Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 516
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Development ; 149(5)2022 03 01.
Article in English | MEDLINE | ID: mdl-35132995

ABSTRACT

Distinct neural stem cells (NSCs) reside in different regions of the subventricular zone (SVZ) and generate multiple olfactory bulb (OB) interneuron subtypes in the adult brain. However, the molecular mechanisms underlying such NSC heterogeneity remain largely unknown. Here, we show that the basic helix-loop-helix transcription factor Olig2 defines a subset of NSCs in the early postnatal and adult SVZ. Olig2-expressing NSCs exist broadly but are most enriched in the ventral SVZ along the dorsoventral axis complementary to dorsally enriched Gsx2-expressing NSCs. Comparisons of Olig2-expressing NSCs from early embryonic to adult stages using single cell transcriptomics reveal stepwise developmental changes in their cell cycle and metabolic properties. Genetic studies further show that cross-repression contributes to the mutually exclusive expression of Olig2 and Gsx2 in NSCs/progenitors during embryogenesis, but that their expression is regulated independently from each other in adult NSCs. Finally, lineage-tracing and conditional inactivation studies demonstrate that Olig2 plays an important role in the specification of OB interneuron subtypes. Altogether, our study demonstrates that Olig2 defines a unique subset of adult NSCs enriched in the ventral aspect of the adult SVZ.


Subject(s)
Interneurons/metabolism , Lateral Ventricles/growth & development , Lateral Ventricles/metabolism , Neural Stem Cells/metabolism , Olfactory Bulb/growth & development , Olfactory Bulb/metabolism , Oligodendrocyte Transcription Factor 2/metabolism , Animals , Cell Cycle/genetics , Cell Lineage/genetics , Cells, Cultured , Female , Gene Knockout Techniques , Lateral Ventricles/embryology , Male , Mice , Mice, Knockout , Neurogenesis/genetics , Olfactory Bulb/embryology , Oligodendrocyte Transcription Factor 2/genetics , Signal Transduction/genetics , Transcriptome/genetics
2.
Development ; 149(1)2022 01 01.
Article in English | MEDLINE | ID: mdl-35023540

ABSTRACT

Estrogens are well-known to regulate development of sexual dimorphism of the brain; however, their role in embryonic brain development prior to sex-differentiation is unclear. Using estrogen biosensor zebrafish models, we found that estrogen activity in the embryonic brain occurs from early neurogenesis specifically in a type of glia in the olfactory bulb (OB), which we name estrogen-responsive olfactory bulb (EROB) cells. In response to estrogen, EROB cells overlay the outermost layer of the OB and interact tightly with olfactory sensory neurons at the olfactory glomeruli. Inhibiting estrogen activity using an estrogen receptor antagonist, ICI182,780 (ICI), and/or EROB cell ablation impedes olfactory glomerular development, including the topological organisation of olfactory glomeruli and inhibitory synaptogenesis in the OB. Furthermore, activation of estrogen signalling inhibits both intrinsic and olfaction-dependent neuronal activity in the OB, whereas ICI or EROB cell ablation results in the opposite effect on neuronal excitability. Altering the estrogen signalling disrupts olfaction-mediated behaviour in later larval stage. We propose that estrogens act on glia to regulate development of OB circuits, thereby modulating the local excitability in the OB and olfaction-mediated behaviour.


Subject(s)
Estrogens/metabolism , Neurogenesis , Neuroglia/cytology , Olfactory Bulb/embryology , Animals , Estrogen Receptor Antagonists/pharmacology , Fulvestrant/pharmacology , Neuroglia/drug effects , Neuroglia/metabolism , Olfactory Bulb/cytology , Olfactory Bulb/drug effects , Olfactory Receptor Neurons/cytology , Olfactory Receptor Neurons/metabolism , Receptors, Estrogen/antagonists & inhibitors , Synapses/metabolism , Synapses/physiology , Zebrafish
3.
Development ; 147(10)2020 05 26.
Article in English | MEDLINE | ID: mdl-32253238

