Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Nucleic Acids Res ; 48(16): 9019-9036, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32725242

ABSTRACT

Cilia assembly is under strict transcriptional control during animal development. In vertebrates, a hierarchy of transcription factors (TFs) are involved in controlling the specification, differentiation and function of multiciliated epithelia. RFX TFs play key functions in the control of ciliogenesis in animals. Whereas only one RFX factor regulates ciliogenesis in C. elegans, several distinct RFX factors have been implicated in this process in vertebrates. However, a clear understanding of the specific and redundant functions of different RFX factors in ciliated cells remains lacking. Using RNA-seq and ChIP-seq approaches we identified genes regulated directly and indirectly by RFX1, RFX2 and RFX3 in mouse ependymal cells. We show that these three TFs have both redundant and specific functions in ependymal cells. Whereas RFX1, RFX2 and RFX3 occupy many shared genomic loci, only RFX2 and RFX3 play a prominent and redundant function in the control of motile ciliogenesis in mice. Our results provide a valuable list of candidate ciliary genes. They also reveal stunning differences between compensatory processes operating in vivo and ex vivo.


Subject(s)
Cilia/physiology , Ependyma/cytology , Gene Expression Regulation, Developmental , Regulatory Factor X Transcription Factors/physiology , Regulatory Factor X1/physiology , Animals , Cilia/genetics , Mice , Mice, Inbred C57BL
2.
Hum Mol Genet ; 28(6): 877-887, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30445565

ABSTRACT

Acrocallosal syndrome (ACLS) is a rare genetic disorder characterized by agenesis or hypoplasia of corpus callosum (CC), polydactyly, craniofacial dysmorphism and severe intellectual deficiency. We previously identified KIF7, a key ciliary component of the Sonic hedgehog (SHH) pathway, as being a causative gene for this syndrome, thus including ACLS in the group of ciliopathies. In both humans and mice, KIF7 depletion leads to abnormal GLI3 processing and over-activation of SHH target genes. To understand the pathological mechanisms involved in CC defects in this syndrome, we took advantage of a previously described Kif7-/- mouse model to demonstrate that in addition to polydactyly and neural tube closure defects, these mice present CC agenesis with characteristic Probst bundles, thus recapitulating major ACLS features. We show that CC agenesis in these mice is associated with specific patterning defects of the cortical septum boundary leading to altered distribution of guidepost cells required to guide the callosal axons through the midline. Furthermore, by crossing Kif7-/- mice with Gli3Δ699 mice exclusively producing the repressive isoform of GLI3 (GLI3R), we demonstrate that decreased GLI3R signaling is fully responsible for the ACLS features in these mice, as all phenotypes are rescued by increasing GLI3R activity. Moreover, we show that increased FGF8 signaling is responsible in part for CC defects associated to KIF7 depletion, as modulating FGF8 signaling rescued CC formation anteriorly in Kif7-/- mice. Taken together our data demonstrate that ACLS features rely on defective GLI3R and FGF8 signaling.


Subject(s)
Acrocallosal Syndrome/etiology , Acrocallosal Syndrome/metabolism , Fibroblast Growth Factor 8/metabolism , Kinesins/genetics , Nerve Tissue Proteins/metabolism , Signal Transduction , Zinc Finger Protein Gli3/metabolism , Acrocallosal Syndrome/diagnosis , Animals , Body Patterning/genetics , Corpus Callosum/embryology , Corpus Callosum/metabolism , Disease Models, Animal , Disease Susceptibility , Genotype , Kinesins/metabolism , Mice , Mice, Knockout , Neuroglia/metabolism , Neurons/metabolism , Phenotype
3.
J Cell Biol ; 214(7): 875-89, 2016 09 26.
Article in English | MEDLINE | ID: mdl-27646273

ABSTRACT

The ciliary transition zone (TZ) is a complex structure found at the cilia base. Defects in TZ assembly are associated with human ciliopathies. In most eukaryotes, three protein complexes (CEP290, NPHP, and MKS) cooperate to build the TZ. We show that in Drosophila melanogaster, mild TZ defects are observed in the absence of MKS components. In contrast, Cby and Azi1 cooperate to build the TZ by acting upstream of Cep290 and MKS components. Without Cby and Azi1, centrioles fail to form the TZ, precluding sensory cilia assembly, and no ciliary membrane cap associated with sperm ciliogenesis is made. This ciliary cap is critical to recruit the tubulin-depolymerizing kinesin Klp59D, required for regulation of axonemal growth. Our results show that Drosophila TZ assembly in sensory neurons and male germ cells involves cooperative actions of Cby and Dila. They further reveal that temporal control of membrane cap assembly by TZ components and microtubule elongation by kinesin-13 is required for axoneme formation in male germ cells.


