Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 269
Filter
1.
PLoS Biol ; 18(9): e3000852, 2020 09.
Article in English | MEDLINE | ID: mdl-32931487

ABSTRACT

Olfaction in most animals is mediated by neurons bearing cilia that are accessible to the environment. Olfactory sensory neurons (OSNs) in chordates usually have multiple cilia, each with a centriole at its base. OSNs differentiate from stem cells in the olfactory epithelium, and how the epithelium generates cells with many centrioles is not yet understood. We show that centrioles are amplified via centriole rosette formation in both embryonic development and turnover of the olfactory epithelium in adult mice, and rosette-bearing cells often have free centrioles in addition. Cells with amplified centrioles can go on to divide, with centrioles clustered at each pole. Additionally, we found that centrioles are amplified in immediate neuronal precursors (INPs) concomitant with elevation of mRNA for polo-like kinase 4 (Plk4) and SCL/Tal1-interrupting locus gene (Stil), key regulators of centriole duplication. These results support a model in which centriole amplification occurs during a transient state characterized by elevated Plk4 and Stil in early INP cells. These cells then go on to divide at least once to become OSNs, demonstrating that cell division with amplified centrioles, known to be tolerated in disease states, can occur as part of a normal developmental program.


Subject(s)
Cell Division/physiology , Centrioles/physiology , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Olfactory Receptor Neurons/physiology , Aging/physiology , Animals , Cell Cycle/physiology , Cells, Cultured , Embryo, Mammalian , Embryonic Development/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Olfactory Cortex/cytology , Olfactory Cortex/embryology , Olfactory Mucosa/cytology , Olfactory Mucosa/embryology , Olfactory Mucosa/ultrastructure , Olfactory Receptor Neurons/cytology , Olfactory Receptor Neurons/ultrastructure
2.
J Anat ; 237(2): 225-240, 2020 08.
Article in English | MEDLINE | ID: mdl-32314400

ABSTRACT

Despite the long-held assumption that olfaction plays a relatively minor role in the behavioral ecology of birds, crown-group avians exhibit marked phylogenetic variation in the size and form of the olfactory apparatus. As part of a larger effort to better understand the role of olfaction and olfactory tissues in the evolution and development of the avian skull, we present the first quantitative analysis of ontogenetic scaling between olfactory features [olfactory bulbs (OBs) and olfactory turbinates] and neighboring structures (cerebrum, total brain, respiratory turbinates) based on the model organism Gallus gallus. The OB develops under the predictions of a concerted evolutionary model with rapid early growth that is quickly overcome by the longer, sustained growth of the larger cerebrum. A similar pattern is found in the nasal cavity where the morphologically simple (non-scrolled) olfactory turbinates appear and mature early, with extended growth characterizing the larger and scrolled respiratory turbinates. Pairwise regressions largely recover allometric relationships among the examined structures, with a notable exception being the isometric trajectory of the OB and olfactory turbinate. Their parallel growth suggests a unique regulatory pathway that is likely driven by the morphogenesis of the olfactory nerve, which serves as a structural bridge between the two features. Still, isometry was not necessarily expected given that the olfactory epithelium covers more than just the turbinate. These data illuminate a number of evolutionary hypotheses that, moving forward, should inform tradeoffs and constraints between the olfactory and neighboring systems in the avian head.


Subject(s)
Nasal Cavity/anatomy & histology , Olfactory Bulb/anatomy & histology , Turbinates/anatomy & histology , Animals , Chick Embryo , Chickens , Nasal Cavity/embryology , Nasal Cavity/growth & development , Olfactory Bulb/embryology , Olfactory Bulb/growth & development , Olfactory Mucosa/anatomy & histology , Olfactory Mucosa/embryology , Olfactory Mucosa/growth & development , Turbinates/embryology , Turbinates/growth & development
3.
J Comp Neurol ; 527(14): 2233-2244, 2019 10 01.
Article in English | MEDLINE | ID: mdl-30864157

