Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
1.
J Neurosci ; 43(6): 1051-1071, 2023 02 08.
Article in English | MEDLINE | ID: mdl-36596700

ABSTRACT

Parkinson's disease (PD) is characterized by multiple symptoms including olfactory dysfunction, whose underlying mechanisms remain unclear. Here, we explored pathologic changes in the olfactory pathway of transgenic (Tg) mice of both sexes expressing the human A30P mutant α-synuclein (α-syn; α-syn-Tg mice) at 6-7 and 12-14 months of age, representing early and late-stages of motor progression, respectively. α-Syn-Tg mice at late stages exhibited olfactory behavioral deficits, which correlated with severe α-syn pathology in projection neurons (PNs) of the olfactory pathway. In parallel, olfactory bulb (OB) neurogenesis in α-syn-Tg mice was reduced in the OB granule cells at six to seven months and OB periglomerular cells at 12-14 months, respectively, both of which could contribute to olfactory dysfunction. Proteomic analyses showed a disruption in endocytic and exocytic pathways in the OB during the early stages which appeared exacerbated at the synaptic terminals when the mice developed olfactory deficits at 12-14 months. Our data suggest that (1) the α-syn-Tg mice recapitulate the olfactory functional deficits seen in PD; (2) olfactory structures exhibit spatiotemporal disparities for vulnerability to α-syn pathology; (3) α-syn pathology is restricted to projection neurons in the olfactory pathway; (4) neurogenesis in adult α-syn-Tg mice is reduced in the OB; and (5) synaptic endocytosis and exocytosis defects in the OB may further explain olfactory deficits.SIGNIFICANCE STATEMENT Olfactory dysfunction is a characteristic symptom of Parkinson's disease (PD). Using the human A30P mutant α-synuclein (α-syn)-expressing mouse model, we demonstrated the appearance of olfactory deficits at late stages of the disease, which was accompanied by the accumulation of α-syn pathology in projection neurons (PNs) of the olfactory system. This dysfunction included a reduction in olfactory bulb (OB) neurogenesis as well as changes in synaptic vesicular transport affecting synaptic function, both of which are likely contributing to olfactory behavioral deficits.


Subject(s)
Olfaction Disorders , Parkinson Disease , Male , Female , Mice , Humans , Animals , Parkinson Disease/genetics , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Smell , Proteomics , Mice, Transgenic , Neurogenesis , Olfaction Disorders/genetics , Disease Models, Animal
2.
J Neurophysiol ; 124(2): 375-387, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32639901

ABSTRACT

The first compartmental computer models of brain neurons using the Rall method predicted novel and unexpected dendrodendritic interactions between mitral and granule cells in the olfactory bulb. We review the models from a 50-year perspective on the work that has challenged, supported, and extended the original proposal that these interactions mediate both lateral inhibition and oscillatory activity, essential steps in the neural basis of olfactory processing and perception. We highlight strategies behind the neurophysiological experiments and the Rall methods that enhance the ability of detailed compartmental modeling to give counterintuitive predictions that lead to deeper insights into neural organization at the synaptic and circuit level. The application of these methods to mechanisms of neurogenesis and plasticity are exciting challenges for the future.


Subject(s)
Brain Waves/physiology , Dendrites/physiology , Models, Theoretical , Neural Inhibition/physiology , Neurogenesis/physiology , Neuronal Plasticity/physiology , Olfactory Bulb/physiology , Olfactory Perception/physiology , Synapses/physiology , Animals
3.
Cereb Cortex ; 29(1): 1-16, 2019 01 01.
Article in English | MEDLINE | ID: mdl-29136113

ABSTRACT

Piriform cortex (PC) is a 3-layer paleocortex receiving primary afferent input from the olfactory bulb. The past decade has seen significant progress in understanding the synaptic, cellular and functional organization of PC, but PC embryogenesis continues to be enigmatic. Here, using birthdating strategies and clonal analyses, we probed the early development and laminar specificity of neurogenesis/gliogenesis as it relates to the organization of the PC. Our data demonstrate a temporal sequence of laminar-specific neurogenesis following the canonical "inside-out" pattern, with the notable exception of PC Layer II which exhibited an inverse "outside-in" temporal neurogenic pattern. Of interest, we found no evidence of a neurogenic gradient along the anterior to posterior axis, although the timing of neuronal migration and laminar development was delayed rostrally by approximately 24 h. To begin probing if lineage affected cell fate in the PC, we labeled PC neuroblasts using a multicolor technique and analyzed their laminar organization. Our results suggested that PC progenitors were phenotypically committed to reach specific layers early in the development. Collectively, these studies shed new light on the determinants of the laminar specificity of neuronal/glial organization in PC and the likely role of subpopulations of committed progenitors in regulating PC embryogenesis.