Subject(s)
Axoneme/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Spermatozoa/cytology , Spermatozoa/metabolism , Animals , Axoneme/ultrastructure , Centrioles/metabolism , Cilia/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/ultrastructure , Fertility , Male , Spermatogenesis , Spermatozoa/ultrastructure
4.
J Med Genet ; 53(2): 98-110, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26502894

ABSTRACT

BACKGROUND: Orodental diseases include several clinically and genetically heterogeneous disorders that can present in isolation or as part of a genetic syndrome. Due to the vast number of genes implicated in these disorders, establishing a molecular diagnosis can be challenging. We aimed to develop a targeted next-generation sequencing (NGS) assay to diagnose mutations and potentially identify novel genes mutated in this group of disorders. METHODS: We designed an NGS gene panel that targets 585 known and candidate genes in orodental disease. We screened a cohort of 101 unrelated patients without a molecular diagnosis referred to the Reference Centre for Oro-Dental Manifestations of Rare Diseases, Strasbourg, France, for a variety of orodental disorders including isolated and syndromic amelogenesis imperfecta (AI), isolated and syndromic selective tooth agenesis (STHAG), isolated and syndromic dentinogenesis imperfecta, isolated dentin dysplasia, otodental dysplasia and primary failure of tooth eruption. RESULTS: We discovered 21 novel pathogenic variants and identified the causative mutation in 39 unrelated patients in known genes (overall diagnostic rate: 39%). Among the largest subcohorts of patients with isolated AI (50 unrelated patients) and isolated STHAG (21 unrelated patients), we had a definitive diagnosis in 14 (27%) and 15 cases (71%), respectively. Surprisingly, COL17A1 mutations accounted for the majority of autosomal-dominant AI cases. CONCLUSIONS: We have developed a novel targeted NGS assay for the efficient molecular diagnosis of a wide variety of orodental diseases. Furthermore, our panel will contribute to better understanding the contribution of these genes to orodental disease. TRIAL REGISTRATION NUMBERS: NCT01746121 and NCT02397824.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Mutation , Tooth Abnormalities/genetics , Amelogenesis Imperfecta/genetics , Autoantigens/genetics , Chromosome Deletion , Chromosome Disorders/genetics , Chromosomes, Human, Pair 11/genetics , Cohort Studies , Coloboma/genetics , Dentin Dysplasia/genetics , France , Hearing Loss, Sensorineural/genetics , Humans , Non-Fibrillar Collagens/genetics , Reproducibility of Results , Collagen Type XVII
5.
J Neurosci ; 35(43): 14467-75, 2015 Oct 28.
Article in English | MEDLINE | ID: mdl-26511239

ABSTRACT

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.


Subject(s)
Corpus Striatum/physiology , Fibroblast Growth Factors/physiology , Homeodomain Proteins/physiology , Neoplasm Proteins/physiology , Neurons/physiology , Receptors, Retinoic Acid/physiology , Substantia Nigra/physiology , Animals , Antimetabolites/pharmacology , Bromodeoxyuridine/pharmacology , Corpus Striatum/cytology , Corpus Striatum/embryology , Female , Fibroblast Growth Factor 3/metabolism , Glutamate Decarboxylase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Ecotropic Viral Integration Site 1 Protein , Neurogenesis/genetics , Neurogenesis/physiology , Pregnancy , Primary Cell Culture , Receptors, Dopamine D1/metabolism , Substantia Nigra/cytology , Substantia Nigra/embryology
6.
PLoS Genet ; 11(7): e1005368, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26162102

