Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Dev Biol ; 457(1): 43-56, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31526806

ABSTRACT

SOX2 is essential for maintaining neurosensory stem cell properties, although its involvement in the early neurosensory development of cranial placodes remains unclear. To address this, we used Foxg1-Cre to conditionally delete Sox2 during eye, ear, and olfactory placode development. Foxg1-Cre mediated early deletion of Sox2 eradicates all olfactory placode development, and disrupts retinal development and invagination of the lens placode. In contrast to the lens and olfactory placodes, the ear placode invaginates and delaminates NEUROD1 positive neurons. Furthermore, we show that SOX2 is not necessary for early ear neurogenesis, since the early inner ear ganglion is formed with near normal central projections to the hindbrain and peripheral projections to the undifferentiated sensory epithelia of E11.5-12.5 ears. However, later stages of ear neurosensory development, in particular, the late forming auditory system, critically depend on the presence of SOX2. Our data establish distinct differences for SOX2 requirements among placodal sensory organs with similarities between olfactory and lens but not ear placode development, consistent with the unique neurosensory development and molecular properties of the ear.


Subject(s)
Ear, Inner/embryology , Neurogenesis , SOXB1 Transcription Factors/metabolism , Animals , Apoptosis , Ear, Inner/cytology , Ear, Inner/metabolism , Lens, Crystalline/embryology , Lens, Crystalline/metabolism , Mice , Mice, Knockout , Nasal Mucosa/embryology , Nasal Mucosa/metabolism , SOXB1 Transcription Factors/genetics
2.
Dev Biol ; 441(1): 67-82, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29928868

ABSTRACT

The identity of individual neuronal cell types is defined and maintained by the expression of specific combinations of transcriptional regulators that control cell type-specific genetic programs. The epithelium of the vomeronasal organ of mice contains two major types of vomeronasal sensory neurons (VSNs): 1) the apical VSNs which express vomeronasal 1 receptors (V1r) and the G-protein subunit Gαi2 and; 2) the basal VSNs which express vomeronasal 2 receptors (V2r) and the G-protein subunit Gαo. Both cell types originate from a common pool of progenitors and eventually acquire apical or basal identity through largely unknown mechanisms. The transcription factor AP-2ε, encoded by the Tfap2e gene, plays a role in controlling the development of GABAergic interneurons in the main and accessory olfactory bulb (AOB), moreover AP-2ε has been previously described to be expressed in the basal VSNs. Here we show that AP-2ε is expressed in post-mitotic VSNs after they commit to the basal differentiation program. Loss of AP-2ε function resulted in reduced number of basal VSNs and in an increased number of neurons expressing markers of the apical lineage. Our work suggests that AP-2ε, which is expressed in late phases of differentiation, is not needed to initiate the apical-basal differentiation dichotomy but for maintaining the basal VSNs' identity. In AP-2ε mutants we observed a large number of cells that entered the basal program can express apical genes, our data suggest that differentiated VSNs of mice retain a notable level of plasticity.


Subject(s)
GABAergic Neurons/metabolism , Gene Expression Regulation, Developmental/physiology , Nasal Mucosa/embryology , Sensory Receptor Cells/metabolism , Transcription Factor AP-2/biosynthesis , Vomeronasal Organ/embryology , Animals , Cell Differentiation/physiology , Mice , Mice, Transgenic , Mutation , Nasal Mucosa/cytology , Sensory Receptor Cells/cytology , Transcription Factor AP-2/genetics , Vomeronasal Organ/cytology
3.
Okajimas Folia Anat Jpn ; 94(3): 101-108, 2017.
Article in English | MEDLINE | ID: mdl-29681587

ABSTRACT

The purpose of this study is to describe the Hasner's membrane which is the main factor of congenital nasolacrimal duct obstruction. Hasner's membrane at the nasal end of the fetal nasolacrimal duct (NLD) is considered to rupture at and after birth. However, topographical anatomy around the membrane as well as a mechanism of rupture seems to be still obscure. We observed frontal or sagittal sections of 20 late-stage fetuses (28-33 weeks) and found the on-going rupture in 2 specimens. The present sections demonstrated that 1) the nasal dilation was not a simple ball-like structure but extended posteriorly and laterally; 2) dilation of the NLD consistently involved the lacrimal sac; 3) Hasner's membrane and ductal mucosal layer contained no macrophages and no or few arteries and nerves. The posterior extension of the NLD end ranged from 1-2 mm, while the lateral extension 3-5 mm although a site of the thinnest membrane varied in location between specimens. Moreover, the thickest NLD due to dilation was in the slightly orbital or upper side of the nasal end. Therefore, before surgical treatment of Hasner's membrane, evaluation using medical images seems to be necessary. Since the nasal epithelium on Hasner's membrane was most likely to destroy earlier than the NLD mucosal lining, observations of the membrane from the nasal cavity seemed helpful for diagnosis at which site would be broken and when.