ABSTRACT

Olfactory sensory neurons (OSNs) located in the dorsomedial and ventromedial regions of the olfactory epithelium (OE) are distinguished from one another based on their molecular expression patterns. This difference is reflected in the separation of the glomerular layer of the olfactory bulb (OB) into dorsomedial and ventrolateral regions. However, it is unclear whether a complementary separation is also evident in the projection neurons that innervate the OB glomeruli. In this study, we compared the development of the OB between different regions by focusing on the transcription factor, Tbx21, which is expressed by mitral and tufted cells in the mature OB. Examining the OB at different developmental ages, we found that Tbx21 expression commenced in the anteromedial region called the tongue-shaped area, followed by the dorsomedial and then ventrolateral areas. We also showed that the tongue-shaped area was innervated by the OSNs located in the most dorsomedial part of the ventrolateral OE, the V-zone:DM. Interestingly, the generation of OSNs occurred first in the dorsomedial zone including the V-zone:DM, suggesting a correlation between the time course of OSN generation in the OE and Tbx21 expression in their target region of the OB. In contrast, expression of vGluT1, which is also found in all mitral cells in the mature OB, was first detected in the ventrolateral region during development. Our findings demonstrate that the development of projection neurons occurs in a compartmentalized manner in the OB; tongue-shaped, dorsomedial, and ventrolateral areas, and that not all projection neurons follow the same developmental pathway.


Subject(s)
Cell Differentiation/physiology , Neurogenesis/physiology , Olfactory Bulb/cytology , Olfactory Bulb/growth & development , Olfactory Mucosa/cytology , Olfactory Mucosa/growth & development , Animals , Animals, Newborn , Female , Glucose Transporter Type 1/physiology , Mice , Mice, Inbred CBA , Mice, Transgenic , Olfactory Bulb/embryology , Olfactory Mucosa/embryology , Olfactory Receptor Neurons/physiology , Pregnancy
4.
Dev Cell ; 46(5): 564-580.e5, 2018 09 10.
Article in English | MEDLINE | ID: mdl-30100263

ABSTRACT

The olfactory epithelium (OE) is a neurosensory organ required for the sense of smell. Turbinates, bony projections from the nasal cavity wall, increase the surface area within the nasal cavity lined by the OE. Here, we use engineered fibroblast growth factor 20 (Fgf20) knockin alleles to identify a population of OE progenitor cells that expand horizontally during development to populate all lineages of the mature OE. We show that these Fgf20-positive epithelium-spanning progenitor (FEP) cells are responsive to Wnt/ß-Catenin signaling. Wnt signaling suppresses FEP cell differentiation into OE basal progenitors and their progeny and positively regulates Fgf20 expression. We further show that FGF20 signals to the underlying mesenchyme to regulate the growth of turbinates. These studies thus identify a population of OE progenitor cells that function to scale OE surface area with the underlying turbinates.


Subject(s)
Fibroblast Growth Factors/physiology , Mesoderm/cytology , Olfactory Mucosa/physiology , Stem Cells/physiology , Turbinates/growth & development , Wnt Signaling Pathway , Animals , Cell Differentiation , Cells, Cultured , Female , Male , Mesoderm/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Olfactory Mucosa/cytology , Olfactory Mucosa/embryology , Stem Cells/cytology
5.
Methods Mol Biol ; 1817: 197-207, 2018.
Article in English | MEDLINE | ID: mdl-29959715

ABSTRACT

The culture of the olfactory epithelium has been a useful model for the study of neurogenesis since olfactory neurons regenerate continuously throughout the adult lifespan. Structurally and functionally mature olfactory neurons are generated in vitro from non-neuronal precursors in a process that resembles the in vivo counterparts. This chapter describes a technique for culture of olfactory neurons from the avian olfactory epithelium of embryonic chickens; this enables the controlled laboratory study of a critical sensory system that is unstudied in this major vertebrate class. The techniques described here are broadly applicable to other endothermic vertebrate species.