ABSTRACT

Spermatogenesis consists broadly of three phases: proliferation of diploid germ cells, meiosis, and finally extensive differentiation of the haploid cells into effective delivery vehicles for the paternal genome. Despite detailed characterization of many haploid developmental steps leading to sperm, only fragmentary information exists on the control of gene expression underlying these processes. Here we report that the RFX2 transcription factor is a master regulator of genes required for the haploid phase. A targeted mutation of Rfx2 was created in mice. Rfx2-/- mice are perfectly viable but show complete male sterility. Spermatogenesis appears to progress unperturbed through meiosis. However, haploid cells undergo a complete arrest in spermatid development just prior to spermatid elongation. Arrested cells show altered Golgi apparatus organization, leading to a deficit in the generation of a spreading acrosomal cap from proacrosomal vesicles. Arrested cells ultimately merge to form giant multinucleated cells released to the epididymis. Spermatids also completely fail to form the flagellar axoneme. RNA-Seq analysis and ChIP-Seq analysis identified 139 genes directly controlled by RFX2 during spermiogenesis. Gene ontology analysis revealed that genes required for cilium function are specifically enriched in down- and upregulated genes showing that RFX2 allows precise temporal expression of ciliary genes. Several genes required for cell adhesion and cytoskeleton remodeling are also downregulated. Comparison of RFX2-regulated genes with those controlled by other major transcriptional regulators of spermiogenesis showed that each controls independent gene sets. Altogether, these observations show that RFX2 plays a major and specific function in spermiogenesis.


Subject(s)
DNA-Binding Proteins/genetics , Infertility, Male/genetics , Spermatids/cytology , Spermatocytes/cytology , Spermatogenesis/genetics , Transcription Factors/genetics , Animals , Apoptosis/genetics , Cell Adhesion/genetics , Cilia/genetics , Cilia/physiology , Cyclic AMP Response Element Modulator/genetics , Cytoskeleton/genetics , Gene Expression Profiling , Gene Expression Regulation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Regulatory Factor X Transcription Factors , Spermatogenesis/physiology , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics , Transcription, Genetic/genetics
7.
Hum Mol Genet ; 24(17): 4997-5014, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26071364

ABSTRACT

Agenesis of the corpus callosum (AgCC) is a frequent brain disorder found in over 80 human congenital syndromes including ciliopathies. Here, we report a severe AgCC in Ftm/Rpgrip1l knockout mouse, which provides a valuable model for Meckel-Grüber syndrome. Rpgrip1l encodes a protein of the ciliary transition zone, which is essential for ciliogenesis in several cell types in mouse including neuroepithelial cells in the developing forebrain. We show that AgCC in Rpgrip1l(-/-) mouse is associated with a disturbed location of guidepost cells in the dorsomedial telencephalon. This mislocalization results from early patterning defects and abnormal cortico-septal boundary (CSB) formation in the medial telencephalon. We demonstrate that all these defects primarily result from altered GLI3 processing. Indeed, AgCC, together with patterning defects and mispositioning of guidepost cells, is rescued by overexpressing in Rpgrip1l(-/-) embryos, the short repressor form of the GLI3 transcription factor (GLI3R), provided by the Gli3(Δ699) allele. Furthermore, Gli3(Δ699) also rescues AgCC in Rfx3(-/-) embryos deficient for the ciliogenic RFX3 transcription factor that regulates the expression of several ciliary genes. These data demonstrate that GLI3 processing is a major outcome of primary cilia function in dorsal telencephalon morphogenesis. Rescuing CC formation in two independent ciliary mutants by GLI3(Δ699) highlights the crucial role of primary cilia in maintaining the proper level of GLI3R required for morphogenesis of the CC.


Subject(s)
Cilia/metabolism , Corpus Callosum/metabolism , Kruppel-Like Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Agenesis of Corpus Callosum/embryology , Agenesis of Corpus Callosum/genetics , Agenesis of Corpus Callosum/metabolism , Animals , Body Patterning/genetics , Ciliary Motility Disorders/genetics , Ciliary Motility Disorders/metabolism , Corpus Callosum/enzymology , Corpus Callosum/pathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Encephalocele/genetics , Encephalocele/metabolism , Gene Expression Regulation, Developmental , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Knockout , Mutation , Neocortex/embryology , Neocortex/metabolism , Neocortex/pathology , Nerve Tissue Proteins/genetics , Neurons/metabolism , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/metabolism , Regulatory Factor X Transcription Factors , Retinitis Pigmentosa , Transcription Factors/genetics , Transcription Factors/metabolism , Zinc Finger Protein Gli3
8.
Hum Mol Genet ; 24(9): 2578-93, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25631876