Subject(s)
Nasolacrimal Duct/embryology , Humans , Lacrimal Duct Obstruction/congenital , Nasal Mucosa/embryology , Nasolacrimal Duct/immunology
4.
Dev Biol ; 419(2): 348-356, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27590203

ABSTRACT

Hypertrophy, hyperplasia and altered mucus secretion from the respiratory submucosal glands (SMG) are characteristics of airway diseases such as cystic fibrosis, asthma and chronic bronchitis. More commonly, hyper-secretion of the nasal SMGs contributes to allergic rhinitis and upper airway infection. Considering the role of these glands in disease states, there is a significant dearth in understanding the molecular signals that regulate SMG development and patterning. Due to the imperative role of FGF signalling during the development of other branched structures, we investigated the role of Fgf10 during initiation and branching morphogenesis of murine nasal SMGs. Fgf10 is expressed in the mesenchyme around developing SMGs while expression of its receptor Fgfr2 is seen within glandular epithelial cells. In the Fgf10 null embryo, Steno's gland and the maxillary sinus gland were completely absent while other neighbouring nasal glands showed normal duct elongation but defective branching. Interestingly, the medial nasal glands were present in Fgf10 homozygotes but missing in Fgfr2b mutants, with expression of Fgf7 specifically expressed around these developing glands, indicating that Fgf7 might compensate for loss of Fgf10 in this group of glands. Intriguingly the lateral nasal glands were only mildly affected by loss of FGF signalling, while these glands were missing in Eda mutant mice, where the Steno's and maxillary sinus gland developed as normal. This analysis reveals that regulation of nasal gland development is complex with different subsets of glands being regulated by different signalling pathways. This analysis helps shed light on the nasal gland defects observed in patients with hypohidrotic ectodermal dysplasia (HED) (defect EDA pathway) and LADD syndrome (defect FGFR2b pathway).


Subject(s)
Ectodysplasins/physiology , Exocrine Glands/embryology , Fibroblast Growth Factor 10/physiology , Receptor, Fibroblast Growth Factor, Type 2/physiology , Signal Transduction/physiology , Animals , Ectodysplasins/deficiency , Ectodysplasins/genetics , Endoscopic Mucosal Resection , Exocrine Glands/metabolism , Exocrine Glands/ultrastructure , Female , Fibroblast Growth Factor 10/deficiency , Fibroblast Growth Factor 10/genetics , Fibroblast Growth Factor 7/physiology , Male , Maxillary Sinus/embryology , Maxillary Sinus/ultrastructure , Mesoderm/metabolism , Mice , Morphogenesis , Nasal Mucosa/embryology , Nasal Mucosa/ultrastructure , Receptor, Fibroblast Growth Factor, Type 2/deficiency , Receptor, Fibroblast Growth Factor, Type 2/genetics
5.
Okajimas Folia Anat Jpn ; 87(1): 5-10, 2010 May.
Article in English | MEDLINE | ID: mdl-20715566

ABSTRACT

Immunoreactivity of the calcium binging protein calretinin is often used as a marker of olfactory neurons. Although the immunoreactivity and density of olfactory neurons are known to change between developmental stages in the human fetus, previous descriptions have been limited to the olfactory epithelium and/or the nasal septum and have not included the entire nasal cavity. Using horizontal semi serial sections of heads of six mid-term fetuses (9-15 weeks of gestation), we examined the topographical anatomy of calretinin-positive olfactory neurons. By 9 weeks of gestation, the distribution of calretinin-positive cells reached levels inferior to the developing inferior meatus. By 12 weeks, concentrations in the inferior end had reached the level of the inferior meatus and the middle meatus carried abundant positive cells. However, by 15 weeks, calretinin positive cells were restricted to levels superior to the middle meatus and in the vomeronasal organ. Placode-derived cells are initially distributed antero-inferiorly along the nasal epithelium, but most lose their calretinin immunoreactivity. They might differentiate into the neuroendocrine cells embedded between nasal respiratory epithelial cells. The final differentiation of calretinin-positive cells was likely to require connection to the olfactory bulb and accessory bulb.


Subject(s)
Fetal Development/physiology , Nasal Mucosa/embryology , S100 Calcium Binding Protein G/metabolism , Calbindin 2 , Gestational Age , Humans , Nasal Mucosa/cytology , Nasal Mucosa/metabolism , Neurons/cytology , Neurons/metabolism , Olfactory Nerve/cytology , Olfactory Nerve/metabolism
6.
Annu Rev Immunol ; 26: 627-50, 2008.
Article in English | MEDLINE | ID: mdl-18370924

ABSTRACT

Secondary lymphoid organs develop during embryogenesis or in the first few weeks after birth according to a highly coordinated series of interactions between newly emerging hematopoietic cells and immature mesenchymal or stromal cells. These interactions are orchestrated by homeostatic chemokines, cytokines, and growth factors that attract hematopoietic cells to sites of future lymphoid organ development and promote their survival and differentiation. In turn, lymphotoxin-expressing hematopoietic cells trigger the differentiation of stromal and endothelial cells that make up the scaffolding of secondary lymphoid organs. Lymphotoxin signaling also maintains the expression of adhesion molecules and chemokines that govern the ultimate structure and function of secondary lymphoid organs. Here we describe the current paradigm of secondary lymphoid organ development and discuss the subtle differences in the timing, molecular interactions, and cell types involved in the development of each secondary lymphoid organ.