Subject(s)
Neurons/cytology , Olfactory Mucosa/embryology , Primary Cell Culture/methods , Animals , Cell Differentiation , Cells, Cultured , Chickens , Neurogenesis , Olfactory Mucosa/cytology
6.
Dev Biol ; 441(2): 242-251, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29709597

ABSTRACT

The fish Astyanax mexicanus comes in two forms: the normal surface-dwelling (SF) and the blind depigmented cave-adapted (CF) morphs. Among many phenotypic differences, cavefish show enhanced olfactory sensitivity to detect amino-acid odors and they possess large olfactory sensory organs. Here, we questioned the relationship between the size of the olfactory organ and olfactory capacities. Comparing olfactory detection abilities of CF, SF and F1 hybrids with various olfactory epithelium (OE) sizes in behavioral tests, we concluded that OE size is not the only factor involved. Other possibilities were envisaged. First, olfactory behavior was tested in SF raised in the dark or after embryonic lens ablation, which leads to eye degeneration and mimics the CF condition. Both absence of visual function and absence of visual organs improved the SF olfactory detection capacities, without affecting the size of their OE. This suggested that developmental plasticity occurs between the visual and the olfactory modalities, and can be recruited in SF after visual deprivation. Second, the development of the olfactory epithelium was compared in SF and CF in their first month of life. Proliferation, cell death, neuronal lifespan, and olfactory progenitor cell cycling properties were identical in the two morphs. By contrast, the proportions of the three main olfactory sensory neurons subtypes (ciliated, microvillous and crypt) in their OE differed. OMP-positive ciliated neurons were more represented in SF, TRPC2-positive microvillous neurons were proportionately more abundant in CF, and S100-positive crypt cells were found in equal densities in the two morphs. Thus, general proliferative properties of olfactory progenitors are identical but neurogenic properties differ and lead to variations in the neuronal composition of the OE in SF and CF. Together, these experiments suggest that there are at least two components in the evolution of cavefish olfactory skills: (1) one part of eye-dependent developmental phenotypic plasticity, which does not depend on the size of the olfactory organ, and (2) one part of developmental evolution of the OE, which may stem from embryonic specification of olfactory neurons progenitor pools.


Subject(s)
Behavior, Animal/physiology , Characiformes/embryology , Neural Stem Cells/metabolism , Olfactory Mucosa/embryology , Olfactory Perception/physiology , Smell/physiology , Animals , Cell Death/physiology , Cell Proliferation/physiology , Neural Stem Cells/cytology , Olfactory Mucosa/cytology
7.
PLoS Comput Biol ; 14(4): e1006128, 2018 04.
Article in English | MEDLINE | ID: mdl-29672531

ABSTRACT

State-of-the-art light-sheet and confocal microscopes allow recording of entire embryos in 3D and over time (3D+t) for many hours. Fluorescently labeled structures can be segmented and tracked automatically in these terabyte-scale 3D+t images, resulting in thousands of cell migration trajectories that provide detailed insights to large-scale tissue reorganization at the cellular level. Here we present EmbryoMiner, a new interactive open-source framework suitable for in-depth analyses and comparisons of entire embryos, including an extensive set of trajectory features. Starting at the whole-embryo level, the framework can be used to iteratively focus on a region of interest within the embryo, to investigate and test specific trajectory-based hypotheses and to extract quantitative features from the isolated trajectories. Thus, the new framework provides a valuable new way to quantitatively compare corresponding anatomical regions in different embryos that were manually selected based on biological prior knowledge. As a proof of concept, we analyzed 3D+t light-sheet microscopy images of zebrafish embryos, showcasing potential user applications that can be performed using the new framework.


Subject(s)
Cell Tracking/statistics & numerical data , Zebrafish/embryology , Animals , Animals, Genetically Modified , Cell Movement , Computational Biology , Embryonic Development , Embryonic Stem Cells/cytology , Gastrulation , Germ Layers/cytology , Imaging, Three-Dimensional , Microscopy, Fluorescence , Olfactory Mucosa/cytology , Olfactory Mucosa/embryology , Software
8.
Mol Neurobiol ; 55(11): 8306-8327, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29532253