Subject(s)
Cell Lineage/physiology , Cell Movement/physiology , Neurogenesis/physiology , Neuroglia/physiology , Piriform Cortex/cytology , Piriform Cortex/growth & development , Animals , Female , HEK293 Cells , Humans , Male , Mice , Pregnancy
4.
Mol Cell Neurosci ; 98: 82-96, 2019 07.
Article in English | MEDLINE | ID: mdl-31200100

ABSTRACT

The olfactory tubercle (OT) is located in the ventral-medial region of the brain where it receives primary input from olfactory bulb (OB) projection neurons and processes olfactory behaviors related to motivation, hedonics of smell and sexual encounters. The OT is part of the dopamine reward system that shares characteristics with the striatum. Together with the nucleus accumbens, the OT has been referred to as the "ventral striatum". However, despite its functional importance little is known about the embryonic development of the OT and the phenotypic properties of the OT cells. Here, using thymidine analogs, we establish that mouse OT neurogenesis occurs predominantly between E11-E15 in a lateral-to-medial gradient. Then, using a piggyBac multicolor technique we characterized the migratory route of OT neuroblasts from their embryonic point of origin. Following neurogenesis in the ventral lateral ganglionic eminence (vLGE), neuroblasts destined for the OT followed a dorsal-ventral pathway we named "ventral migratory course" (VMC). Upon reaching the nascent OT, neurons established a prototypical laminar distribution that was determined, in part, by the progenitor cell of origin. A phenotypic analysis of OT neuroblasts using a single-color piggyBac technique, showed that OT shared the molecular specification of striatal neurons. In addition to primary afferent input from the OB, the OT also receives a robust dopaminergic input from ventral tegmentum (Ikemoto, 2007). We used tyrosine hydroxylase (TH) expression as a proxy for dopaminergic innervation and showed that TH onset occurs at E13 and progressively increased until postnatal stages following an 'inside-out' pattern. Postnatally, we established the myelination in the OT occurring between P7 and P14, as shown with CNPase staining, and we characterized the cellular phenotypes populating the OT by immunohistochemistry. Collectively, this work provides the first detailed analysis of the developmental and maturation processes occurring in mouse OT, and demonstrates the striatal nature of the OT as part of the ventral striatum (vST).


Subject(s)
Neurogenesis , Olfactory Tubercle/embryology , Animals , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Female , Male , Mice , Myelin Sheath/metabolism , Olfactory Tubercle/cytology , Olfactory Tubercle/growth & development
5.
Proc Natl Acad Sci U S A ; 112(18): 5821-6, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25902488

ABSTRACT

Odorant receptors (OR) are strongly implicated in coalescence of olfactory sensory neuron (OSN) axons and the formation of olfactory bulb (OB) glomeruli. However, when ORs are first expressed relative to basal cell division and OSN axon extension is unknown. We developed an in vivo fate-mapping strategy that enabled us to follow OSN maturation and axon extension beginning at basal cell division. In parallel, we mapped the molecular development of OSNs beginning at basal cell division, including the onset of OR expression. Our data show that ORs are first expressed around 4 d following basal cell division, 24 h after OSN axons have reached the OB. Over the next 6+ days the OSN axons navigate the OB nerve layer and ultimately coalesce in glomeruli. These data provide a previously unidentified perspective on the role of ORs in homophilic OSN axon adhesion and lead us to propose a new model dividing axon extension into two phases. Phase I is OR-independent and accounts for up to 50% of the time during which axons approach the OB and begin navigating the olfactory nerve layer. Phase II is OR-dependent and concludes as OSN axons coalesce in glomeruli.