Subject(s)
Lymphoid Tissue/embryology , Lymphoid Tissue/growth & development , Animals , Cytokines/physiology , Lymph Nodes/cytology , Lymph Nodes/embryology , Lymph Nodes/growth & development , Lymphoid Tissue/cytology , Mice , Nasal Mucosa/cytology , Nasal Mucosa/embryology , Nasal Mucosa/growth & development , Peyer's Patches/cytology , Peyer's Patches/embryology , Peyer's Patches/growth & development , Signal Transduction/physiology
7.
Dev Dyn ; 236(4): 961-70, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17330888

ABSTRACT

The mammalian epidermis is the first line of defense against external environmental challenges including dehydration. The epidermis undergoes a highly intricate developmental program in utero, transforming from a simple to a complex stratified epithelium. During this process of stratification and differentiation, epidermal keratinocytes express a defined set of structural proteins, mainly keratins, whose expression is controlled by largely unknown mechanisms. In order to identify novel factors contributing to epidermal morphogenesis, we performed a global transcriptional analysis of the developing mouse epidermis after separating it from the underlying dermis (E12.5-E15.5). Unexpectedly, the recently identified genes encoding secreted peptides dermokine (Dmkn), keratinocyte differentiation-associated protein (krtdap), and suprabasin (Sbsn) as well as a largely uncharacterized embryonic keratin (Krt77), were among the most highly differentially expressed genes. The three genes encoding the secreted proteins form a cluster in an approximately 40-Kb locus on human chromosome 19 and the syntenic region on mouse chromosome 7 known as the stratified epithelium secreted peptides complex (SSC). Using whole mount in situ hybridization, we show that these genes show a coordinated spatio-temporal expression pattern during epidermal morphogenesis. The expression of these genes initiates in the nasal epithelium and correlates with the initiation of other epidermal differentiation markers such as K1 and loricrin (Byrne et al. [1994] Development 120:2369-2383), as well as the initiation of barrier formation. Our observations reveal a coordinated mode of expression of the SSC genes as well as the correlation of their initiation in the nasal epithelium with the initiation of barrier formation at this site.


Subject(s)
Epidermis/embryology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Animals , Antigens, Differentiation/genetics , Aspartic Acid Endopeptidases/genetics , Cell Adhesion/genetics , Cell Communication/genetics , Epidermis/enzymology , Epidermis/metabolism , Intercellular Signaling Peptides and Proteins , Keratins/genetics , Mice , Mice, Inbred C57BL , Morphogenesis/genetics , Nasal Mucosa/embryology , Phosphoproteins/genetics , Proteins/genetics
8.
Int J Dev Biol ; 51(1): 67-77, 2007.
Article in English | MEDLINE | ID: mdl-17183466

ABSTRACT

Cleft of the secondary palate is one of the most common congenital birth defects in humans. The primary cause of cleft palate formation is a failure of fusion of bilateral palatal shelves, but rupture of the once fused palate has also been suggested to take place in utero. The possibility of post-fusion rupture of the palate in humans has hardly been accepted, mainly because in all the cleft palate cases, the cleft palatal edge is always covered with intact epithelium. To verify whether the intrauterine environment of the fetus plays roles in wound healing when the once fused palate is torn apart, we artificially tore apart fetal mouse palates after fusion and cultivated them in culture medium with or without mouse or human amniotic fluid. We thereby found that the wounded palatal edge became completely covered with flattened epithelium after 36 hours in culture with amniotic fluid, but not in culture without amniotic fluid. Using histological and scanning electron microscopic analyses of the healing process, it was revealed that the epithelium covering the wound was almost exclusively derived from the adjacent nasal epithelium, but not from the oral epithelium. Such actions of amniotic fluid on the fetal wound were never simulated by exogenous epidermal growth factor (EGF), albumin, or both. In addition, the rapid epithelialization induced by amniotic fluid was not prevented by either PD168393 (an inhibitor of the EGF receptor-specific tyrosine kinase) or SB431542 (a specific inhibitor of TGFbeta receptor type I/ALK5). The present study provides new insights into the unique biological actions of amniotic fluid in the repair of injured fetal palate.