ABSTRACT

The postnatal mammalian olfactory epithelium (OE) represents a major aspect of the peripheral olfactory system. It is a pseudostratified tissue that originates from the olfactory placode and is composed of diverse cells, some of which are specialized receptor neurons capable of transducing odorant stimuli to afford the perception of smell (olfaction). The OE is known to offer a tractable miniature model for studying the systematic generation of neurons and glia that typify neural tissue development. During OE development, stem/progenitor cells that will become olfactory sensory neurons and/or non-neuronal cell types display fine spatiotemporal expression of neuronal and non-neuronal genes that ensures their proper proliferation, differentiation, survival, and regeneration. Many factors, including transcription and epigenetic factors, have been identified as key regulators of the expression of such requisite genes to permit normal OE morphogenesis. Typically, specific interactive regulatory networks established between transcription and epigenetic factors/cofactors orchestrate histogenesis in the embryonic and adult OE. Hence, investigation of these regulatory networks critical for OE development promises to disclose strategies that may be employed in manipulating the stepwise transition of olfactory precursor cells to become fully differentiated and functional neuronal and non-neuronal cell types. Such strategies potentially offer formidable means of replacing injured or degenerated neural cells as therapeutics for nervous system perturbations. This review recapitulates the developmental cellular diversity of the olfactory neuroepithelium and discusses findings on how the precise and cooperative molecular control by transcriptional and epigenetic machinery is indispensable for OE ontogeny.


Subject(s)
Mammals/genetics , Olfactory Mucosa/embryology , Olfactory Mucosa/metabolism , Animals , Chromatin Assembly and Disassembly , Epigenesis, Genetic , Olfactory Mucosa/cytology , Transcription, Genetic
9.
Development ; 145(2)2018 01 19.
Article in English | MEDLINE | ID: mdl-29352015

ABSTRACT

The transcription factor Sox2 is necessary to maintain pluripotency of embryonic stem cells, and to regulate neural development. Neurogenesis in the vertebrate olfactory epithelium persists from embryonic stages through adulthood. The role Sox2 plays for the development of the olfactory epithelium and neurogenesis within has, however, not been determined. Here, by analysing Sox2 conditional knockout mouse embryos and chick embryos deprived of Sox2 in the olfactory epithelium using CRISPR-Cas9, we show that Sox2 activity is crucial for the induction of the neural progenitor gene Hes5 and for subsequent differentiation of the neuronal lineage. Our results also suggest that Sox2 activity promotes the neurogenic domain in the nasal epithelium by restricting Bmp4 expression. The Sox2-deficient olfactory epithelium displays diminished cell cycle progression and proliferation, a dramatic increase in apoptosis and finally olfactory pit atrophy. Moreover, chromatin immunoprecipitation data show that Sox2 directly binds to the Hes5 promoter in both the PNS and CNS. Taken together, our results indicate that Sox2 is essential to establish, maintain and expand the neuronal progenitor pool by suppressing Bmp4 and upregulating Hes5 expression.


Subject(s)
Avian Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Bone Morphogenetic Proteins/metabolism , Neurogenesis/physiology , Olfactory Mucosa/embryology , Olfactory Mucosa/metabolism , Repressor Proteins/genetics , SOXB1 Transcription Factors/metabolism , Animals , Apoptosis , Avian Proteins/deficiency , Avian Proteins/genetics , Base Sequence , Binding Sites/genetics , Bone Morphogenetic Protein 4/metabolism , Cell Cycle , Cell Lineage , Cell Proliferation , Chick Embryo , Female , Gene Knockout Techniques , Mice , Mice, Knockout , Neurogenesis/genetics , Pregnancy , Promoter Regions, Genetic , SOXB1 Transcription Factors/deficiency , SOXB1 Transcription Factors/genetics , Up-Regulation
10.
Elife ; 72018 01 02.
Article in English | MEDLINE | ID: mdl-29292696

ABSTRACT

The zebrafish olfactory epithelium comprises a variety of neuronal populations, which are thought to have distinct embryonic origins. For instance, while ciliated sensory neurons arise from preplacodal ectoderm (PPE), previous lineage tracing studies suggest that both Gonadotropin releasing hormone 3 (Gnrh3) and microvillous sensory neurons derive from cranial neural crest (CNC). We find that the expression of Islet1/2 is restricted to Gnrh3 neurons associated with the olfactory epithelium. Unexpectedly, however, we find no change in Islet1/2+ cell numbers in sox10 mutant embryos, calling into question their CNC origin. Lineage reconstruction based on backtracking in time-lapse confocal datasets, and confirmed by photoconversion experiments, reveals that Gnrh3 neurons derive from the anterior PPE. Similarly, all of the microvillous sensory neurons we have traced arise from preplacodal progenitors. Our results suggest that rather than originating from separate ectodermal populations, cell-type heterogeneity is generated from overlapping pools of progenitors within the preplacodal ectoderm.