Subject(s)
Axons/metabolism , Olfactory Bulb/physiology , Receptors, Odorant/metabolism , Sensory Receptor Cells/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Adhesion , Cell Differentiation , Cell Movement , Electroporation , GAP-43 Protein/metabolism , Immunohistochemistry , In Situ Hybridization , Kidney/metabolism , Mice , Mitosis , Neurogenesis , Neurons/metabolism , Neurons, Afferent/cytology , Odorants , Olfactory Bulb/cytology , Olfactory Nerve/cytology , Olfactory Receptor Neurons/metabolism , Smell/genetics , Stem Cells/cytology , Tamoxifen/chemistry
6.
Nature ; 472(7342): 186-90, 2011 Apr 14.
Article in English | MEDLINE | ID: mdl-21441906

ABSTRACT

Loss of function of the gene SCN9A, encoding the voltage-gated sodium channel Na(v)1.7, causes a congenital inability to experience pain in humans. Here we show that Na(v)1.7 is not only necessary for pain sensation but is also an essential requirement for odour perception in both mice and humans. We examined human patients with loss-of-function mutations in SCN9A and show that they are unable to sense odours. To establish the essential role of Na(v)1.7 in odour perception, we generated conditional null mice in which Na(v)1.7 was removed from all olfactory sensory neurons. In the absence of Na(v)1.7, these neurons still produce odour-evoked action potentials but fail to initiate synaptic signalling from their axon terminals at the first synapse in the olfactory system. The mutant mice no longer display vital, odour-guided behaviours such as innate odour recognition and avoidance, short-term odour learning, and maternal pup retrieval. Our study creates a mouse model of congenital general anosmia and provides new strategies to explore the genetic basis of the human sense of smell.


Subject(s)
Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation/genetics , Olfaction Disorders/genetics , Olfaction Disorders/physiopathology , Sodium Channels/genetics , Action Potentials , Animals , Behavior, Animal , Disease Models, Animal , Female , Gene Expression Profiling , Humans , Male , Mice , NAV1.7 Voltage-Gated Sodium Channel , Odorants/analysis , Olfaction Disorders/congenital , Olfaction Disorders/pathology , Olfactory Mucosa/cytology , Olfactory Mucosa/pathology , Olfactory Pathways/metabolism , Olfactory Pathways/pathology , Olfactory Pathways/physiopathology , Olfactory Perception/genetics , Olfactory Perception/physiology , Olfactory Receptor Neurons/metabolism , Olfactory Receptor Neurons/pathology , Pain/genetics , Pain/physiopathology , Phenotype , Smell/genetics , Smell/physiology , Sodium Channels/deficiency , Sodium Channels/metabolism , Synapses/metabolism , Synapses/pathology , Urine/chemistry
7.
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
8.
Proc Natl Acad Sci U S A ; 111(47): 16931-6, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25385630

ABSTRACT

The piriform cortex (PCX) is the largest component of the olfactory cortex and is hypothesized to be the locus of odor object formation. The distributed odorant representation found in PCX contrasts sharply with the topographical representation seen in other primary sensory cortices, making it difficult to test this view. Recent work in PCX has focused on functional characteristics of these distributed afferent and association fiber systems. However, information regarding the efferent projections of PCX and how those may be involved in odor representation and object recognition has been largely ignored. To investigate this aspect of PCX, we have used the efferent pathway from mouse PCX to the orbitofrontal cortex (OFC). Using double fluorescent retrograde tracing, we identified the output neurons (OPNs) of the PCX that project to two subdivisions of the OFC, the agranular insula and the lateral orbitofrontal cortex (AI-OPNs and LO-OPNs, respectively). We found that both AI-OPNs and LO-OPNs showed a distinct spatial topography within the PCX and fewer than 10% projected to both the AI and the LO as judged by double-labeling. These data revealed that the efferent component of the PCX may be topographically organized. Further, these data suggest a model for functional organization of the PCX in which the OPNs are grouped into parallel output circuits that provide olfactory information to different higher centers. The distributed afferent input from the olfactory bulb and the local PCX association circuits would then ensure a complete olfactory representation, pattern recognition capability, and neuroplasticity in each efferent circuit.


Subject(s)
Piriform Cortex/anatomy & histology , Sensory Receptor Cells/cytology , Animals , Mice , Piriform Cortex/cytology
9.
Proc Natl Acad Sci U S A ; 111(38): 13984-9, 2014 Sep 23.
Article in English | MEDLINE | ID: mdl-25189772