ABSTRACT

The transcription factor Zeb2 controls fate specification and subsequent differentiation and maturation of multiple cell types in various embryonic tissues. It binds many protein partners, including activated Smad proteins and the NuRD co-repressor complex. How Zeb2 subdomains support cell differentiation in various contexts has remained elusive. Here, we studied the role of Zeb2 and its domains in neurogenesis and neural differentiation in the young postnatal ventricular-subventricular zone (V-SVZ), in which neural stem cells generate olfactory bulb-destined interneurons. Conditional Zeb2 knockouts and separate acute loss- and gain-of-function approaches indicated that Zeb2 is essential for controlling apoptosis and neuronal differentiation of V-SVZ progenitors before and after birth, and we identified Sox6 as a potential downstream target gene of Zeb2. Zeb2 genetic inactivation impaired the differentiation potential of the V-SVZ niche in a cell-autonomous fashion. We also provide evidence that its normal function in the V-SVZ also involves non-autonomous mechanisms. Additionally, we demonstrate distinct roles for Zeb2 protein-binding domains, suggesting that Zeb2 partners co-determine neuronal output from the mouse V-SVZ in both quantitative and qualitative ways in early postnatal life.


Subject(s)
Lateral Ventricles/embryology , Lateral Ventricles/growth & development , Neurogenesis/genetics , Olfactory Bulb/embryology , Olfactory Bulb/growth & development , Zinc Finger E-box Binding Homeobox 2/metabolism , Animals , Apoptosis/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Gene Knockout Techniques , Interneurons/metabolism , Lateral Ventricles/metabolism , Mice , Mice, Knockout , Neural Stem Cells/metabolism , Olfactory Bulb/metabolism , SOXD Transcription Factors/metabolism , Signal Transduction/immunology , Zinc Finger E-box Binding Homeobox 2/genetics
4.
Development ; 146(4)2019 02 18.
Article in English | MEDLINE | ID: mdl-30777863

ABSTRACT

In the adult rodent brain, neural stem cells (NSCs) persist in the ventricular-subventricular zone (V-SVZ) and the subgranular zone (SGZ), which are specialized niches in which young neurons for the olfactory bulb (OB) and hippocampus, respectively, are generated. Recent studies have significantly modified earlier views on the mechanisms of NSC self-renewal and neurogenesis in the adult brain. Here, we discuss the molecular control, heterogeneity, regional specification and cell division modes of V-SVZ NSCs, and draw comparisons with NSCs in the SGZ. We highlight how V-SVZ NSCs are regulated by local signals from their immediate neighbors, as well as by neurotransmitters and factors that are secreted by distant neurons, the choroid plexus and vasculature. We also review recent advances in single cell RNA analyses that reveal the complexity of adult neurogenesis. These findings set the stage for a better understanding of adult neurogenesis, a process that one day may inspire new approaches to brain repair.


Subject(s)
Adult Stem Cells/physiology , Hippocampus/physiology , Lateral Ventricles/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Olfactory Bulb/physiology , Animals , Cell Communication , Cell Differentiation , Cell Lineage , Embryonic Stem Cells/physiology , Hippocampus/embryology , Humans , Interneurons/physiology , Lateral Ventricles/embryology , Mice , Neurons/physiology , Olfactory Bulb/embryology , Sequence Analysis, RNA , Signal Transduction , Single-Cell Analysis , Transcriptome
5.
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
6.
Cereb Cortex ; 29(11): 4831-4849, 2019 12 17.
Article in English | MEDLINE | ID: mdl-30796806

ABSTRACT

Generation of olfactory bulb (OB) interneurons requires neural stem/progenitor cell specification, proliferation, differentiation, and young interneuron migration and maturation. Here, we show that the homeobox transcription factors Dlx1/2 are central and essential components in the transcriptional code for generating OB interneurons. In Dlx1/2 constitutive null mutants, the differentiation of GSX2+ and ASCL1+ neural stem/progenitor cells in the dorsal lateral ganglionic eminence is blocked, resulting in a failure of OB interneuron generation. In Dlx1/2 conditional mutants (hGFAP-Cre; Dlx1/2F/- mice), GSX2+ and ASCL1+ neural stem/progenitor cells in the postnatal subventricular zone also fail to differentiate into OB interneurons. In contrast, overexpression of Dlx1&2 in embryonic mouse cortex led to ectopic production of OB-like interneurons that expressed Gad1, Sp8, Sp9, Arx, Pbx3, Etv1, Tshz1, and Prokr2. Pax6 mutants generate cortical ectopia with OB-like interneurons, but do not do so in compound Pax6; Dlx1/2 mutants. We propose that DLX1/2 promote OB interneuron development mainly through activating the expression of Sp8/9, which further promote Tshz1 and Prokr2 expression. Based on this study, in combination with earlier ones, we propose a transcriptional network for the process of OB interneuron development.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Interneurons/metabolism , Neural Stem Cells/metabolism , Olfactory Bulb/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation , Female , Male , Mice, Inbred C57BL , Mice, Transgenic , Neocortex/embryology , Neocortex/metabolism , Olfactory Bulb/embryology
7.
Development ; 143(20): 3817-3825, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27578798