Subject(s)
Amniotic Fluid/metabolism , Fetus/physiology , Palate/embryology , Palate/injuries , Wound Healing , Activin Receptors, Type I/metabolism , Animals , Cleft Palate/embryology , Epidermal Growth Factor/pharmacology , Epithelium/metabolism , ErbB Receptors/antagonists & inhibitors , Female , Humans , Male , Mice , Mice, Inbred ICR , Microscopy, Electron, Scanning , Nasal Mucosa/embryology , Nasal Mucosa/metabolism , Organ Culture Techniques/methods , Palate/cytology , Palate/metabolism , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/metabolism , Rupture
9.
Histochem Cell Biol ; 125(4): 337-49, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16273384

ABSTRACT

The olfactory marker protein (OMP) is expressed in mature chemosensory neurons in the nasal neuroepithelium. Here, we report the identification of a novel population of OMP-expressing neurons located bilaterally in the anterior/dorsal region of each nasal cavity at the septum. These cells are clearly separated from the regio olfactoria, harboring the olfactory sensory neurons. During mouse development, the arrangement of the anterior OMP-cells undergoes considerable change. They appear at about stage E13 and are localized in the nasal epithelium during early stages; by epithelial budding, ganglion-shaped clusters are formed in the mesenchyme during the perinatal phase, and a filiform layer directly underneath the nasal epithelium is established in adults. The anterior OMP-cells extend long axonal processes which form bundles and project towards the brain. The data suggest that the newly discovered group of OMP-cells in the anterior region of the nasal cavity may serve a distinct sensory function.


Subject(s)
Nasal Cavity/metabolism , Nasal Mucosa/metabolism , Olfactory Marker Protein/metabolism , Olfactory Receptor Neurons/metabolism , Animals , Animals, Newborn/metabolism , Mice , Nasal Cavity/embryology , Nasal Mucosa/embryology , Nasal Septum/embryology , Nasal Septum/metabolism , Olfactory Receptor Neurons/embryology , Tubulin/metabolism
10.
Drug Metab Dispos ; 33(10): 1423-8, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16014766

ABSTRACT

Despite recent progress in the identification and characterization of numerous nasal biotransformation enzymes in laboratory animals, the expression of biotransformation genes in human nasal mucosa remains difficult to study. Given the potential role of nasal biotransformation enzymes in the metabolism of airborne chemicals, including fragrance compounds and therapeutic agents, as well as the potential interspecies differences between laboratory animals and humans, it would be highly desirable to identify those biotransformation genes that are expressed in human nasal mucosa. In this study, a global gene expression analysis was performed to compare biotransformation enzymes expressed in human fetal and adult nasal mucosa to those expressed in liver. The identities of a list of biotransformation genes with apparently nasal mucosa-selective expression were subsequently confirmed by RNA-polymerase chain reaction (PCR) and DNA sequencing of the PCR products. Further quantitative RNA-PCR experiments indicated that, in the fetus, aldehyde dehydrogenase 6 (ALDH6), CYP1B1, CYP2F1, CYP4B1, and UDP glucuronosyltransferase 2A1 are expressed preferentially in the nasal mucosa and that ALDH7, flavin-containing monooxygenase 1, and glutathione S-transferase P1 are at least as abundant in the nasal mucosa as in the liver. The nasal mucosal expression of CYP2E1 was also detected. These findings provide a basis for further explorations of the metabolic capacity of the human nasal mucosa for xenobiotic compounds.


Subject(s)
Nasal Mucosa/embryology , Nasal Mucosa/enzymology , Oxidoreductases/genetics , Transferases/genetics , Adult , Aged , Biotransformation , Female , Gene Expression Profiling , Humans , Liver/embryology , Liver/enzymology , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Oxidoreductases/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transferases/metabolism
11.
Chem Res Toxicol ; 18(6): 913-8, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15962925

ABSTRACT

Among human P450s studied to date, P450 2A13 is the most efficient catalyst of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) alpha-hydroxylation. This reaction is a key bioactivation pathway in NNK-induced carcinogenesis. P450 2A13 mRNA has been detected in human tissues, but it is unknown whether the enzyme is functional in vivo. Therefore, we studied NNK alpha-hydroxylation in human fetal nasal mucosal microsomes, which have been shown to contain high levels of P450 2A protein, presumed to be a mixture of P450 2A6 and 2A13. The microsomes efficiently catalyzed NNK alpha-hydroxylation at the methylene and methyl carbons, as well as carbonyl reduction. Antibodies against mouse P450 2A5 inhibited alpha-hydroxylation by these microsomes greater than 90%. K(m) and V(max) values for alpha-methylene hydroxylation were 6.5 +/- 1.1 muM and 3.0 +/- 0.1 pmol/min/mg; for alpha-methyl hydroxylation, they were 6.7 +/- 0.8 microM and 0.85 +/- 0.03 pmol/min/mg. The K(m) values agree closely with those for NNK metabolism by P450 2A13. Using a new technique, we separated P450 2A13 from P450 2A6 by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Quantitative immunoblot analysis indicated that the level of P450 2A13 in the pooled fetal nasal microsome sample used for kinetic analysis was approximately 1.6 pmol/mg protein. In the same sample, P450 2A6 was not detected (detection limit, 67 fmol/mg protein). These kinetic, immunoinhibition, and immunoblot data confirm that P450 2A13 is a functional enzyme and the catalyst of NNK alpha-hydroxylation in human fetal nasal mucosa. The results are also the first to demonstrate high efficiency NNK alpha-hydroxylation in a human tissue.