ABSTRACT

The production of new neurons in the olfactory bulb (OB) through adulthood is a major mechanism of structural and functional plasticity underlying learning-induced circuit remodeling. The recruitment of adult-born OB neurons depends not only on sensory input but also on the context in which the olfactory stimulus is received. Among the multiple steps of adult neurogenesis, the integration and survival of adult-born neurons are both strongly influenced by olfactory learning. Conversely, optogenetic stimulation of adult-born neurons has been shown to specifically improve olfactory learning and long-term memory. However, the nature of the circuit and the synaptic mechanisms underlying this reciprocal influence are not yet known. Here, we showed that olfactory learning increases the spine density in a region-restricted manner along the dendritic tree of adult-born granule cells (GCs). Anatomical and electrophysiological analysis of adult-born GCs showed that olfactory learning promotes a remodeling of both excitatory and inhibitory inputs selectively in the deep dendritic domain. Circuit mapping revealed that the malleable dendritic portion of adult-born neurons receives excitatory inputs mostly from the regions of the olfactory cortex that project back to the OB. Finally, selective optogenetic stimulation of olfactory cortical projections to the OB showed that learning strengthens these inputs onto adult-born GCs. We conclude that learning promotes input-specific synaptic plasticity in adult-born neurons, which reinforces the top-down influence from the olfactory cortex to early stages of olfactory information processing.


Subject(s)
Dendrites/metabolism , Memory, Long-Term/physiology , Neurogenesis/physiology , Neuronal Plasticity/physiology , Olfactory Bulb/metabolism , Olfactory Pathways/metabolism , Animals , Male , Mice , Mice, Transgenic , Olfactory Bulb/cytology , Olfactory Pathways/cytology
10.
Glia ; 63(9): 1646-59, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25856239

ABSTRACT

The olfactory nerve is permissive for axon growth throughout life. This has been attributed in part to the olfactory ensheathing glial cells that encompass the olfactory sensory neuron fascicles. Olfactory ensheathing cells (OECs) also promote axon growth in vitro and when transplanted in vivo to sites of injury. The mechanisms involved remain largely unidentified owing in part to the limited knowledge of the physiological properties of ensheathing cells. Glial cells rely for many functions on the properties of the potassium channels expressed; however, those expressed in ensheathing cells are unknown. Here we show that OECs express voltage-dependent potassium currents compatible with inward rectifier (Kir ) and delayed rectifier (KDR ) channels. Together with gap junction coupling, these contribute to the heterogeneity of membrane properties observed in OECs. The relevance of K(+) currents expressed by ensheathing cells is discussed in relation to plasticity of the olfactory nerve.


Subject(s)
Myelin Sheath/physiology , Olfactory Nerve/cytology , Olfactory Nerve/physiology , Potassium Channels, Voltage-Gated/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/physiology , Connexin 43/metabolism , Female , Gap Junctions/drug effects , Gap Junctions/metabolism , Immunohistochemistry , Male , Mice , Myelin Sheath/drug effects , Olfactory Nerve/drug effects , Patch-Clamp Techniques , Potassium/metabolism , Tissue Culture Techniques
11.
Eur J Neurosci ; 41(2): 147-56, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25393912

ABSTRACT

Mitral and tufted cells are the projection neurons of the olfactory bulb (OB). We previously reported that somata location and innervation patterns were different between early- and late-born mitral cells (Imamura et al., 2011). Here, we introduced a plasmid that drives the expression of a GFP gene into the mouse OB using in utero electroporation, and demonstrated that we can deliver the plasmid vectors into distinct subsets of OB projection neurons by changing the timing of electroporation after fertilisation. The electroporation performed at embryonic day (E)10 preferentially labeled mitral cells in the accessory OB and main OB mitral cells in dorsomedial mitral cell layer (MCL). In contrast, the E12 electroporation introduced the plasmid vectors preferentially into main OB mitral cells in the ventrolateral MCL and tufted cells. Combining these data with BrdU injections, we confirmed that E10 and E12 electroporation preferentially labeled early- and late-born projection neurons, respectively. This work introduces a novel method for segregated labeling of mouse olfactory bulb projection neurons based on their birthdates. With this technique we found that early- and late-born projection neurons extend their secondary dendrites in the deep and superficial external plexiform layer (EPL), respectively. Although a similar segregation has been suggested for mitral vs. tufted cell dendrites, we found mitral cells projecting secondary dendrites into the superficial EPL in E12-electroporated main OB. Our observations indicate that timing of neurogenesis regulates not only somata location and innervation patterns but also the laminar organisation of projection neuron dendrites in the EPL.