ABSTRACT

Building the topographic map in the mammalian olfactory bulb is explained by a model based on two axes along which sensory neurons are guided: one dorsoventral and one anteroposterior. This latter axis relies on specific expression levels of Nrp1. To evaluate the role of this receptor in this process, we used an in vivo genetic approach to decrease or suppress Nrp1 in specific neuronal populations and at different time points during axonal targeting. We observed, in neurons that express the M71 or M72 odorant receptors, that Nrp1 inactivation leads to two distinct wiring alterations, depending on the time at which Nrp1 expression is altered: first, a surprising dorsal shift of the M71 and M72 glomeruli, which often fuse with their contralateral counterparts, and second the formation of anteriorized glomeruli. The two phenotypes are partly recapitulated in mice lacking the Nrp1 ligand Sema3A and in mice whose sensory neurons express an Nrp1 mutant unable to bind Sema3A. Using a mosaic conditional approach, we show that M71 axonal fibers can bypass the Nrp1 signals that define their target area, since they are hijacked and coalesce with Nrp1-deficient M71-expressing axons that target elsewhere. Together, these findings show drastically different axonal targeting outcomes dependent on the timing at which Nrp1/Sema3A signaling is altered.


Subject(s)
Neuropilin-1/metabolism , Olfactory Bulb/cytology , Olfactory Bulb/metabolism , Animals , Axons/metabolism , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Immunohistochemistry , Mice , Neuropilin-1/genetics , Olfactory Bulb/embryology , Olfactory Receptor Neurons/cytology , Olfactory Receptor Neurons/metabolism , Semaphorin-3A/genetics , Semaphorin-3A/metabolism
8.
Part Fibre Toxicol ; 16(1): 5, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30654819

ABSTRACT

BACKGROUND: Airborne pollution, especially from diesel exhaust (DE), is known to have a negative effect on the central nervous system in exposed human populations. However, the consequences of gestational exposure to DE on the fetal brain remain poorly explored, with various effects depending on the conditions of exposure, as well as little information on early developmental stages. We investigated the short-term effects of indirect DE exposure throughout gestation on the developing brain using a rabbit model. We analyzed fetal olfactory tissues at the end of gestation and tested behaviors relevant to pups' survival at birth. Pregnant dams were exposed by nose-only inhalation to either clean air or DE with a content of particles (DEP) adjusted to 1 mg/m3 by diluting engine exhaust, for 2 h/day, 5 days/week, from gestational day 3 (GD3) to day 27 (GD27). At GD28, fetal olfactory mucosa, olfactory bulbs and whole brains were collected for anatomical and neurochemical measurements. At postnatal day 2 (PND2), pups born from another group of exposed or control female were examined for their odor-guided behavior in response to the presentation of the rabbit mammary pheromone 2-methyl-3-butyn-2-ol (2MB2). RESULTS: At GD28, nano-sized particles were observed in cilia and cytoplasm of the olfactory sensory neurons in the olfactory mucosa and in the cytoplasm of periglomerular cells in the olfactory bulbs of exposed fetuses. Moreover, cellular and axonal hypertrophies were observed throughout olfactory tissues. Concomitantly, fetal serotoninergic and dopaminergic systems were affected in the olfactory bulbs. Moreover, the neuromodulatory homeostasis was disturbed in a sex-dependent manner in olfactory tissues. At birth, the olfactory sensitivity to 2MB2 was reduced in exposed PND2 pups. CONCLUSION: Gestational exposure to DE alters olfactory tissues and affects monoaminergic neurotransmission in fetuses' olfactory bulbs, resulting in an alteration of olfactory-based behaviors at birth. Considering the anatomical and functional continuum between the olfactory system and other brain structures, and due to the importance of monoamine neurotransmission in the plasticity of neural circuits, such alterations could participate to disturbances in higher integrative structures, with possible long-term neurobehavioral consequences.