ABSTRACT

Primary cilia are complex subcellular structures that play key roles during embryogenesis by controlling the cellular response to several signaling pathways. Defects in the function and/or structure of primary cilia underlie a large number of human syndromes collectively referred to as ciliopathies. Often, ciliopathies are associated with mental retardation (MR) and malformation of the corpus callosum. However, the possibility of defects in other forebrain axon tracts, which could contribute to the cognitive disorders of these patients, has not been explored. Here, we investigate the formation of the corticothalamic/thalamocortical tracts in mice mutant for Rfx3, which regulates the expression of many genes involved in ciliogenesis and cilia function. Using DiI axon tracing and immunohistochemistry experiments, we show that some Rfx3(-/-) corticothalamic axons abnormally migrate toward the pial surface of the ventral telencephalon (VT). Some thalamocortical axons (TCAs) also fail to leave the diencephalon or abnormally project toward the amygdala. Moreover, the Rfx3(-/-) VT displays heterotopias containing attractive guidance cues and expressing the guidance molecules Slit1 and Netrin1. Finally, the abnormal projection of TCAs toward the amygdala is also present in mice carrying a mutation in the Inpp5e gene, which is mutated in Joubert Syndrome and which controls cilia signaling and stability. The presence of identical thalamocortical malformations in two independent ciliary mutants indicates a novel role for primary cilia in the formation of the corticothalamic/thalamocortical tracts by establishing the correct cellular environment necessary for its development.


Subject(s)
Body Patterning/genetics , Cerebral Cortex/metabolism , DNA-Binding Proteins/genetics , Telencephalon/metabolism , Thalamus/metabolism , Transcription Factors/genetics , Animals , Embryo, Mammalian , Homozygote , Immunohistochemistry , Kruppel-Like Transcription Factors/metabolism , Mice , Mutation , Nerve Tissue Proteins/metabolism , Neural Pathways , Neurons/metabolism , Phosphoric Monoester Hydrolases/genetics , Regulatory Factor X Transcription Factors , Telencephalon/embryology , Telencephalon/pathology , Thalamus/embryology , Thalamus/pathology , Zinc Finger Protein Gli3
9.
PLoS Genet ; 10(1): e1004118, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24465223

ABSTRACT

Inner ear mechanosensory hair cells transduce sound and balance information. Auditory hair cells emerge from a Sox2-positive sensory patch in the inner ear epithelium, which is progressively restricted during development. This restriction depends on the action of signaling molecules. Fibroblast growth factor (FGF) signalling is important during sensory specification: attenuation of Fgfr1 disrupts cochlear hair cell formation; however, the underlying mechanisms remain unknown. Here we report that in the absence of FGFR1 signaling, the expression of Sox2 within the sensory patch is not maintained. Despite the down-regulation of the prosensory domain markers, p27(Kip1), Hey2, and Hes5, progenitors can still exit the cell cycle to form the zone of non-proliferating cells (ZNPC), however the number of cells that form sensory cells is reduced. Analysis of a mutant Fgfr1 allele, unable to bind to the adaptor protein, Frs2/3, indicates that Sox2 maintenance can be regulated by MAP kinase. We suggest that FGF signaling, through the activation of MAP kinase, is necessary for the maintenance of sensory progenitors and commits precursors to sensory cell differentiation in the mammalian cochlea.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Ear, Inner/growth & development , Hair Cells, Auditory, Inner/cytology , Membrane Proteins/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle , Cell Differentiation/genetics , Cochlea/growth & development , Cochlea/metabolism , Ear, Inner/cytology , Epithelium/growth & development , Epithelium/metabolism , Gene Expression Regulation, Developmental , Membrane Proteins/metabolism , Protein Binding , Receptor, Fibroblast Growth Factor, Type 1/metabolism , SOXB1 Transcription Factors/genetics , Signal Transduction
10.
PLoS One ; 9(1): e84343, 2014.
Article in English | MEDLINE | ID: mdl-24416220

ABSTRACT

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.