Subject(s)
Neurons/cytology , Olfactory Bulb/cytology , Olfactory Bulb/embryology , Animals , Bromodeoxyuridine , Electroporation/methods , Genetic Vectors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Mice , Neural Pathways/cytology , Neurogenesis , Plasmids/genetics
12.
J Neurosci ; 33(16): 6742-52, 2013 Apr 17.
Article in English | MEDLINE | ID: mdl-23595732

ABSTRACT

Most dendrite branches and a large fraction of dendritic spines in the adult rodent forebrain are stable for extended periods of time. Destabilization of these structures compromises brain function and is a major contributing factor to psychiatric and neurodegenerative diseases. Integrins are a class of transmembrane extracellular matrix receptors that function as αß heterodimers and activate signaling cascades regulating the actin cytoskeleton. Here we identify integrin α3 as a key mediator of neuronal stability. Dendrites, dendritic spines, and synapses develop normally in mice with selective loss of integrin α3 in excitatory forebrain neurons, reaching their mature sizes and densities through postnatal day 21 (P21). However, by P42, integrin α3 mutant mice exhibit significant reductions in hippocampal dendrite arbor size and complexity, loss of dendritic spine and synapse densities, and impairments in hippocampal-dependent behavior. Furthermore, gene-dosage experiments demonstrate that integrin α3 interacts functionally with the Arg nonreceptor tyrosine kinase to activate p190RhoGAP, which inhibits RhoA GTPase and regulates hippocampal dendrite and synapse stability and mouse behavior. Together, our data support a fundamental role for integrin α3 in regulating dendrite arbor stability, synapse maintenance, and proper hippocampal function. In addition, these results provide a biochemical and structural explanation for the defects in long-term potentiation, learning, and memory reported previously in mice lacking integrin α3.


Subject(s)
Dendrites/genetics , Dendritic Spines/genetics , Integrin alpha3/metabolism , Neurons/cytology , Recognition, Psychology/physiology , Synapses/physiology , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Dendrites/ultrastructure , Dendritic Spines/ultrastructure , Disks Large Homolog 4 Protein , Female , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Guanylate Kinases/metabolism , Hippocampus/cytology , Immunoprecipitation , Integrin alpha3/genetics , Lysine/analogs & derivatives , Lysine/metabolism , Male , Membrane Proteins/metabolism , Memory Disorders/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Nerve Tissue Proteins/genetics , Phosphopyruvate Hydratase/metabolism , Synapses/ultrastructure , alpha-Fetoproteins/deficiency , rhoA GTP-Binding Protein/metabolism
13.
Mol Cell Neurosci ; 54: 58-70, 2013 May.
Article in English | MEDLINE | ID: mdl-23353076

ABSTRACT

Tracking olfactory bulb mitral cell development with BrdU labeling, we find that mitral cells are generated from Pax6+ radial glial cells in the ventricular zone of the embryonic olfactory bulb. Unlike cortical projection neurons, postmitotic mitral cell precursors express both Tbr1 and Tbr2. Our tracking experiments revealed that down-regulation of Pax6 preceded up-regulation of Tbrs, and that Tbr1 emerged earlier than Tbr2. Using in utero electroporation, we also show that Pax6 negatively regulates the expression of Tbr1 and Tbr2 in postmitotic mitral cell precursors. Exogenous expression of Pax6 in embryonic olfactory bulb postmitotic precursors decreased the number of cells that progressed to a mitral cell fate. In contrast, exogenous expression of Pax6 resulted in an increase of GABAergic and/or dopaminergic interneurons. These results indicate that Pax6 is a regulator of fate determination of precursor cells.


Subject(s)
DNA-Binding Proteins/genetics , Eye Proteins/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Interneurons/cytology , Olfactory Bulb/cytology , Paired Box Transcription Factors/genetics , Repressor Proteins/genetics , T-Box Domain Proteins/genetics , Animals , DNA-Binding Proteins/metabolism , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Eye Proteins/metabolism , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Homeodomain Proteins/metabolism , Interneurons/metabolism , Mice , Mitosis , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis , Neuroglia/cytology , Neuroglia/metabolism , Olfactory Bulb/embryology , Olfactory Bulb/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , T-Box Domain Proteins/metabolism
14.
eNeuro ; 11(6)2024 Jun.
Article in English | MEDLINE | ID: mdl-38834299