Subject(s)
Air Pollutants/toxicity , Behavior, Animal/drug effects , Fetal Development/drug effects , Olfactory Bulb/drug effects , Prenatal Exposure Delayed Effects/chemically induced , Vehicle Emissions/toxicity , Air Pollutants/pharmacokinetics , Animals , Animals, Newborn , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Female , Inhalation Exposure , Male , Olfactory Bulb/embryology , Olfactory Bulb/growth & development , Olfactory Bulb/ultrastructure , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Rabbits , Serotonergic Neurons/drug effects , Serotonergic Neurons/metabolism , Sex Factors , Synaptic Transmission/drug effects , Tissue Distribution
9.
Dev Dyn ; 247(1): 250-261, 2018 01.
Article in English | MEDLINE | ID: mdl-29106072

ABSTRACT

BACKGROUND: In vertebrates, cranial sensory placodes give rise to neurosensory and endocrine structures, such as the olfactory epithelium, inner ear, and anterior pituitary. We report here the establishment of a transgenic mouse line that expresses Cre recombinase under the control of Six1-21, a major placodal enhancer of the homeobox gene Six1. RESULTS: In the new Cre-expressing line, mSix1-21-NLSCre, the earliest Cre-mediated recombination was induced at embryonic day 8.5 in the region overlapping with the otic-epibranchial progenitor domain (OEPD), a transient, common precursor domain for the otic and epibranchial placodes. Recombination was later observed in the OEPD-derived structures (the entire inner ear and the VIIth-Xth cranial sensory ganglia), olfactory epithelium, anterior pituitary, pharyngeal ectoderm and pouches. Other Six1-positive structures, such as salivary/lacrimal glands and limb buds, were also positive for recombination. Moreover, comparison with another mouse line expressing Cre under the control of the sensory neuron enhancer, Six1-8, indicated that the continuous and complex expression pattern of Six1 during sensory organ formation is pieced together by separate enhancers. CONCLUSIONS: mSix1-21-NLSCre has several unique characteristics to make it suitable for analysis of cell lineage and gene function in sensory placodes as well as nonplacodal Six1-positive structures. Developmental Dynamics 247:250-261, 2018. © 2017 Wiley Periodicals, Inc.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Olfactory Bulb/metabolism , Sensory Receptor Cells/metabolism , Animals , Homeodomain Proteins/metabolism , Mice , Mice, Transgenic , Neural Plate/embryology , Neural Plate/metabolism , Olfactory Bulb/embryology , Olfactory Receptor Neurons/metabolism
10.
Glia ; 66(12): 2617-2631, 2018 12.
Article in English | MEDLINE | ID: mdl-30256452

ABSTRACT

We and others previously showed that in mouse embryos lacking the transcription factor Sox10, olfactory ensheathing cell (OEC) differentiation is disrupted, resulting in defective olfactory axon targeting and fewer gonadotropin-releasing hormone (GnRH) neurons entering the embryonic forebrain. The underlying mechanisms are unclear. Here, we report that OECs in the olfactory nerve layer express Frzb-encoding a secreted Wnt inhibitor with roles in axon targeting and basement membrane breakdown-from embryonic day (E)12.5, when GnRH neurons first enter the forebrain, until E16.5, the latest stage examined. The highest levels of Frzb expression are seen in OECs in the inner olfactory nerve layer, abutting the embryonic olfactory bulb. We find that Sox10 is required for Frzb expression in OECs, suggesting that loss of Frzb could explain the olfactory axon targeting and/or GnRH neuron migration defects seen in Sox10-null mice. At E16.5, Frzb-null embryos show significant reductions in both the volume of the olfactory nerve layer expressing the maturation marker Omp and the number of Omp-positive olfactory receptor neurons in the olfactory epithelium. As Omp upregulation correlates with synapse formation, this suggests that Frzb deletion indeed disrupts olfactory axon targeting. In contrast, GnRH neuron entry into the forebrain is not significantly affected. Hence, loss of Frzb may contribute to the olfactory axon targeting phenotype, but not the GnRH neuron phenotype, of Sox10-null mice. Overall, our results suggest that Frzb secreted from OECs in the olfactory nerve layer is important for olfactory axon targeting.