Subject(s)
Craniofacial Abnormalities/enzymology , Head/growth & development , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Abnormalities, Multiple/enzymology , Abnormalities, Multiple/pathology , Abnormalities, Multiple/physiopathology , Animals , Craniofacial Abnormalities/pathology , Craniofacial Abnormalities/physiopathology , Enzyme Activation , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , MAP Kinase Signaling System , Male , Mice , Odontogenesis , Phenotype , RNA, Small Interfering/genetics , Ribosomal Protein S6 Kinases, 90-kDa/deficiency , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Tooth/anatomy & histology , Tooth/growth & development
11.
Eur J Pharmacol ; 718(1-3): 383-92, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23978568

ABSTRACT

We have investigated the effects of tCFA15, a non-peptidic compound, on the differentiation of neural stem cell-derived neurospheres, and have found that tCFA15 promotes their differentiation into neurons and reduces their differentiation into astrocytes, in a dose-dependent manner. This response is reminiscent of that resulting from the loss-of-function of Notch signaling after inactivation of the Delta-like 1 (Dll1) gene. Further analysis of the expression of genes from the Notch pathway by reverse transcriptase-PCR revealed that tCFA15 treatment results in a consistent decrease in the level of Notch1 mRNA. We have confirmed this result in other cell lines and propose that it reflects a general effect of the tCFA15 molecule. We discuss the implications of this finding with respect to regulation of Notch activity in neural stem cells, and the possible use of tCFA15 as a therapeutic tool for various pathologies that result from impairment of Notch signaling.


Subject(s)
Cell Differentiation/drug effects , Cyclohexanones/pharmacology , Fatty Alcohols/pharmacology , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Receptor, Notch1/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Gene Expression Regulation/drug effects , Mice , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Notch1/genetics
12.
Neural Dev ; 8: 13, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23829703

ABSTRACT

BACKGROUND: In order to fulfill their chemosensory function, olfactory neurons are in direct contact with the external environment and are therefore exposed to environmental aggressive factors. Olfaction is maintained through life because, unlike for other sensory neuroepithelia, olfactory neurons have a unique capacity to regenerate after trauma. The mechanisms that control the ontogenesis and regenerative ability of these neurons are not fully understood. Here, we used various experimental approaches in two model systems (chick and mouse) to assess the contribution of retinoic acid signaling in the induction of the olfactory epithelium, the generation and maintenance of progenitor populations, and the ontogenesis and differentiation of olfactory neurons. RESULTS: We show that retinoic acid signaling, although dispensable for initial induction of the olfactory placode, plays a key role in neurogenesis within this neuroepithelium. Retinoic acid depletion in the olfactory epithelium, both in chick and mouse models, results in a failure of progenitor cell maintenance and, consequently, differentiation of olfactory neurons is not sustained. Using an explant system, we further show that renewal of olfactory neurons is hindered if the olfactory epithelium is unable to synthesize retinoic acid. CONCLUSIONS: Our data show that retinoic acid is not a simple placodal inductive signal, but rather controls olfactory neuronal production by regulating the fate of olfactory progenitor cells. Retinaldehyde dehydrogenase 3 (RALDH3) is the key enzyme required to generate retinoic acid within the olfactory epithelium.


Subject(s)
Cell Differentiation/drug effects , Cell Lineage/drug effects , Neurogenesis/drug effects , Olfactory Bulb/drug effects , Olfactory Receptor Neurons/drug effects , Stem Cells/drug effects , Tretinoin/pharmacology , Animals , Cell Differentiation/physiology , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Mice , Neurogenesis/physiology , Olfactory Bulb/cytology , Olfactory Mucosa/cytology , Olfactory Mucosa/drug effects , Olfactory Receptor Neurons/cytology , Signal Transduction/drug effects , Signal Transduction/physiology , Stem Cells/cytology
13.
PLoS One ; 8(4): e62274, 2013.
Article in English | MEDLINE | ID: mdl-23638021