ABSTRACT

Viruses, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), use respiratory epithelial cells as an entry point for infection. Within the nasal cavity, the olfactory epithelium (OE) is particularly sensitive to infections which may lead to olfactory dysfunction. In patients suffering from coronavirus disease 2019, deficits in olfaction have been characterized as a distinctive symptom. Here, we used the K18hACE2 mice to study the spread of SARS-CoV-2 infection and inflammation in the olfactory system (OS) after 7 d of infection. In the OE, we found that SARS-CoV-2 selectively targeted the supporting/sustentacular cells (SCs) and macrophages from the lamina propria. In the brain, SARS-CoV-2 infected some microglial cells in the olfactory bulb (OB), and there was a widespread infection of projection neurons in the OB, piriform cortex (PC), and tubular striatum (TuS). Inflammation, indicated by both elevated numbers and morphologically activated IBA1+ cells (monocyte/macrophage lineages), was preferentially increased in the OE septum, while it was homogeneously distributed throughout the layers of the OB, PC, and TuS. Myelinated OS axonal tracts, the lateral olfactory tract, and the anterior commissure, exhibited decreased levels of 2',3'-cyclic-nucleotide 3'-phosphodiesterase, indicative of myelin defects. Collectively, our work supports the hypothesis that SARS-CoV-2 infected SC and macrophages in the OE and, centrally, microglia and subpopulations of OS neurons. The observed inflammation throughout the OS areas and central myelin defects may account for the long-lasting olfactory deficit.


Subject(s)
COVID-19 , Myelin Sheath , Olfactory Bulb , Olfactory Mucosa , SARS-CoV-2 , Animals , COVID-19/pathology , COVID-19/complications , Mice , Olfactory Mucosa/pathology , Olfactory Mucosa/virology , Olfactory Bulb/pathology , Olfactory Bulb/virology , Myelin Sheath/pathology , Myelin Sheath/metabolism , Microglia/pathology , Microglia/metabolism , Microglia/virology , Mice, Transgenic , Angiotensin-Converting Enzyme 2/metabolism , Olfaction Disorders/pathology , Olfaction Disorders/virology , Disease Models, Animal , Male , Inflammation/pathology , Inflammation/virology , Macrophages/pathology , Female
15.
bioRxiv ; 2024 Aug 04.
Article in English | MEDLINE | ID: mdl-39131289

ABSTRACT

Genetic variants in TRIO are associated with neurodevelopmental disorders (NDDs) including schizophrenia (SCZ), autism spectrum disorder (ASD) and intellectual disability. TRIO uses its two guanine nucleotide exchange factor (GEF) domains to activate GTPases (GEF1: Rac1 and RhoG; GEF2: RhoA) that control neuronal development and connectivity. It remains unclear how discrete TRIO variants differentially impact these neurodevelopmental events. Here, we investigate how heterozygosity for NDD-associated Trio variants - +/K1431M (ASD), +/K1918X (SCZ), and +/M2145T (bipolar disorder, BPD) - impact mouse behavior, brain development, and synapse structure and function. Heterozygosity for different Trio variants impacts motor, social, and cognitive behaviors in distinct ways that align with clinical phenotypes in humans. Trio variants differentially impact head and brain size with corresponding changes in dendritic arbors of motor cortex layer 5 pyramidal neurons (M1 L5 PNs). Although neuronal structure was only modestly altered in the Trio variant heterozygotes, we observe significant changes in synaptic function and plasticity. We also identified distinct changes in glutamate synaptic release in +/K1431M and +/M2145T cortico-cortical synapses. The TRIO K1431M GEF1 domain has impaired ability to promote GTP exchange on Rac1, but +/K1431M mice exhibit increased Rac1 activity, associated with increased levels of the Rac1 GEF Tiam1. Acute Rac1 inhibition with NSC23766 rescued glutamate release deficits in +/K1431M variant cortex. Our work reveals that discrete NDD-associated Trio variants yield overlapping but distinct phenotypes in mice, demonstrates an essential role for Trio in presynaptic glutamate release, and underscores the importance of studying the impact of variant heterozygosity in vivo.

16.
J Neurosci ; 32(8): 2824-34, 2012 Feb 22.
Article in English | MEDLINE | ID: mdl-22357865

ABSTRACT

Integrins are heterodimeric extracellular matrix receptors that are essential for the proper development of the vertebrate nervous system. We report here that selective loss of integrin ß1 in excitatory neurons leads to reductions in the size and complexity of hippocampal dendritic arbors, hippocampal synapse loss, impaired hippocampus-dependent learning, and exaggerated psychomotor sensitivity to cocaine in mice. Our biochemical and genetic experiments demonstrate that the intracellular tail of integrin ß1 binds directly to Arg kinase and that this interaction stimulates activity of the Arg substrate p190RhoGAP, an inactivator of the RhoA GTPase. Moreover, genetic manipulations that reduce integrin ß1 signaling through Arg recapitulate the integrin ß1 knock-out phenotype in a gene dose-sensitive manner. Together, these results describe a novel integrin ß1-Arg-p190RhoGAP pathway that regulates dendritic arbor size, promotes synapse maintenance, supports proper hippocampal function, and mitigates the behavioral consequences of cocaine exposure.