Subject(s)
Axons/metabolism , Gene Expression Regulation, Developmental/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Neuroglia/metabolism , Olfactory Bulb , Olfactory Receptor Neurons/pathology , Animals , Antigens, Neoplasm/metabolism , Embryo, Mammalian , Gonadotropin-Releasing Hormone/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Transgenic , Neuropeptide Y/metabolism , Olfactory Bulb/cytology , Olfactory Bulb/embryology , Olfactory Bulb/metabolism , Olfactory Marker Protein/genetics , Olfactory Marker Protein/metabolism , Olfactory Mucosa/cytology , Olfactory Mucosa/metabolism , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Tubulin/metabolism
11.
Development ; 142(24): 4318-28, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26525675

ABSTRACT

The olfactory system provides mammals with the abilities to investigate, communicate and interact with their environment. These functions are achieved through a finely organized circuit starting from the nasal cavity, passing through the olfactory bulb and ending in various cortical areas. We show that the absence of transient axonal glycoprotein-1 (Tag1)/contactin-2 (Cntn2) in mice results in a significant and selective defect in the number of the main projection neurons in the olfactory bulb, namely the mitral cells. A subpopulation of these projection neurons is reduced in Tag1-deficient mice as a result of impaired migration. We demonstrate that the detected alterations in the number of mitral cells are well correlated with diminished odor discrimination ability and social long-term memory formation. Reduced neuronal activation in the olfactory bulb and the corresponding olfactory cortex suggest that Tag1 is crucial for the olfactory circuit formation in mice. Our results underpin the significance of a numerical defect in the mitral cell layer in the processing and integration of odorant information and subsequently in animal behavior.


Subject(s)
Cell Movement , Contactin 2/deficiency , Olfactory Bulb/pathology , Olfactory Bulb/physiopathology , Animals , Cell Count , Contactin 2/metabolism , Mice, Inbred C57BL , Models, Biological , Olfactory Bulb/embryology , Olfactory Bulb/metabolism , Olfactory Receptor Neurons/metabolism , Olfactory Receptor Neurons/pathology
12.
J Neurosci ; 36(31): 8210-27, 2016 08 03.
Article in English | MEDLINE | ID: mdl-27488640

ABSTRACT

UNLABELLED: Neural circuits that undergo reorganization by newborn interneurons in the olfactory bulb (OB) are necessary for odor detection and discrimination, olfactory memory, and innate olfactory responses, including predator avoidance and sexual behaviors. The OB possesses many interneurons, including various types of granule cells (GCs); however, the contribution that each type of interneuron makes to olfactory behavioral control remains unknown. Here, we investigated the in vivo functional role of oncofetal trophoblast glycoprotein 5T4, a regulator for dendritic arborization of 5T4-expressing GCs (5T4 GCs), the level of which is reduced in the OB of 5T4 knock-out (KO) mice. Electrophysiological recordings with acute OB slices indicated that external tufted cells (ETCs) can be divided into two types, bursting and nonbursting. Optogenetic stimulation of 5T4 GCs revealed their connection to both bursting and nonbursting ETCs, as well as to mitral cells (MCs). Interestingly, nonbursting ETCs received fewer inhibitory inputs from GCs in 5T4 KO mice than from those in wild-type (WT) mice, whereas bursting ETCs and MCs received similar inputs in both mice. Furthermore, 5T4 GCs received significantly fewer excitatory inputs in 5T4 KO mice. Remarkably, in olfactory behavior tests, 5T4 KO mice had higher odor detection thresholds than the WT, as well as defects in odor discrimination learning. Therefore, the loss of 5T4 attenuates inhibitory inputs from 5T4 GCs to nonbursting ETCs and excitatory inputs to 5T4 GCs, contributing to disturbances in olfactory behavior. Our novel findings suggest that, among the various types of OB interneurons, the 5T4 GC subtype is required for odor detection and discrimination behaviors. SIGNIFICANCE STATEMENT: Neuronal circuits in the brain include glutamatergic principal neurons and GABAergic interneurons. Although the latter is a minority cell type, they are vital for normal brain function because they regulate the activity of principal neurons. If interneuron function is impaired, brain function may be damaged, leading to behavior disorder. The olfactory bulb (OB) possesses various types of interneurons, including granule cells (GCs); however, the contribution that each type of interneuron makes to the control of olfactory behavior remains unknown. Here, we analyzed electrophysiologically and behaviorally the function of oncofetal trophoblast glycoprotein 5T4, a regulator for dendritic branching in OB GCs. We found that, among the various types of OB interneuron, the 5T4 GC subtype is required for odor detection and odor discrimination behaviors.