ABSTRACT

Retinoic acid (RA), an active derivative of the liposoluble vitamin A (retinol), acts as an important signaling molecule during embryonic development, regulating phenomenons as diverse as anterior-posterior axial patterning, forebrain and optic vesicle development, specification of hindbrain rhombomeres, pharyngeal arches and second heart field, somitogenesis, and differentiation of spinal cord neurons. This small molecule directly triggers gene activation by binding to nuclear receptors (RARs), switching them from potential repressors to transcriptional activators. The repertoire of RA-regulated genes in embryonic tissues is poorly characterized. We performed a comparative analysis of the transcriptomes of murine wild-type and Retinaldehyde Dehydrogenase 2 null-mutant (Raldh2 (-/-)) embryos - unable to synthesize RA from maternally-derived retinol - using Affymetrix DNA microarrays. Transcriptomic changes were analyzed in two embryonic regions: anterior tissues including forebrain and optic vesicle, and posterior (trunk) tissues, at early stages preceding the appearance of overt phenotypic abnormalities. Several genes expected to be downregulated under RA deficiency appeared in the transcriptome data (e.g. Emx2, Foxg1 anteriorly, Cdx1, Hoxa1, Rarb posteriorly), whereas reverse-transcriptase-PCR and in situ hybridization performed for additional selected genes validated the changes identified through microarray analysis. Altogether, the affected genes belonged to numerous molecular pathways and cellular/organismal functions, demonstrating the pleiotropic nature of RA-dependent events. In both tissue samples, genes upregulated were more numerous than those downregulated, probably due to feedback regulatory loops. Bioinformatic analyses highlighted groups (clusters) of genes displaying similar behaviors in mutant tissues, and biological functions most significantly affected (e.g. mTOR, VEGF, ILK signaling in forebrain tissues; pyrimidine and purine metabolism, calcium signaling, one carbon metabolism in posterior tissues). Overall, these data give an overview of the gene expression changes resulting from embryonic RA deficiency, and provide new candidate genes and pathways that may help understanding retinoid-dependent molecular events.


Subject(s)
Embryo, Mammalian/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Signal Transduction , Tretinoin/metabolism , Aldehyde Oxidoreductases/genetics , Animals , Cell Lineage/drug effects , Cell Lineage/genetics , Cluster Analysis , Computational Biology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Female , Gene Expression Regulation, Developmental/drug effects , Mice , Mice, Knockout , Phenotype , Pregnancy , Regulatory Sequences, Nucleic Acid , Reproducibility of Results , Tretinoin/pharmacology
14.
BMC Res Notes ; 6: 113, 2013 Mar 26.
Article in English | MEDLINE | ID: mdl-23531410

ABSTRACT

BACKGROUND: One of the key questions in developmental biology is how, from a relatively small number of conserved signaling pathways, is it possible to generate organs displaying a wide range of shapes, tissue organization, and function. The dentition and its distinct specific tooth types represent a valuable system to address the issues of differential molecular signatures. To identify such signatures, we performed a comparative transcriptomic analysis of developing murine lower incisors, mandibular molars and maxillary molars at the developmental cap stage (E14.5). RESULTS: 231 genes were identified as being differentially expressed between mandibular incisors and molars, with a fold change higher than 2 and a false discovery rate lower than 0.1, whereas only 96 genes were discovered as being differentially expressed between mandibular and maxillary molars. Numerous genes belonging to specific signaling pathways (the Hedgehog, Notch, Wnt, FGF, TGFß/BMP, and retinoic acid pathways), and/or to the homeobox gene superfamily, were also uncovered when a less stringent fold change threshold was used. Differential expressions for 10 out of 12 (mandibular incisors versus molars) and 9 out of 10 selected genes were confirmed by quantitative reverse transcription-PCR (qRT-PCR). A bioinformatics tool (Ingenuity Pathway Analysis) used to analyze biological functions and pathways on the group of incisor versus molar differentially expressed genes revealed that 143 genes belonged to 9 networks with intermolecular connections. Networks with the highest significance scores were centered on the TNF/NFκB complex and the ERK1/2 kinases. Two networks ERK1/2 kinases and tretinoin were involved in differential molar morphogenesis. CONCLUSION: These data allowed us to build several regulatory networks that may distinguish incisor versus molar identity, and may be useful for further investigations of these tooth-specific ontogenetic programs. These programs may be dysregulated in transgenic animal models and related human diseases leading to dental anomalies.


Subject(s)
Incisor/metabolism , Molar/metabolism , Oligonucleotide Array Sequence Analysis , Gene Expression Profiling , Gene Regulatory Networks , Humans , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
15.
PLoS One ; 7(3): e32447, 2012.
Article in English | MEDLINE | ID: mdl-22396766