Subject(s)
Dendrites/metabolism , Exploratory Behavior/physiology , Integrin beta1/metabolism , Neurons/cytology , Signal Transduction/genetics , Synapses/physiology , alpha-Fetoproteins/metabolism , Analysis of Variance , Animals , Animals, Newborn , Avoidance Learning/drug effects , Avoidance Learning/physiology , Basic Helix-Loop-Helix Transcription Factors/deficiency , Cells, Cultured , Cocaine/administration & dosage , Dendrites/ultrastructure , Enzyme-Linked Immunosorbent Assay , Exploratory Behavior/drug effects , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Hippocampus/ultrastructure , Immunoprecipitation , Integrin beta1/genetics , Male , Mice , Mice, Knockout , Mutation/physiology , Nerve Tissue Proteins/deficiency , Neurons/physiology , Neurons/ultrastructure , Organ Culture Techniques , Post-Synaptic Density/genetics , Post-Synaptic Density/pathology , Post-Synaptic Density/ultrastructure , Protein Binding/drug effects , Protein Binding/genetics , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction/drug effects , Synapses/ultrastructure , alpha-Fetoproteins/genetics , src Homology Domains/drug effects , src Homology Domains/physiology
17.
Proc Natl Acad Sci U S A ; 107(35): 15613-8, 2010 Aug 31.
Article in English | MEDLINE | ID: mdl-20679234

ABSTRACT

Little is known about how normal aging affects the brain. Recent evidence suggests that neuronal loss is not ubiquitous in aging neocortex. Instead, subtle and still controversial, region- and layer-specific alterations of neuron morphology and synapses are reported during aging, leading to the notion that discrete changes in neural circuitry may underlie age-related cognitive deficits. Although deficits in sensory function suggest that primary sensory cortices are affected by aging, our understanding of the age-related cellular and molecular changes is sparse. To assess the effect of aging on the organization of olfactory bulb (OB) circuitry, we carried out quantitative morphometric analyses in the mouse OB at 2, 6, 12, 18, and 24 mo. Our data establish that the volumes of the major OB layers do not change during aging. Parallel to this, we are unique in demonstrating that the stereotypic glomerular convergence of M72-GFP OSN axons in the OB is preserved during aging. We then provide unique evidence of the stability of projection neurons and interneurons subpopulations in the aging mouse OB, arguing against the notion of an age-dependent widespread loss of neurons. Finally, we show ultrastructurally a significant layer-specific loss of synapses; synaptic density is reduced in the glomerular layer but not the external plexiform layer, leading to an imbalance in OB circuitry. These results suggest that reduction of afferent synaptic input and local modulatory circuit synapses in OB glomeruli may contribute to specific age-related alterations of the olfactory function.


Subject(s)
Aging/physiology , Nerve Net/physiology , Olfactory Bulb/physiology , Synapses/physiology , Animals , Axons/physiology , Dendrites/physiology , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Interneurons/cytology , Interneurons/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Microscopy, Confocal , Microscopy, Electron , Nerve Net/cytology , Olfactory Bulb/metabolism , Olfactory Bulb/ultrastructure , Receptors, Odorant/genetics , Receptors, Odorant/metabolism , Sensory Receptor Cells/cytology , Sensory Receptor Cells/physiology
18.
eNeuro ; 10(2)2023 02.
Article in English | MEDLINE | ID: mdl-36697258

ABSTRACT

Microglia invade the neuroblast migratory corridor of the rostral migratory stream (RMS) early in development. The early postnatal RMS does not yet have the dense astrocyte and vascular scaffold that helps propel forward migrating neuroblasts, which led us to consider whether microglia help regulate conditions permissive to neuroblast migration in the RMS. GFP-labeled microglia in CX3CR-1GFP/+ mice assemble primarily along the outer borders of the RMS during the first postnatal week, where they exhibit predominantly an ameboid morphology and associate with migrating neuroblasts. Microglia ablation for 3 d postnatally does not impact the density of pulse labeled BrdU+ neuroblasts nor the distance migrated by tdTomato electroporated neuroblasts in the RMS. However, microglia wrap DsRed-labeled neuroblasts in the RMS of P7 CX3CR-1GFP/+;DCXDsRed/+ mice and express the markers CD68, CLEC7A, MERTK, and IGF-1, suggesting active regulation in the developing RMS. Microglia depletion for 14 d postnatally further induced an accumulation of CC3+ DCX+ apoptotic neuroblasts in the RMS, a wider RMS and extended patency of the lateral ventricle extension in the olfactory bulb. These findings illustrate the importance of microglia in maintaining a healthy neuroblast population and an environment permissive to neuroblast migration in the early postnatal RMS.