Subject(s)
Interneurons/cytology , Interneurons/physiology , Odorants/analysis , Olfactory Bulb/cytology , Olfactory Bulb/physiology , Olfactory Perception/physiology , Animals , Behavior, Animal/physiology , Discrimination Learning/physiology , Interneurons/classification , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Olfactory Bulb/embryology
13.
Development ; 141(7): 1580-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24598163

ABSTRACT

New neurons, originating from the subventricular zone, are continuously integrating into neuronal circuitry in the olfactory bulb (OB). Using a transgenic sensor mouse, we found that adult-born OB interneurons express microRNA-125 (miR-125), whereas the pre-existing developmentally generated OB interneurons represent a unique population of cells in the adult brain, without miR-125 activity. Stable inhibition of miR-125 in newborn OB neurons resulted in enhanced dendritic morphogenesis, as well as in increased synaptic activation in response to odour sensory stimuli. These data demonstrate that miR-125 controls functional synaptic integration of adult-born OB interneurons. Our results also suggest that absence of an otherwise broadly expressed miRNA is a novel mechanism with which to achieve neuronal subtype specification.


Subject(s)
Adult Stem Cells/physiology , Embryonic Stem Cells/physiology , Interneurons/physiology , MicroRNAs/physiology , Olfactory Bulb/cytology , Animals , Animals, Newborn , Biomarkers/metabolism , Cell Differentiation/genetics , Female , Interneurons/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Stem Cells/physiology , Neurogenesis/genetics , Olfactory Bulb/embryology , Olfactory Bulb/metabolism , Synapses/genetics
14.
Development ; 141(1): 28-38, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24284204

ABSTRACT

Meis homeodomain transcription factors control cell proliferation, cell fate specification and differentiation in development and disease. Previous studies have largely focused on Meis contribution to the development of non-neuronal tissues. By contrast, Meis function in the brain is not well understood. Here, we provide evidence for a dual role of the Meis family protein Meis2 in adult olfactory bulb (OB) neurogenesis. Meis2 is strongly expressed in neuroblasts of the subventricular zone (SVZ) and rostral migratory stream (RMS) and in some of the OB interneurons that are continuously replaced during adult life. Targeted manipulations with retroviral vectors expressing function-blocking forms or with small interfering RNAs demonstrated that Meis activity is cell-autonomously required for the acquisition of a general neuronal fate by SVZ-derived progenitors in vivo and in vitro. Additionally, Meis2 activity in the RMS is important for the generation of dopaminergic periglomerular neurons in the OB. Chromatin immunoprecipitation identified doublecortin and tyrosine hydroxylase as direct Meis targets in newly generated neurons and the OB, respectively. Furthermore, biochemical analyses revealed a previously unrecognized complex of Meis2 with Pax6 and Dlx2, two transcription factors involved in OB neurogenesis. The full pro-neurogenic activity of Pax6 in SVZ derived neural stem and progenitor cells requires the presence of Meis. Collectively, these results show that Meis2 cooperates with Pax6 in generic neurogenesis and dopaminergic fate specification in the adult SVZ-OB system.


Subject(s)
Dopaminergic Neurons/cytology , Eye Proteins/metabolism , Homeodomain Proteins/metabolism , Neurogenesis/physiology , Olfactory Bulb/embryology , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , Animals , Base Sequence , Cell Proliferation , Dopaminergic Neurons/metabolism , Doublecortin Domain Proteins , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Molecular Sequence Data , Neural Stem Cells/metabolism , Neurogenesis/genetics , Neuropeptides/metabolism , Olfactory Bulb/cytology , Olfactory Bulb/growth & development , PAX6 Transcription Factor , RNA Interference , RNA, Small Interfering/genetics , Transcription Factors/metabolism , Tyrosine 3-Monooxygenase/metabolism
15.
Dev Growth Differ ; 59(9): 701-712, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29124740

ABSTRACT

The anterior part of the embryonic telencephalon gives rise to several brain regions that are important for animal behavior, including the frontal cortex (FC) and the olfactory bulb. The FC plays an important role in decision-making behaviors, such as social and cognitive behavior, and the olfactory bulb is involved in olfaction. Here, we show the organizing activity of fibroblast growth factor 8 (Fgf8) in the regionalization of the anterior telencephalon, specifically the FC and the olfactory bulb. Misexpression of Fgf8 in the most anterior part of the mouse telencephalon at embryonic day 11.5 (E11.5) by ex utero electroporation resulted in a lateral shift of dorsal FC subdivision markers and a lateral expansion of the dorsomedial part of the FC, the future anterior cingulate and prelimbic cortex. Fgf8-transfected brains had lacked ventral FC, including the future orbital cortex, which was replaced by the expanded olfactory bulb. The olfactory region occupied a larger area of the FC when transfection efficiency of Fgf8 was higher. These results suggest that Fgf8 regulates the proportions of the FC and olfactory bulb in the anterior telencephalon and has a medializing effect on the formation of FC subdivisions.