ABSTRACT

Studies in avian models have demonstrated an involvement of retinoid signaling in early neural tube patterning. The roles of this signaling pathway at later stages of spinal cord development are only partly characterized. Here we use Raldh2-null mouse mutants rescued from early embryonic lethality to study the consequences of lack of endogenous retinoic acid (RA) in the differentiating spinal cord. Mid-gestation RA deficiency produces prominent structural and molecular deficiencies in dorsal regions of the spinal cord. While targets of Wnt signaling in the dorsal neuronal lineage are unaltered, reductions in Fibroblast Growth Factor (FGF) and Notch signaling are clearly observed. We further provide evidence that endogenous RA is capable of driving stem cell differentiation. Raldh2 deficiency results in a decreased number of spinal cord derived neurospheres, which exhibit a reduced differentiation potential. Raldh2-null neurospheres have a decreased number of cells expressing the neuronal marker ß-III-tubulin, while the nestin-positive cell population is increased. Hence, in vivo retinoid deficiency impaired neural stem cell growth. We propose that RA has separable functions in the developing spinal cord to (i) maintain high levels of FGF and Notch signaling and (ii) drive stem cell differentiation, thus restricting both the numbers and the pluripotent character of neural stem cells.


Subject(s)
Aldehyde Oxidoreductases/genetics , Spinal Cord/embryology , Tretinoin/metabolism , Aldehyde Oxidoreductases/metabolism , Animals , Cell Differentiation , Cell Lineage , Fibroblast Growth Factors/metabolism , Ganglia, Spinal/metabolism , Intermediate Filament Proteins/biosynthesis , Mice , Mice, Transgenic , Mutation , Nerve Tissue Proteins/biosynthesis , Nestin , Neural Stem Cells/cytology , Phenotype , Receptors, Notch/metabolism , Signal Transduction , Spinal Cord/cytology , Spinal Cord/metabolism , Transcription, Genetic , Tubulin/metabolism
16.
Dev Biol ; 330(2): 389-98, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19362544

ABSTRACT

The eye field is initially a large single domain at the anterior end of the neural plate and is the first indication of optic potential in the vertebrate embryo. During the course of development, this domain is subject to interactions that shape and refine the organogenic field. The action of the prechordal mesoderm in bisecting this single region into two bilateral domains has been well described, however the role of signalling interactions in the further restriction and refinement of this domain has not been previously characterised. Here we describe a role for the rostral cephalic paraxial mesoderm in limiting the extent of the eye field. The anterior transposition of this mesoderm or its ablation disrupted normal development of the eye. Importantly, perturbation of optic vesicle development occurred in the absence of any detectable changes in the pattern of neighbouring regions of the neural tube. Furthermore, negative regulation of eye development is a property unique to the rostral paraxial mesoderm. The rostral paraxial mesoderm expresses members of the bone morphogenetic protein (BMP) family of signalling molecules and manipulation of endogenous BMP signalling resulted in abnormalities of the early optic primordia.


Subject(s)
Bone Morphogenetic Proteins/physiology , Eye/embryology , Mesoderm/embryology , Animals , Chick Embryo , Coturnix/embryology , Electroporation , In Situ Hybridization , Recombinant Proteins/metabolism
17.
J Med Chem ; 47(25): 6270-82, 2004 Dec 02.
Article in English | MEDLINE | ID: mdl-15566297

ABSTRACT

In a search for inducers of neuronal differentiation to treat neurodegenerative diseases such as Alzheimer's disease, a series of indole fatty alcohols (IFAs) were prepared. 13c (n = 18) was able to promote the differentiation of neural stem cell derived neurospheres into neurons at a concentration of 10 nM. Analysis of the expression of the Notch pathway genes in neurospheres treated during the differentiation phase with 13c (n = 18) revealed a significant decrease in the transcription of the Notch 4 receptor.


Subject(s)
Alcohols/chemical synthesis , Fatty Alcohols/chemical synthesis , Free Radical Scavengers/chemical synthesis , Indoles/chemical synthesis , Neurons/drug effects , Stem Cells/drug effects , Alcohols/chemistry , Alcohols/pharmacology , Animals , Benzothiazoles , Cell Differentiation/drug effects , Fatty Alcohols/chemistry , Fatty Alcohols/pharmacology , Free Radical Scavengers/chemistry , Free Radical Scavengers/pharmacology , Free Radicals/chemistry , In Vitro Techniques , Indoles/chemistry , Indoles/pharmacology , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Neurons/cytology , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Receptor, Notch1 , Receptor, Notch2 , Receptor, Notch4 , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Receptors, Notch , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology , Structure-Activity Relationship , Sulfonic Acids/chemistry , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...