Subject(s)
Carcinogens/pharmacokinetics , Microsomes, Liver/enzymology , Nasal Mucosa/enzymology , Nitrosamines/pharmacokinetics , Adult , Biotransformation , Carcinogens/metabolism , Female , Fetus , Gestational Age , Humans , Hydroxylation , Male , Middle Aged , Nasal Mucosa/embryology , Nitrosamines/metabolism , Nicotiana/metabolism
12.
Anat Sci Int ; 78(1): 25-35, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12680467

ABSTRACT

Abstract Epidermal growth factor (EGF) and receptor (-R) signaling pathway is required for epithelial cell growth and differentiation such as the degeneration of the medial edge epithelial cells during the fusion process of secondary palate formation. As epithelial fusion takes place during primary palate formation, we investigated the involvement of the EGF-R in fusion of the medial (MNP) and lateral (LNP) nasal prominences of the mouse embryo was examined. Immunoreactivity of EGF-R was investigated in embryonic day 10 embryos (32-37 somite stages). The EGF-R immunoreactivity was observed in the nasal epithelia of the presumptive fusion area before fusion. It became undetectable just prior to the fusion and faintly reappeared at the time of the fusion. In contrast, the non-fusing epithelial cells of the nasal groove maintained the immunoreactivity throughout these stages. In order to elucidate whether the EGF/EGF-R signaling pathway was involved in nasal epithelial fusion, EGF solution was injected into the exocoelum of explanted mouse embryos, and the embryos were cultured for 18-24 h by whole embryo culture (WEC). This exogenous EGF inhibited fusion of nasal prominences in 66.7-81.5% of the embryos. Treatment with EGF for 4-14 h showed that exogenous EGF disturbed the EGF-R disappearance and normal alteration of epithelial cell morphology in the fusion area. These results suggest that temporal disappearance of the EGF/EGF-R signaling from presumptive fusion of the nasal prominences is required for morphological change of the epithelial cells leading to the fusion of MNP and LNP.


Subject(s)
Cell Differentiation/physiology , Embryo, Mammalian/embryology , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Nasal Cavity/embryology , Nasal Mucosa/embryology , Palate/embryology , Animals , Body Patterning/drug effects , Body Patterning/physiology , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/physiology , Embryo, Mammalian/metabolism , Embryo, Mammalian/ultrastructure , Epidermal Growth Factor/pharmacology , ErbB Receptors/drug effects , Female , Fetus , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Scanning , Nasal Cavity/metabolism , Nasal Cavity/ultrastructure , Nasal Mucosa/metabolism , Nasal Mucosa/ultrastructure , Organ Culture Techniques , Palate/metabolism , Palate/ultrastructure , Pregnancy
13.
Proc Natl Acad Sci U S A ; 99(25): 16111-6, 2002 Dec 10.
Article in English | MEDLINE | ID: mdl-12454286

ABSTRACT

Knockout of the murine retinoic acid (RA)-synthesizing enzyme retinaldehyde dehydrogenase 2 (RALDH2) gene leads to early morphogenetic defects and embryonic lethality. Using a RA-responsive reporter transgene, we have looked for RA-generating activities in Raldh2-null mouse embryos and investigated whether these activities could be ascribed to the other known RALDH enzymes (RALDH1 and RALDH3). To this end, the early defects of Raldh2(-/-) embryos were rescued through maternal dietary RA supplementation under conditions that do not interfere with the activity of the reporter transgene in WT embryos. We show that RALDH2 is responsible for most of the patterns of reporter transgene activity in the spinal cord and trunk mesodermal derivatives. However, reporter transgene activity was selectively detected in Raldh2(-/-) embryos within the mesonephric area that expresses RALDH3 and in medial-ventral cells of the spinal cord and posterior hindbrain, up to the level of the fifth rhombomere. The craniofacial patterns of RA-reporter activity were unaltered in Raldh2(-/-) mutants. Although these patterns correlated with the presence of Raldh1 andor Raldh3 transcripts in eye, nasal, and inner ear epithelia, no such correlation was found within forebrain neuroepithelium. These data suggest the existence of additional RA-generating activities in the differentiating forebrain, hindbrain, and spinal cord, which, along with RALDH1 and RALDH3, may account for the development of Raldh2(-/-) mutants once these have been rescued for early lethality.