Subject(s)
Cell Lineage , Ectoderm/embryology , Neurons/physiology , Olfactory Mucosa/embryology , Zebrafish/embryology , Animals , Microscopy, Confocal , Time-Lapse Imaging
11.
Development ; 144(19): 3521-3532, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28974641

ABSTRACT

Placodes are discrete thickenings of the vertebrate cranial ectoderm that generate morpho-functionally distinct structures, such as the adenohypophysis, olfactory epithelium and lens. All placodes arise from a horseshoe-shaped preplacodal ectoderm in which the precursors of individual placodes are intermingled. However, fate-map studies indicated that cells positioned at the preplacodal midline give rise to only the adenohypophyseal placode, suggesting a unique organization of these precursors within the preplacode. To test this possibility, we combined embryological and molecular approaches in chick embryos to show that, at gastrula stage, adenohypophyseal precursors are clustered in the median preplacodal ectoderm, largely segregated from those of the adjacent olfactory placode. Median precursors are elongated, densely packed and, at neurula stage, express a molecular signature that distinguishes them from the remaining preplacodal cells. Olfactory placode precursors and midline neural cells can replace ablated adenohypophyseal precursors up to head-fold stage, although with a more plastic organization. We thus propose that adenohypophyseal placode precursors are unique within the preplacodal ectoderm possibly because they originate the only single placode and the only one with an endocrine character.


Subject(s)
Ectoderm/embryology , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/embryology , Stem Cells/cytology , Animals , Body Patterning , Cell Shape , Chick Embryo , Ectoderm/cytology , Lens, Crystalline/cytology , Lens, Crystalline/embryology , Olfactory Mucosa/cytology , Olfactory Mucosa/embryology
12.
Sci Rep ; 7(1): 8804, 2017 08 18.
Article in English | MEDLINE | ID: mdl-28821769

ABSTRACT

Organisms have evolved strikingly parallel phenotypes in response to similar selection pressures suggesting that there may be shared constraints limiting the possible evolutionary trajectories. For example, the behavioral adaptation of specialist Drosophila species to specific host plants can exhibit parallel changes in their adult olfactory neuroanatomy. We investigated the genetic basis of these parallel changes by comparing gene expression during the development of the olfactory system of two specialist Drosophila species to that of four other generalist species. Our results suggest that the parallelism observed in the adult olfactory neuroanatomy of ecological specialists extends more broadly to their developmental antennal expression profiles, and to the transcription factor combinations specifying olfactory receptor neuron (ORN) fates. Additionally, comparing general patterns of variation for the antennal transcriptional profiles in the adult and developing olfactory system of the six species suggest the possibility that specific, non-random components of the developmental programs underlying the Drosophila olfactory system harbor a disproportionate amount of interspecies variation. Further examination of these developmental components may be able to inform a deeper understanding of how traits evolve.


Subject(s)
Arthropod Antennae/embryology , Drosophila/embryology , Drosophila/genetics , Gene Expression Regulation, Developmental , Genetic Variation , Organogenesis/genetics , Transcriptome , Animals , Olfactory Mucosa/embryology , Olfactory Receptor Neurons/metabolism , Reproducibility of Results
13.
Zoology (Jena) ; 123: 11-15, 2017 08.
Article in English | MEDLINE | ID: mdl-28550945

ABSTRACT

Animals recognize their surrounding environments through the sense of smell by detecting thousands of chemical odorants. Wild boars (Sus scrofa) completely depend on their ability to recognize chemical odorants: to detect food, during scavenging and searching partners, during breeding periods and to avoid potential predators. Wild piglets must be prepared for the chemical universe that they will enter after birth, and they show intense neuronal activity in the olfactory mucosa. With this in mind, we investigated the morpho-functional embryonic development of the olfactory mucosa in the wild boar (in five stages before birth). Using mRNA expression analysis of olfactory marker protein and neuropeptide Y, involved in the function of olfactory sensory neurons, we show early activation of the appropriate genes in the wild boar. We hypothesize olfactory pre-birth development in wild boar is highly adaptive.


