISH gene expression patterns, as well as functional annotation and interaction of gene sets. It also provides direct links to human pathways and diseases. Hierarchical clustering of expression patterns in the 25 tissues revealed three main branches corresponding to tissues with common functions and/or embryonic origins. To illustrate the integrative power of ImAnno, we explored the expression, function and disease traits of the sensory epithelia of the five presumptive sensory organs. The study identified 623 genes (out of 2000) concomitantly expressed in the five embryonic epithelia, among which many (∼12%) were involved in human disorders. Finally, various multilevel interaction networks were characterized, highlighting differential functional enrichments of directly or indirectly interacting genes. These analyses exemplify an under-represention of "sensory" functions in the sensory gene set suggests that E14.5 is a pivotal stage between the developmental stage and the functional phase that will be fully reached only after birth.


Sujet(s)
Biologie informatique/méthodes , Analyse de profil d'expression de gènes/méthodes , Régulation de l'expression des gènes au cours du développement , Hybridation in situ/méthodes , Animaux , Plexus choroïde/embryologie , Plexus choroïde/métabolisme , Bases de données génétiques , Oreille interne/embryologie , Oreille interne/métabolisme , Développement foetal/génétique , Gene Ontology , Réseaux de régulation génique , Génomique/méthodes , Humains , Mémorisation et recherche des informations/méthodes , Souris , Muqueuse olfactive/embryologie , Muqueuse olfactive/métabolisme , Rétine/embryologie , Rétine/métabolisme , Cellules réceptrices sensorielles/métabolisme , Vibrisses/embryologie , Vibrisses/métabolisme
18.
Front Physiol ; 5: 345, 2014.
Article de Anglais | MEDLINE | ID: mdl-25309446

RÉSUMÉ

Precise control of self-renewal and differentiation of progenitor cells into the cranial neural crest (CNC) pool ensures proper head development, guided by signaling pathways such as BMPs, FGFs, Shh and Notch. Here, we show that murine Sox2 plays an essential role in controlling progenitor cell behavior during craniofacial development. A "Conditional by Inversion" Sox2 allele (Sox2(COIN) ) has been employed to generate an epiblast ablation of Sox2 function (Sox2(EpINV) ). Sox2 (EpINV/+(H)) haploinsufficient and conditional (Sox2(EpINV/mosaic) ) mutant embryos proceed beyond gastrulation and die around E11. These mutant embryos exhibit severe anterior malformations, with hydrocephaly and frontonasal truncations, which could be attributed to the deregulation of CNC progenitor cells during their epithelial to mesenchymal transition. This irregularity results in an exacerbated and aberrant migration of Sox10(+) NCC in the branchial arches and frontonasal process of the Sox2 mutant embryos. These results suggest a novel role for Sox2 as a regulator of the epithelial to mesenchymal transitions (EMT) that are important for the cell flow in the developing head.

19.
PLoS One ; 9(1): e84343, 2014.
Article de Anglais | MEDLINE | ID: mdl-24416220

RÉSUMÉ

BACKGROUND: The RSK2 gene is responsible for Coffin-Lowry syndrome, an X-linked dominant genetic disorder causing mental retardation, skeletal growth delays, with craniofacial and digital abnormalities typically associated with this syndrome. Craniofacial and dental anomalies encountered in this rare disease have been poorly characterized. METHODOLOGY/PRINCIPAL FINDINGS: We examined, using X-Ray microtomographic analysis, the variable craniofacial dysmorphism and dental anomalies present in Rsk2 knockout mice, a model of Coffin-Lowry syndrome, as well as in triple Rsk1,2,3 knockout mutants. We report Rsk mutation produces surpernumerary teeth midline/mesial to the first molar. This highly penetrant phenotype recapitulates more ancestral tooth structures lost with evolution. Most likely this leads to a reduction of the maxillary diastema. Abnormalities of molar shape were generally restricted to the mesial part of both upper and lower first molars (M1). Expression analysis of the four Rsk genes (Rsk1, 2, 3 and 4) was performed at various stages of odontogenesis in wild-type (WT) mice. Rsk2 is expressed in the mesenchymal, neural crest-derived compartment, correlating with proliferative areas of the developing teeth. This is consistent with RSK2 functioning in cell cycle control and growth regulation, functions potentially responsible for severe dental phenotypes. To uncover molecular pathways involved in the etiology of these defects, we performed a comparative transcriptomic (DNA microarray) analysis of mandibular wild-type versus Rsk2-/Y molars. We further demonstrated a misregulation of several critical genes, using a Rsk2 shRNA knock-down strategy in molar tooth germs cultured in vitro. CONCLUSIONS: This study reveals RSK2 regulates craniofacial development including tooth development and patterning via novel transcriptional targets.


Sujet(s)
Malformations crâniofaciales/enzymologie , Tête/croissance et développement , Ribosomal Protein S6 Kinases, 90-kDa/métabolisme , Malformations multiples/enzymologie , Malformations multiples/anatomopathologie , Malformations multiples/physiopathologie , Animaux , Malformations crâniofaciales/anatomopathologie , Malformations crâniofaciales/physiopathologie , Activation enzymatique , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes au cours du développement , Techniques de knock-down de gènes , Système de signalisation des MAP kinases , Mâle , Souris , Odontogenèse , Phénotype , Petit ARN interférent/génétique , Ribosomal Protein S6 Kinases, 90-kDa/déficit , Ribosomal Protein S6 Kinases, 90-kDa/génétique , Dent/anatomie et histologie , Dent/croissance et développement
20.
PeerJ ; 1: e142, 2013.
Article de Anglais | MEDLINE | ID: mdl-24010020

RÉSUMÉ

Dorsal horn neurons in the spinal cord integrate and relay sensory information to higher brain centers. These neurons are organized in specific laminae and different transcription factors are involved in their specification. The murine homeodomain Gbx1 protein is expressed in the mantle zone of the spinal cord at E12.5-13.5, correlating with the appearance of a discernable dorsal horn around E14 and eventually defining a narrow layer in the dorsal horn around perinatal stages. At postnatal stages, Gbx1 identifies a specific subpopulation of GABAergic neurons in the dorsal spinal cord. We have generated a loss of function mutation for Gbx1 and analyzed its consequences during spinal cord development. Gbx1 (-/-) mice are viable and can reproduce as homozygous null mutants. However, the adult mutant mice display an altered gait during forward movement that specifically affects the hindlimbs. This abnormal gait was evaluated by a series of behavioral tests, indicating that locomotion is impaired, but not muscle strength or motor coordination. Molecular analysis showed that the development of the dorsal horn is not profoundly affected in Gbx1 (-/-) mutant mice. However, analysis of terminal neuronal differentiation revealed that the proportion of GABAergic inhibitory interneurons in the superficial dorsal horn is diminished. Our study unveiled a role for Gbx1 in specifying a subset of GABAergic neurons in the dorsal horn of the spinal cord involved in the control of posterior limb movement.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...