Subject(s)
Fibroblast Growth Factor 8/metabolism , Telencephalon/metabolism , Animals , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental , Mice , Olfactory Bulb/embryology , Olfactory Bulb/metabolism , Telencephalon/embryology
16.
Nitric Oxide ; 71: 27-31, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29031735

ABSTRACT

Experimental work over the past several years has revealed an unexpected abundance of long natural antisense transcripts (NATs) in eukaryotic species. In light of the proposed role of such RNA molecules in the regulation of gene expression in the brain, attention is now focused on specific examples of neuronal NATs. Of particular interest are NATs that are complementary to mRNAs encoding nitric oxide synthase (NOS), the enzyme responsible for production of the important gaseous neurotransmitter nitric oxide (NO). Here we study the temporal expression profile of murine Nos3as NAT in the brain. Notably, Nos3as NAT is known to act as a negative regulator of Nos3 gene expression. The results of our quantitative analysis reveal differential expression of Nos3as NAT during embryonic and post-embryonic stages of development of the brain. Also, they show that the low levels of Nos3as NAT coincides with active neurogenesis. In addition we report on an inverse correlation between the relative expression level of Nos3as NAT and the level of Nos3 protein. Thus our data raise the hypothesis that the Nos3as NAT regulates neurogenesis through suppression of Nos3 gene activity. This idea is further supported by experiments conducted on the olfactory bulbs and cultured neuroblastoma cells.


Subject(s)
Brain/metabolism , Neurogenesis/genetics , RNA, Antisense/metabolism , Animals , Brain/embryology , Brain/growth & development , Cell Line, Tumor , Embryonic Development , Gene Expression Profiling , Gene Expression Regulation, Developmental , Mice , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Olfactory Bulb/embryology , Olfactory Bulb/growth & development , Olfactory Bulb/metabolism , RNA, Antisense/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors
17.
Cell Mol Life Sci ; 73(13): 2467-90, 2016 07.
Article in English | MEDLINE | ID: mdl-26994098

ABSTRACT

The components of the nervous system are assembled in development by the process of cell migration. Although the principles of cell migration are conserved throughout the brain, different subsystems may predominantly utilize specific migratory mechanisms, or may display unusual features during migration. Examining these subsystems offers not only the potential for insights into the development of the system, but may also help in understanding disorders arising from aberrant cell migration. The olfactory system is an ancient sensory circuit that is essential for the survival and reproduction of a species. The organization of this circuit displays many evolutionarily conserved features in vertebrates, including molecular mechanisms and complex migratory pathways. In this review, we describe the elaborate migrations that populate each component of the olfactory system in rodents and compare them with those described in the well-studied neocortex. Understanding how the components of the olfactory system are assembled will not only shed light on the etiology of olfactory and sexual disorders, but will also offer insights into how conserved migratory mechanisms may have shaped the evolution of the brain.


Subject(s)
Cell Movement , Olfactory Bulb/embryology , Olfactory Cortex/embryology , Olfactory Pathways , Rodentia/embryology , Animals , Biological Evolution , Hypothalamus/cytology , Hypothalamus/embryology , Neurons/cytology , Olfactory Bulb/cytology , Olfactory Cortex/cytology , Prosencephalon/cytology , Prosencephalon/embryology , Smell , Vomeronasal Organ/cytology , Vomeronasal Organ/embryology
18.
Mol Cell Neurosci ; 74: 78-86, 2016 07.
Article in English | MEDLINE | ID: mdl-27073125