Subject(s)
Aldehyde Oxidoreductases/physiology , Tretinoin/metabolism , Administration, Oral , Aldehyde Oxidoreductases/deficiency , Aldehyde Oxidoreductases/genetics , Animals , Ear, Inner/embryology , Ear, Inner/metabolism , Epithelial Cells/metabolism , Eye/embryology , Eye/metabolism , Female , Fetal Diseases/drug therapy , Gene Expression Regulation, Developmental , Genes, Lethal , Genes, Reporter , Gestational Age , Lac Operon , Mesonephros/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Nasal Mucosa/embryology , Nasal Mucosa/metabolism , Organ Specificity , Pregnancy , Prosencephalon/embryology , Prosencephalon/metabolism , Rhombencephalon/embryology , Rhombencephalon/metabolism , Spinal Cord/embryology , Spinal Cord/metabolism , Tretinoin/therapeutic use
14.
Surg Radiol Anat ; 24(1): 27-32, 2002 Feb.
Article in English | MEDLINE | ID: mdl-12197007

ABSTRACT

The aim of this study was to describe the microvascularization of the area of junction located between the integuments of the nasal ala and the respiratory mucosa. This study is part of an overall study on the microvascularization of the mucocutaneous junctions of the head. It was undertaken on histological or clarified sections of noses from fetuses, newborns and adults whose vascular system was injected with Indian ink agar. The mucocutaneous junction of the nose shows similarities with the mucocutaneous junction of the oral cavity. Under a well-defined avascular and thick epithelium lies a vascular papillary network with typical loops less high than in the lips. The angioarchitectonics of superficial and deep vascular reticular networks is similar to that of the lips but they are less dense.


Subject(s)
Fetus/anatomy & histology , Nasal Mucosa/blood supply , Nose/blood supply , Adult , Humans , Infant, Newborn , Microcirculation/embryology , Nasal Mucosa/anatomy & histology , Nasal Mucosa/embryology , Nose/anatomy & histology , Nose/embryology
15.
Endocrinology ; 143(7): 2503-7, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12072381

ABSTRACT

Sex steroids influence LHRH neuronal activity, exerting a negative or positive feedback action, depending on the reproductive state of the animal. Recent evidence indicates that LHRH neurons possess the estrogen receptor beta (ERbeta) subtype postnatally, suggesting that estrogen may act, in part, directly on LHRH neurons. In this study, we identified ERbeta transcript in prenatal LHRH neurons as a function of age. Single-cell cDNA pools were made from LHRH neurons maintained for 7, 14, and 28 d in vitro (div). Screening of the cDNA pools by PCR with ERbeta-specific primers revealed ERbeta-positive LHRH neurons at all three ages. However, the number of LHRH cells coexpressing ERbeta transcript decreased dramatically between 14 (6/10) and 28 div (1/10). None of the LHRH cells were positive for ERalpha transcript. These results indicate that developing LHRH neurons express the transcript for ERbeta and suggest that continued expression of ERbeta is either a characteristic of LHRH neurons that may require cues from the central nervous system and/or periphery or predetermined to be maintained in a subpopulation of LHRH neurons.


Subject(s)
Gonadotropin-Releasing Hormone/physiology , Nasal Mucosa/innervation , Nasal Mucosa/metabolism , Neurons/metabolism , Receptors, Estrogen/biosynthesis , Receptors, Estrogen/genetics , Animals , Cell Separation , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Embryonic and Fetal Development/physiology , Estrogen Receptor beta , Female , Immunohistochemistry , Mice , Nasal Mucosa/embryology , Nerve Fibers/metabolism , Organ Culture Techniques , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Transcription, Genetic
16.
Ital J Anat Embryol ; 107(4): 257-65, 2002.
Article in English | MEDLINE | ID: mdl-12611477

ABSTRACT

The mechanism of formation of the maxillary sinuses is not elucidated as yet, although their morphology during embryogenesis is well described. In the prenatal period, the pneumatization hypothesis is not valid. As the molecular approach to this problem is difficult to apply to human samples, we decided to apply immunohistochemical reactions to analyse the synthesis of selected molecules involved in the rebuilding of tissues. Hematoxylin-eosin staining and immunohistochemical reactions for the detection of MMPs (matrix metalloproteinases), one of their inhibitor TIMP 1 (tissue inhibitor of MMPs), BMP 6 (bone morphogenetic protein 6) and TGF-beta (transforming growth factor beta) were performed in the epithelium the mucosa of the maxillary sinuses of several human foetuses from the collection of the Anatomical Institute. The age of the foetuses was 8, 11, 15, 16, 17, 18 and 22 weeks. An intense positive reaction for MMPs 1, 2 and 3 was found in the mucosal epithehum of developing sinuses in the whole series of foetuses was found. The reaction was more intense in advanced stages of foetal development. Tissue derived inhibitor TIMP was hardly detectable, regardless of the age of samples. However, the intensity of the reaction for TGFbeta was strong in both young and more mature sinus epithelium. The presence of BMP 6, a member of the superfamily of TGFbeta, was detected although the intensity of this reaction in the epithelium was rather weak. Both TGFbeta and BMP 6 are well known as regulators of differentiation in the course of organogenesis. Results of the histochemical analysis suggest the possible involvement of the epithelium in the growth and formation of the maxillary sinuses. The main argument for this is intense reaction for MMP proteases which, as in bone, regulate the turnover and rebuilding processes of the extracellular matrix (ECM).


Subject(s)
Cell Differentiation/physiology , Epithelial Cells/cytology , Fetus/embryology , Maxillary Sinus/embryology , Nasal Mucosa/embryology , Body Patterning/physiology , Bone Morphogenetic Protein 6 , Bone Morphogenetic Proteins/metabolism , Epithelial Cells/metabolism , Fetus/cytology , Fetus/metabolism , Gene Expression Regulation, Developmental/physiology , Humans , Immunohistochemistry , Matrix Metalloproteinases/metabolism , Maxillary Sinus/cytology , Maxillary Sinus/metabolism , Nasal Mucosa/cytology , Nasal Mucosa/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transforming Growth Factor beta/metabolism
17.
Dev Biol ; 220(2): 343-57, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10753521

ABSTRACT

To explain the disappearance of medial edge epithelial (MEE) cells during palatal fusion, programmed cell death, epithelial-mesenchymal transformation, and migration of these cells to the oral and nasal epithelia have been proposed. However, MEE cell death has not always been accepted as a mechanism involved in midline epithelial seam disappearance. Similarly, labeling of MEE cells with vital lipophilic markers has not led to a clear conclusion as to whether MEE cells migrate, transform into mesenchyme, or both. To clarify these controversies, we first utilized TUNEL techniques to detect apoptosis in mouse palates at the fusion stage and concomitantly analyzed the presence of macrophages by immunochemistry and confocal microscopy. Second, we in vitro infected the MEE with the replication-defective helper-free retroviral vector CXL, which carries the Escherichia coli lacZ gene, and analyzed beta-galactosidase activity in cells after fusion to follow their fate. Our results demonstrate that MEE cells die and transform into mesenchyme during palatal fusion and that dead cells are phagocytosed by macrophages. In addition, we have investigated the effects of the absence of transforming growth factor beta(3) (TGF-beta(3)) during palatal fusion. Using environmental scanning electron microscopy and TUNEL labeling we compared the MEE of the clefted TGF-beta(3) null and wild-type mice. We show that MEE cell death in TGF-beta(3) null palates is greatly reduced at the time of fusion, revealing that TGF-beta(3) has an important role as an inducer of apoptosis during palatal fusion. Likewise, the bulging cells observed on the MEE surface of wild-type mice prior to palatal shelf contact are very rare in the TGF-beta(3) null mutants. We hypothesize that these protruding cells are critical for palatal adhesion, being morphological evidence of increased cell motility/migration.


Subject(s)
Nasal Mucosa/embryology , Palate/embryology , Animals , Apoptosis , Epithelium/embryology , Epithelium/metabolism , Genotype , Immunohistochemistry , In Situ Nick-End Labeling , Macrophages/metabolism , Mesoderm/metabolism , Mice , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron, Scanning , Nasal Mucosa/metabolism , Palate/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
18.
J Biol Chem ; 274(19): 13698-703, 1999 May 07.
Article in English | MEDLINE | ID: mdl-10224143

ABSTRACT

We have identified a novel gene transcript of approximately 1.1 kilobases in length that is expressed in the presumptive nasal epithelium of the mouse embryo. In situ hybridization analysis shows discrete regions of expression associated with the palate, nasal septum, and nasal conchae. This transcript is also expressed strongly in the trachea and bronchi of the adult lung. Screening of a mouse heart cDNA library yielded several overlapping clones to give a continuous sequence of 1113 bases, containing an open reading frame of 278 codons comprising the complete mRNA. No significant homologies with known genes were observed at the nucleotide level; limited amino acid homology with two salivary gland-specific proteins was noted. A search for functionally significant protein motifs revealed consensus sequences for N-glycosylation, protein kinase C and casein kinase phosphorylation, and a leucine zipper. Additionally, we observed a unique amino acid sequence pattern, consisting of the residues Gly-(Leu/Pro/Gln)-(Pro/Leu)-Leu-Pro-Leu, repeated four times near the amino-terminal portion of the protein with two amino acid residues separating the repeats. Based on these observations, we propose that we have identified a new gene, which we call plunc (for palate, lung, and nasal epithelium clone; GenBankTM accession number U69172).