Subject(s)
Olfactory Mucosa/embryology , Smell/physiology , Sus scrofa/embryology , Animals , Female , Fetus/physiology , Gene Expression Regulation, Developmental/physiology , Olfactory Mucosa/metabolism , Pregnancy , Sus scrofa/physiology
14.
Elife ; 62017 04 19.
Article in English | MEDLINE | ID: mdl-28422009

ABSTRACT

While axon fasciculation plays a key role in the development of neural networks, very little is known about its dynamics and the underlying biophysical mechanisms. In a model system composed of neurons grown ex vivo from explants of embryonic mouse olfactory epithelia, we observed that axons dynamically interact with each other through their shafts, leading to zippering and unzippering behavior that regulates their fasciculation. Taking advantage of this new preparation suitable for studying such interactions, we carried out a detailed biophysical analysis of zippering, occurring either spontaneously or induced by micromanipulations and pharmacological treatments. We show that zippering arises from the competition of axon-axon adhesion and mechanical tension in the axons, and provide the first quantification of the force of axon-axon adhesion. Furthermore, we introduce a biophysical model of the zippering dynamics, and we quantitatively relate the individual zipper properties to global characteristics of the developing axon network. Our study uncovers a new role of mechanical tension in neural development: the regulation of axon fasciculation.


Subject(s)
Axon Fasciculation , Axons/physiology , Biophysical Phenomena , Animals , Cell Adhesion , Cells, Cultured , Mice , Models, Biological , Olfactory Mucosa/embryology , Stress, Mechanical
15.
Neural Dev ; 12(1): 2, 2017 Feb 13.
Article in English | MEDLINE | ID: mdl-28193234

ABSTRACT

BACKGROUND: The mammalian primary olfactory system has a spatially-ordered projection in which olfactory sensory neurons (OSNs) located in the dorsomedial (DM) and ventrolateral (VL) region of the olfactory epithelium (OE) send their axons to the dorsal and ventral region of the olfactory bulb (OB), respectively. We previously found that OSN axonal projections occur sequentially, from the DM to the VL region of the OE. The differential timing of axonal projections is important for olfactory map formation because early-arriving OSN axons secrete guidance cues at the OB to help navigate late-arriving OSN axons. We hypothesized that the differential timing of axonal projections is regulated by the timing of OSN neurogenesis. To test this idea, we investigated spatiotemporal patterns of OSN neurogenesis during olfactory development. METHODS AND RESULTS: To determine the time of OSN origin, we used two thymidine analogs, BrdU and EdU, which can be incorporated into cells in the S-phase of the cell-cycle. We injected these two analogs at different developmental time points and analyzed distribution patterns of labeled OSNs. We found that OSNs with different dates of origin were differentially distributed in the OE. The majority of OSNs generated at the early stage of development were located in the DM region of the OE, whereas OSNs generated at the later stage of development were preferentially located in the VL region of the OE. CONCLUSIONS: These results indicate that the number of OSNs is sequentially increased from the DM to the VL axis of the OE. Moreover, the temporal sequence of OSN proliferation correlates with that of axonal extension and emergence of glomerular structures in the OB. Thus, we propose that the timing of OSN neurogenesis regulates that of OSN axonal projection and thereby helps preserve the topographic order of the olfactory glomerular map along the dorsal-ventral axis of the OB.


Subject(s)
Axons/physiology , Neurogenesis , Olfactory Bulb/embryology , Olfactory Receptor Neurons/physiology , Animals , Axon Guidance , Mice , Olfactory Mucosa/embryology
16.
Mol Cell Neurosci ; 80: 75-88, 2017 04.
Article in English | MEDLINE | ID: mdl-28188885