Subject(s)
Microglia , Neural Stem Cells , Mice , Animals , Neural Stem Cells/physiology , Lateral Ventricles , Cell Movement/physiology , Olfactory Bulb/physiology
19.
J Neurosci ; 31(6): 2205-15, 2011 Feb 09.
Article in English | MEDLINE | ID: mdl-21307257

ABSTRACT

The fragile X mental retardation protein (FMRP) is an RNA-binding protein essential for multiple aspects of neuronal mRNA metabolism. Its absence leads to the fragile X syndrome, the most prevalent genetic form of mental retardation. The anatomical landmark of the disease, also present in the Fmr1 knock-out (KO) mice, is the hyperabundance of immature-looking lengthened dendritic spines. We used the well known continuous production of adult-born granule cells (GCs) in the mouse olfactory bulb (OB) to analyze the consequences of Fmrp loss on the differentiation of GCs. Morphological analysis of GCs in the Fmr1 KO mice showed an increase in spine density without a change in spine length. We developed an RNA interference strategy to cell-autonomously mutate Fmr1 in a wild-type OB network. Mutated GCs displayed an increase in spine density and spine length. Detailed analysis of the spines through immunohistochemistry, electron microscopy, and electrophysiology surprisingly showed that, despite these abnormalities, spines receive normal glutamatergic synapses, and thus that mutated adult-born neurons are synaptically integrated into the OB circuitry. Time-course analysis of the spine defects showed that Fmrp cell-autonomously downregulates the level and rate of spine production and limits their overgrowth. Finally, we report that Fmrp does not regulate dendritogenesis in standard conditions but is necessary for activity-dependent dendritic remodeling. Overall, our study of Fmrp in the context of adult neurogenesis has enabled us to carry out a precise dissection of the role of Fmrp in neuronal differentiation and underscores its pleiotropic involvement in both spinogenesis and dendritogenesis.


Subject(s)
Cell Differentiation/genetics , Fragile X Mental Retardation Protein/metabolism , Neurogenesis/physiology , Neurons/physiology , Olfactory Bulb/cytology , Analysis of Variance , Animals , Cell Differentiation/drug effects , Dendrites/drug effects , Dendrites/physiology , Dendrites/ultrastructure , Dendritic Spines/drug effects , Dendritic Spines/physiology , Dendritic Spines/ultrastructure , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Fragile X Mental Retardation Protein/genetics , Green Fluorescent Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Mutation/genetics , Neurogenesis/genetics , Neurons/drug effects , Patch-Clamp Techniques/methods , RNA, Small Interfering/pharmacology , Synapses/metabolism , Synapses/ultrastructure , Time Factors
20.
Cereb Cortex ; 21(6): 1231-45, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21041199

ABSTRACT

The piriform cortex (PCX) is a trilaminar paleocortex that is of interest for its role in odor coding and as a model for studying general principles of cortical sensory processing. While the structure of the mature PCX has been well characterized, its development is poorly understood. Notably, the kinetics as well as the cellular and morphological basis of the postnatal events that shape the PCX remain unknown. We followed the cellular fates of early- versus late-born cells in layer II of the anterior PCX, with a focus on the molecular maturation of pyramidal cells and the kinetics of their differentiation. We showed that: 1) early-born pyramidal cells differentiate more rapidly than late-born cells and 2) the position of pyramidal cells within the thickness of layer II determines the kinetics of their molecular maturation. We then examined the postnatal development of cortical lamination and showed that the establishment of inhibitory networks in the PCX proceeds through an increase in the density of inhibitory synapses despite a decrease in the number of interneurons. Together, our results provide a more comprehensive view of the postnatal development of the anterior PCX and reveal both similarities and differences in the development of this paleocortex versus the neocortex.


Subject(s)
Cerebral Cortex/anatomy & histology , Cerebral Cortex/growth & development , Gene Expression Regulation, Developmental/physiology , Neurons/physiology , Nonlinear Dynamics , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Bromodeoxyuridine/metabolism , Cell Differentiation/genetics , Cerebral Cortex/embryology , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental/genetics , Glutamate Decarboxylase/genetics , Green Fluorescent Proteins/genetics , Indoles , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurons/cytology , Pregnancy , Rats
SELECTION OF CITATIONS
SEARCH DETAIL