ABSTRACT

Transmission of olfactory information to higher brain regions is mediated by olfactory bulb (OB) projection neurons, the mitral and tufted cells. Although mitral/tufted cells are often characterized as the OB counterpart of cortical projection neurons (also known as pyramidal neurons), they possess several unique morphological characteristics and project specifically to the olfactory cortices. Moreover, the molecular networks contributing to the generation of mitral/tufted cells during development are largely unknown. To understand the developmental patterns of gene expression in mitral/tufted cells in the OB, we performed transcriptome analyses targeting purified OB projection neurons at different developmental time points with next-generation RNA sequencing (RNA-seq). Through these analyses, we found 1202 protein-coding genes that are temporally differentially-regulated in developing projection neurons. Among them, 388 genes temporally changed their expression level only in projection neurons. The data provide useful resource to study the molecular mechanisms regulating development of mitral/tufted cells. We further compared the gene expression profiles of developing mitral/tufted cells with those of three cortical projection neuron subtypes, subcerebral projection neurons, corticothalamic projection neurons, and callosal projection neurons, and found that the molecular signature of developing olfactory projection neuron bears resemblance to that of subcerebral neurons. We also identified 3422 events that change the ratio of splicing isoforms in mitral/tufted cells during maturation. Interestingly, several genes expressed a novel isoform not previously reported. These results provide us with a broad perspective of the molecular networks underlying the development of OB projection neurons.


Subject(s)
Neurons/metabolism , Olfactory Bulb/metabolism , Transcriptome , Animals , Gene Expression Regulation, Developmental , Mice , Olfactory Bulb/cytology , Olfactory Bulb/embryology , Open Reading Frames
19.
Int J Mol Sci ; 18(2)2017 Jan 29.
Article in English | MEDLINE | ID: mdl-28146061

ABSTRACT

The peripheral nervous system (PNS) exhibits a much larger capacity for regeneration than the central nervous system (CNS). One reason for this difference is the difference in glial cell types between the two systems. PNS glia respond rapidly to nerve injury by clearing debris from the injury site, supplying essential growth factors and providing structural support; all of which enhances neuronal regeneration. Thus, transplantation of glial cells from the PNS is a very promising therapy for injuries to both the PNS and the CNS. There are two key types of PNS glia: olfactory ensheathing cells (OECs), which populate the olfactory nerve, and Schwann cells (SCs), which are present in the rest of the PNS. These two glial types share many similar morphological and functional characteristics but also exhibit key differences. The olfactory nerve is constantly turning over throughout life, which means OECs are continuously stimulating neural regeneration, whilst SCs only promote regeneration after direct injury to the PNS. This review presents a comparison between these two PNS systems in respect to normal physiology, developmental anatomy, glial functions and their responses to injury. A thorough understanding of the mechanisms and differences between the two systems is crucial for the development of future therapies using transplantation of peripheral glia to treat neural injuries and/or disease.


Subject(s)
Nerve Regeneration , Neuroglia/metabolism , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/pathology , Animals , Cell Transplantation , Homeostasis , Humans , Immunomodulation , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Neuroglia/immunology , Olfactory Bulb/cytology , Olfactory Bulb/embryology , Olfactory Bulb/physiology , Olfactory Nerve/cytology , Olfactory Nerve/embryology , Olfactory Nerve/physiology , Peripheral Nerve Injuries/immunology , Peripheral Nerve Injuries/therapy , Schwann Cells/physiology , Sensory Receptor Cells/metabolism , Signal Transduction
20.
Eur J Neurosci ; 43(8): 1111-21, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26833763

ABSTRACT

In a previous study, we applied a multiple-site optical recording technique with a voltage-sensitive dye to the embryonic chick olfactory system and showed that functional synaptic transmission in the olfactory bulb was expressed at embryonic 6-7-day stages. It is known that oscillations, i.e. stereotyped sinusoidal neural activity, appear in the olfactory system of various species. The focus of the present study is to determine whether the oscillation is also generated in the embryonic chick olfactory bulb and, if this is the case, when the oscillation appears and how its profiles change during embryogenesis. At the early stages of development (embryonic 6- to 8-day stages), postsynaptic response-related optical signals evoked by olfactory nerve stimulation exhibited a simple monophasic waveform that lasted for a few seconds. At embryonic 9-day stage, the optical signal became multi-phasic, and the oscillatory event was detected in some preparations. The oscillation was restricted to the distal half of the olfactory bulb. As development proceeded, the incidence and duration of the oscillation gradually increased, and the waveform became complicated. In some cases at embryonic 12-day stage, the oscillation lasted for nearly a minute. The frequency of the oscillation increased slightly with development, but it remained in the range of theta oscillation during the 9- to 12-day stages. We discuss the ontogenetic dynamics of the oscillation and the significance of this activity in the developing olfactory bulb.


Subject(s)
Action Potentials , Olfactory Bulb/physiology , Animals , Chick Embryo , Olfactory Bulb/embryology , Voltage-Sensitive Dye Imaging
SELECTION OF CITATIONS
SEARCH DETAIL