ABSTRACT

The Ca2+-activated monovalent cation channel Trpm5 is a key element in chemotransduction of taste receptor cells of the tongue, but the extent to which Trpm5 channels are expressed in olfactory sensory neurons (OSNs) of the main olfactory epithelium (MOE) of adult mice as part of a specific pheromonal detection system is debated. Here, we used a novel Trpm5-IRES-Cre knockin strain to drive Cre recombinase expression, employed previously validated Trpm5 antibodies, performed in situ hybridization experiments to localize Trpm5 RNA, and searched extensively for Trpm5 splice variants in genetically-labeled, Trpm5-expressing MOE cells. In contrast to previous reports, we find no evidence for the existence in adult mouse OSNs of the classical Trpm5 channel known from taste cells. We show that Trpm5-expressing adult OSNs express a novel Trpm5 splice variant, Trpm5-9, that is unlikely to form a functional cation channel by itself. We also demonstrate that Trpm5 is transiently expressed in a subpopulation of mature OSNs in the embryonic olfactory epithelium, indicating that Trpm5 channels could play a specific role in utero during a narrow developmental time window. Ca2+ imaging with GCaMP3 under the control of the Trpm5-IRES-Cre allele using a newly developed MOE wholemount preparation of the adult olfactory epithelium reveals that Trpm5-GCaMP3 OSNs comprise a heterogeneous group of sensory neurons many of which can detect general odorants. Together, these studies are essential for understanding the role of transient receptor potential channels in mammalian olfaction.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Olfactory Mucosa/metabolism , TRPM Cation Channels/metabolism , Age Factors , Animals , Animals, Newborn , Calcium/metabolism , Embryo, Mammalian , GAP-43 Protein/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Olfactory Marker Protein/genetics , Olfactory Marker Protein/metabolism , Olfactory Mucosa/cytology , Olfactory Mucosa/embryology , Olfactory Mucosa/growth & development , Olfactory Receptor Neurons/metabolism , RNA, Messenger/metabolism , TRPM Cation Channels/genetics , Vomeronasal Organ/embryology , Vomeronasal Organ/growth & development , Vomeronasal Organ/metabolism
17.
Sci Rep ; 6: 38199, 2016 12 02.
Article in English | MEDLINE | ID: mdl-27910949

ABSTRACT

The olfactory epithelium (OE) is composed of olfactory sensory neurons (OSNs), sustentacular supporting cells, and several types of non-neuronal cells. Stem and progenitor cells are located basally, and are the source of all cell types needed to maintain OE homeostasis. Here, we report that Ascl3, a basic helix-loop-helix transcription factor, is expressed in the developing OE. Lineage tracing experiments demonstrate that the non-neuronal microvillar cells and Bowman's glands are exclusively derived from Ascl3+ progenitor cells in the OE during development. Following chemically-induced injury, Ascl3 expression is activated in a subset of horizontal basal cells (HBCs), which repopulate all microvillar cells and Bowman's glands during OE regeneration. After ablation of Ascl3-expressing cells, the OE can regenerate, but lacks the non-neuronal microvillar and Bowman's gland support cells. These results demonstrate that Ascl3 marks progenitors that are lineage-committed strictly to microvillar cells and Bowman's glands, and highlight the requirement for these cell types to support OE homeostasis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Lineage/physiology , Olfactory Mucosa/embryology , Stem Cells/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Mice , Mice, Transgenic , Olfactory Mucosa/cytology , Stem Cells/cytology
18.
PLoS Genet ; 12(9): e1006274, 2016 09.
Article in English | MEDLINE | ID: mdl-27611684

ABSTRACT

Neurogenesis is a key developmental event through which neurons are generated from neural stem/progenitor cells. Chromatin remodeling BAF (mSWI/SNF) complexes have been reported to play essential roles in the neurogenesis of the central nervous system. However, whether BAF complexes are required for neuron generation in the olfactory system is unknown. Here, we identified onscBAF and ornBAF complexes, which are specifically present in olfactory neural stem cells (oNSCs) and olfactory receptor neurons (ORNs), respectively. We demonstrated that BAF155 subunit is highly expressed in both oNSCs and ORNs, whereas high expression of BAF170 subunit is observed only in ORNs. We report that conditional deletion of BAF155, a core subunit in both onscBAF and ornBAF complexes, causes impaired proliferation of oNSCs as well as defective maturation and axonogenesis of ORNs in the developing olfactory epithelium (OE), while the high expression of BAF170 is important for maturation of ORNs. Interestingly, in the absence of BAF complexes in BAF155/BAF170 double-conditional knockout mice (dcKO), OE is not specified. Mechanistically, BAF complex is required for normal activation of Pax6-dependent transcriptional activity in stem cells/progenitors of the OE. Our findings unveil a novel mechanism mediated by the mSWI/SNF complex in OE neurogenesis and development.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Neurogenesis , Olfactory Mucosa/metabolism , Transcription Factors/genetics , Animals , Cells, Cultured , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins , Gene Deletion , Mice , Mice, Inbred C57BL , Olfactory Mucosa/cytology , Olfactory Mucosa/embryology , Olfactory Receptor Neurons/cytology , Olfactory Receptor Neurons/metabolism , Transcription Factors/metabolism
19.
Surg Radiol Anat ; 38(10): 1161-1168, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27142661