Subject(s)
Gene Expression Regulation, Developmental , Glycoproteins/genetics , Lung/metabolism , Nasal Mucosa/metabolism , Palate/metabolism , Phosphoproteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary , Embryonic and Fetal Development/genetics , Female , Mice , Mice, Inbred ICR , Molecular Sequence Data , Nasal Mucosa/embryology , Palate/embryology , Pregnancy , Sequence Homology, Amino Acid
19.
Brain Res Dev Brain Res ; 110(1): 69-81, 1998 Sep 10.
Article in English | MEDLINE | ID: mdl-9733924

ABSTRACT

Three mouse olfactory receptors have been cloned and sequenced and were found to be expressed in different zones of the olfactory epithelium. In situ hybridisation (ISH) results showed that each olfactory receptor was expressed at an early stage in development (E12), was not dependent on the maturation of the receptor neurons, and was present long before the onset of odour detection. Cells positive for these same olfactory receptors and the G-protein (Gbeta) were also found in non-neural regions of the nasal epithelium in the earlier stages of development (E12-16). Ncam, and Big-2 expression were, however, restricted to the region of developing olfactory neurons. Ncam expression appeared in advance of the olfactory receptor expression, while Big-2 appeared after olfactory receptor expression and neither were expressed in cells outside the olfactory epithelium. Both showed the highest number of positive cells in the early post-partum period when olfactory detection is functional. Ncam is known to be involved in guidance of the developing olfactory axons and was expressed earlier than any of the olfactory receptors, while Big-2 appears somewhat later (E14) at a time when developing axons reach the olfactory bulb. Moreover the highest periods of expression occur at post-natal day 7 when a proliferation of bulbar glomeruli are observed, suggesting the role of Big-2 to be primarily concerned with synaptogenesis.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , GTP-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Nasal Mucosa/innervation , Neural Cell Adhesion Molecules/genetics , Olfactory Receptor Neurons/metabolism , Receptors, Odorant/genetics , Aging/metabolism , Amino Acid Sequence , Animals , Cell Adhesion Molecules, Neuronal/biosynthesis , Conserved Sequence , Contactins , DNA Primers , Embryonic and Fetal Development , GTP-Binding Proteins/biosynthesis , In Situ Hybridization , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Nasal Mucosa/embryology , Nasal Mucosa/growth & development , Neural Cell Adhesion Molecules/biosynthesis , Polymerase Chain Reaction , Receptors, Odorant/biosynthesis , Receptors, Odorant/chemistry , Sequence Alignment , Sequence Homology, Amino Acid
20.
Anat Rec ; 248(3): 307-21, 1997 07.
Article in English | MEDLINE | ID: mdl-9214547

ABSTRACT

BACKGROUND: The olfactory epithelial sustentacular cells may support the survival and function of olfactory receptor neurons, but few reagents are available to mark and manipulate such cells. METHODS: Novel nasal cell-specific monoclonal antibodies were generated using whole cultured rat olfactory mucosal cells as the antigenic stimuli. They were characterized by immunostaining at the light level in rat tissues and newborn rat olfactory cell cultures, and at the electron microscopic level in adult tissues using freeze-substitution, post-embedding staining. RESULTS: An IgMkappa monoclonal antibody designated 1F4 selectively labeled apical surfaces of the rat olfactory and respiratory epithelia in tissue sections and what appeared to be sustentacular cells in olfactory cell cultures. Using electron microscopy, 1F4 bound selectively to the microvilli of sustentacular cells and ductal cells of Bowman's glands in the olfactory epithelium, and to the microvilli and cilia of ciliated but not secretory cells in the respiratory epithelium. No staining was detected in olfactory receptor neurons, basal cells, or two types of microvilli-bearing cells that differed from sustentacular cells. A contrasting antibody, 2H4, bound to granules of secretory respiratory cells. Developmental expression of 1F4 binding began at E17 and increased at and after E18/E19. Bulbectomy did not alter 1F4 immunoreactivity. Cell culture studies found that the 1F4 epitope was external and insensitive to trypsin treatment, and that both 1F4 and 2H4 positive cells required contact with aggregated cells for survival up to fifteen days in vitro. CONCLUSIONS: The antibody 1F4 is a useful marker and potential manipulation reagent specific for sustentacular cells and their microvilli.


Subject(s)
Antibodies, Monoclonal , Microvilli/immunology , Nasal Mucosa/immunology , Nasal Mucosa/ultrastructure , Animals , Animals, Newborn , Antibodies, Monoclonal/biosynthesis , Antibody Specificity , Biomarkers , Cells, Cultured , Cilia/immunology , Cytoplasmic Granules/immunology , Immunoglobulin M/biosynthesis , Immunohistochemistry , Microscopy, Electron , Microvilli/ultrastructure , Nasal Mucosa/embryology , Olfactory Bulb/physiology , Rats , Rats, Sprague-Dawley , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...