ABSTRACT

PURPOSE: Evo-devo is the science that studies the link between evolution of species and embryological development. This concept helps to understand the complex anatomy of the human nose. The evo-devo theory suggests the persistence in the adult of an anatomical entity, the olfactory fascia, that unites the cartilages of the nose to the olfactory mucosa. METHODS: We dissected two fresh specimens. After resecting the superficial tissues of the nose, dissection was focused on the disarticulation of the fibrocartilaginous noses from the facial and skull base skeleton. RESULTS: Dissection shows two fibrocartilaginous sacs that were invaginated side-by-side in the midface and attached to the anterior skull base. These membranous sacs were separated in the midline by the perpendicular plate of the ethmoid. Their walls contained the alar cartilages and the lateral expansions of the septolateral cartilage, which we had to separate from the septal cartilage. The olfactory mucosa was located inside their cranial ends. CONCLUSION: The olfactory fascia is a continuous membrane uniting the nasal cartilages to the olfactory mucosa. Its origin can be found in the invagination and differentiation processes of the olfactory placodes. The fibrous portions of the olfactory fascia may be described as ligaments that unit the different components of the olfactory fascia one to the other and the fibrocartilaginous nose to the facial and skull base skeleton. The basicranial ligaments, fixing the fibrocartilaginous nose to the skull base, represent key elements in the concept of septorhinoplasty by disarticulation.


Subject(s)
Fascia/anatomy & histology , Nasal Cartilages/anatomy & histology , Olfactory Mucosa/anatomy & histology , Rhinoplasty/methods , Adult , Biological Evolution , Cadaver , Developmental Biology , Dissection , Ethmoid Bone/anatomy & histology , Fascia/embryology , Humans , Nasal Cartilages/embryology , Olfactory Mucosa/embryology
20.
Development ; 143(9): 1534-46, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27143755

ABSTRACT

Cellular interactions are key for the differentiation of distinct cell types within developing epithelia, yet the molecular mechanisms engaged in these interactions remain poorly understood. In the developing olfactory epithelium (OE), neural stem/progenitor cells give rise to odorant-detecting olfactory receptor neurons (ORNs) and glial-like sustentacular (SUS) cells. Here, we show in mice that the transmembrane receptor neogenin (NEO1) and its membrane-bound ligand RGMB control the balance of neurons and glial cells produced in the OE. In this layered epithelium, neogenin is expressed in progenitor cells, while RGMB is restricted to adjacent newly born ORNs. Ablation of Rgmb via gene-targeting increases the number of dividing progenitor cells in the OE and leads to supernumerary SUS cells. Neogenin loss-of-function phenocopies these effects observed in Rgmb(-/-) mice, supporting the proposal that RGMB-neogenin signaling regulates progenitor cell numbers and SUS cell production. Interestingly, Neo1(-/-) mice also exhibit increased apoptosis of ORNs, implicating additional ligands in the neogenin-dependent survival of ORNs. Thus, our results indicate that RGMB-neogenin-mediated cell-cell interactions between newly born neurons and progenitor cells control the ratio of glia and neurons produced in the OE.


Subject(s)
Gene Expression Regulation, Developmental , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Neural Stem Cells/cytology , Neurogenesis/genetics , Olfactory Mucosa/embryology , Olfactory Receptor Neurons/cytology , Animals , Apoptosis/genetics , Cell Adhesion Molecules, Neuronal , Cell Proliferation/genetics , GPI-Linked Proteins , Membrane Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/biosynthesis , Neurogenesis/physiology , Neuroglia/cytology , Olfactory Mucosa/cytology , Olfactory Receptor Neurons/metabolism , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL