Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.722
Filter
Add more filters

Publication year range
1.
Cell ; 180(2): 373-386.e15, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31955847

ABSTRACT

Molecular interactions at the cellular interface mediate organized assembly of single cells into tissues and, thus, govern the development and physiology of multicellular organisms. Here, we developed a cell-type-specific, spatiotemporally resolved approach to profile cell-surface proteomes in intact tissues. Quantitative profiling of cell-surface proteomes of Drosophila olfactory projection neurons (PNs) in pupae and adults revealed global downregulation of wiring molecules and upregulation of synaptic molecules in the transition from developing to mature PNs. A proteome-instructed in vivo screen identified 20 cell-surface molecules regulating neural circuit assembly, many of which belong to evolutionarily conserved protein families not previously linked to neural development. Genetic analysis further revealed that the lipoprotein receptor LRP1 cell-autonomously controls PN dendrite targeting, contributing to the formation of a precise olfactory map. These findings highlight the power of temporally resolved in situ cell-surface proteomic profiling in discovering regulators of brain wiring.


Subject(s)
Olfactory Pathways/metabolism , Olfactory Receptor Neurons/metabolism , Proteomics/methods , Animals , Axons/metabolism , Brain/metabolism , Dendrites/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental/genetics , Membrane Proteins/metabolism , Neurogenesis/physiology , Olfactory Nerve/metabolism , Olfactory Pathways/cytology , Olfactory Pathways/physiology , Receptors, Lipoprotein/metabolism , Smell/physiology
2.
Cell ; 175(1): 57-70.e17, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30220455

ABSTRACT

Neurons in Caenorhabditis elegans and other nematodes have been thought to lack classical action potentials. Unexpectedly, we observe membrane potential spikes with defining characteristics of action potentials in C. elegans AWA olfactory neurons recorded under current-clamp conditions. Ion substitution experiments, mutant analysis, pharmacology, and modeling indicate that AWA fires calcium spikes, which are initiated by EGL-19 voltage-gated CaV1 calcium channels and terminated by SHK-1 Shaker-type potassium channels. AWA action potentials result in characteristic signals in calcium imaging experiments. These calcium signals are also observed when intact animals are exposed to odors, suggesting that natural odor stimuli induce AWA spiking. The stimuli that elicit action potentials match AWA's specialized function in climbing odor gradients. Our results provide evidence that C. elegans neurons can encode information through regenerative all-or-none action potentials, expand the computational repertoire of its nervous system, and inform future modeling of its neural coding and network dynamics.


Subject(s)
Action Potentials/physiology , Olfactory Nerve/physiology , Smell/physiology , Animals , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/metabolism , Calcium/metabolism , Calcium Channels/physiology , Chemotaxis/physiology , Membrane Potentials/physiology , Odorants , Olfactory Receptor Neurons/metabolism
3.
Eur J Neurosci ; 60(1): 3719-3741, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38758670

ABSTRACT

Across vertebrate species, the olfactory epithelium (OE) exhibits the uncommon feature of lifelong neuronal turnover. Epithelial stem cells give rise to new neurons that can adequately replace dying olfactory receptor neurons (ORNs) during developmental and adult phases and after lesions. To relay olfactory information from the environment to the brain, the axons of the renewed ORNs must reconnect with the olfactory bulb (OB). In Xenopus laevis larvae, we have previously shown that this process occurs between 3 and 7 weeks after olfactory nerve (ON) transection. In the present study, we show that after 7 weeks of recovery from ON transection, two functionally and spatially distinct glomerular clusters are reformed in the OB, akin to those found in non-transected larvae. We also show that the same odourant response tuning profiles observed in the OB of non-transected larvae are again present after 7 weeks of recovery. Next, we show that characteristic odour-guided behaviour disappears after ON transection but recovers after 7-9 weeks of recovery. Together, our findings demonstrate that the olfactory system of larval X. laevis regenerates with high accuracy after ON transection, leading to the recovery of odour-guided behaviour.


Subject(s)
Larva , Olfactory Bulb , Xenopus laevis , Animals , Olfactory Bulb/physiology , Nerve Regeneration/physiology , Odorants , Olfactory Nerve Injuries , Olfactory Nerve/physiology , Olfactory Mucosa/cytology , Olfactory Mucosa/physiology , Smell/physiology , Olfactory Receptor Neurons/physiology
4.
J Neurophysiol ; 129(6): 1515-1533, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37222431

ABSTRACT

Mitral/tufted cells (M/TCs), the principal output neuron classes form complex circuits with bulbar neurons and long-range centrifugal circuits with higher processing areas such as the horizontal limb of the diagonal band of Broca (HDB). The precise excitability of output neurons is sculpted by local inhibitory circuits. Here, light-gated cation channel channelrhodopsin-2 (ChR2) was expressed in HDB GABAergic neurons to investigate the short-term plasticity of evoked postsynaptic currents/potentials of HDB input to all classes of M/TCs and effects on firing in the acute slice preparation. Activation of the HDB directly inhibited all classes of output neurons exhibiting frequency-dependent short-term depression of evoked inhibitory postsynaptic current (eIPSC)/potential (eIPSP), resulting in decreased inhibition of responses to olfactory nerve input as a function of input frequency. In contrast, activation of an indirect circuit of HDB→interneurons→M/TCs induced frequency-dependent disinhibition, resulting in short-term facilitation of evoked excitatory postsynaptic current (eEPSC) eliciting a burst or cluster of spiking in M/TCs. The facilitatory effects of elevated HDB input frequency were strongest on deeper output neurons (deep tufted and mitral cells) and negligible on peripheral output neurons (external and superficial tufted cells). Taken together, GABAergic HDB activation generates frequency-dependent regulation that differentially affects the excitability and responses across the five classes of M/TCs. This regulation may help maintain the precise balance between inhibition and excitation of neuronal circuits across the populations of output neurons in the face of changes in an animal sniffing rate, putatively to enhance and sharpen the tuning specificity of individual or classes of M/TCs to odors.NEW & NOTEWORTHY Neuronal circuits in the olfactory bulb closely modulate olfactory bulb output activity. Activation of GABAergic circuits from the HDB to the olfactory bulb has both direct and indirect action differentially across the five classes of M/TC bulbar output neurons. The net effect enhances the excitability of deeper output neurons as HDB frequency increases, altering the relative inhibition-excitation balance of output circuits. We hypothesize that this sharpens the tuning specificity of classes of M/TCs to odors during sensory processing.


Subject(s)
Odorants , Olfactory Bulb , Animals , Olfactory Bulb/physiology , Sensation , Synaptic Potentials , Olfactory Nerve
5.
Eur J Neurosci ; 56(6): 4914-4929, 2022 09.
Article in English | MEDLINE | ID: mdl-35920370

ABSTRACT

Multiple-site optical recordings with NK2761, a voltage-sensitive absorption dye, were applied to the embryonic chick olfactory system, and the functional development of olfactory nerve (N.I)-related neural circuits was examined in the forebrain. The stimulation of the N. I elicited neural responses in N.I-olfactory bulb (OB)-forebrain preparations at the embryonic 8-12 day (E8-E12) stages. At the E11 stage, we functionally identified two circuits projecting from the OB to the forebrain. The first circuit passed through the ventral side of the forebrain and spread in the dorso-caudal direction, whereas the second circuit passed through the dorsal side to the first circuit. Pharmacological experiments showed that N-methyl-D -aspartate (NMDA) receptor function was more significant for the transfer of sensory information in these circuits. The functional development of N.I-related circuits was investigated, and the results obtained revealed that the ventral circuit was generated earlier than the dorsal circuit. Neural responses in the ventral circuit were detected from the E9 stage in normal physiological solution and the E8 stage in Mg2+ -free solution, which activated NMDA receptor function. At the E10 stage, neural responses in the dorsal circuit were clearly recognised in addition to ventral responses. We attempted to identify possible candidates for relay nuclei in the forebrain by comparing contour line maps of the optical signal amplitude with previously reported neuroanatomical data. The results suggest that N.I-related neural circuits from the periphery to the subpallium functionally mature earlier than those to the pallium during ontogenesis.


Subject(s)
Olfactory Nerve , Voltage-Sensitive Dye Imaging , Electric Stimulation/methods , Olfactory Bulb/physiology , Prosencephalon , Receptors, N-Methyl-D-Aspartate
6.
Stem Cells ; 39(5): 617-635, 2021 05.
Article in English | MEDLINE | ID: mdl-33470495

ABSTRACT

The olfactory epithelium (OE) possesses unique lifelong neuroregenerative capacities and undergoes constitutive neurogenesis throughout mammalian lifespan. Two populations of stem cells, frequently dividing globose basal cells (GBCs) and quiescent horizontal basal cells (HBCs), readily replace olfactory neurons throughout lifetime. Although lineage commitment and neuronal differentiation of stem cells has already been described in terms of transcription factor expression, little is known about external factors balancing between differentiation and self-renewal. We show here that expression of the CXC-motif chemokine receptor 4 (CXCR4) distinguishes both types of stem cells. Extensive colocalization analysis revealed exclusive expression of CXCR4 in proliferating GBCs and their neuronal progenies. Moreover, only neuronal lineage cells were derived from CXCR4-CreER-tdTomato reporter mice in the OE. Furthermore, Cre-tdTomato mice specific for HBCs (Nestin+ and Cytokeratin14+) did not reduce CXCR4 expression when bred to mice bearing floxed CXCR4 alleles, and did not show labeling of the neuronal cells. CXCR4 and its ligand CXCL12 were markedly upregulated upon induction of GBC proliferation during injury-induced regeneration. in vivo overexpression of CXCL12 did downregulate CXCR4 levels, which results in reduced GBC maintenance and neuronal differentiation. We proved that these effects were caused by CXCR4 downregulation rather than over-activation by showing that the phenotypes of CXCL12-overexpressing mice were highly similar to the phenotypes of CXCR4 knockout mice. Our results demonstrate functional CXCR4 signaling in GBCs regulates cell cycle exit and neural differentiation. We propose that CXCR4/CXCL12 signaling is an essential regulator of olfactory neurogenesis and provide new insights into the dynamics of neurogenesis in the OE.


Subject(s)
Chemokine CXCL12/genetics , Nerve Regeneration/genetics , Neurogenesis/genetics , Olfactory Nerve/growth & development , Receptors, CXCR4/genetics , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Gene Expression Regulation, Developmental/genetics , Keratin-14/genetics , Mice , Mice, Knockout , Nestin/genetics , Neural Stem Cells/cytology , Neurons/cytology , Olfactory Mucosa/growth & development , Olfactory Mucosa/injuries , Olfactory Nerve/metabolism
7.
Ultrastruct Pathol ; 46(5): 490-496, 2022 Sep 03.
Article in English | MEDLINE | ID: mdl-35913116

ABSTRACT

Schwannomas of the olfactory nerve are rare tumors: to the best of our knowledge, 56 cases have been previously reported. Here we describe a new patient presenting with an isolated olfactory schwannoma, highlighting the importance of multiple ancillary tests to approach the intracranial lesion of the anterior skull base, including electron microscopy. We also discuss the enigmatic origin of this entity, moving from the normal histology of the olfactory nerve compared to a peripheral nerve.


Subject(s)
Neurilemmoma , Olfactory Nerve , Humans , Microscopy, Electron , Neurilemmoma/diagnosis , Neurilemmoma/pathology , Olfactory Nerve/pathology
8.
J Neurosci Res ; 99(3): 750-777, 2021 03.
Article in English | MEDLINE | ID: mdl-33217763

ABSTRACT

Without protective and/or therapeutic agents the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection known as coronavirus disease 2019 is quickly spreading worldwide. It has surprising transmissibility potential, since it could infect all ages, gender, and human sectors. It attacks respiratory, gastrointestinal, urinary, hepatic, and endovascular systems and can reach the peripheral nervous system (PNS) and central nervous system (CNS) through known and unknown mechanisms. The reports on the neurological manifestations and complications of the SARS-CoV-2 infection are increasing exponentially. Herein, we enumerate seven candidate routes, which the mature or immature SARS-CoV-2 components could use to reach the CNS and PNS, utilizing the within-body cross talk between organs. The majority of SARS-CoV-2-infected patients suffer from some neurological manifestations (e.g., confusion, anosmia, and ageusia). It seems that although the mature virus did not reach the CNS or PNS of the majority of patients, its unassembled components and/or the accompanying immune-mediated responses may be responsible for the observed neurological symptoms. The viral particles and/or its components have been specifically documented in endothelial cells of lung, kidney, skin, and CNS. This means that the blood-endothelial barrier may be considered as the main route for SARS-CoV-2 entry into the nervous system, with the barrier disruption being more logical than barrier permeability, as evidenced by postmortem analyses.


Subject(s)
COVID-19/complications , COVID-19/metabolism , Central Nervous System/metabolism , Nervous System Diseases/etiology , Nervous System Diseases/metabolism , Peripheral Nervous System/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/virology , COVID-19/transmission , Central Nervous System/virology , Humans , Nervous System Diseases/virology , Olfactory Nerve/metabolism , Olfactory Nerve/virology , Peripheral Nervous System/virology
9.
Acta Neuropathol ; 141(6): 809-822, 2021 06.
Article in English | MEDLINE | ID: mdl-33903954

ABSTRACT

One of the most frequent symptoms of COVID-19 is the loss of smell and taste. Based on the lack of expression of the virus entry proteins in olfactory receptor neurons, it was originally assumed that the new coronavirus (severe acute respiratory syndrome coronavirus 2, SARS-CoV-2) does not infect olfactory neurons. Recent studies have reported otherwise, opening the possibility that the virus can directly infect the brain by traveling along the olfactory nerve. Multiple animal models have been employed to assess mechanisms and routes of brain infection of SARS-CoV-2, often with conflicting results. We here review the current evidence for an olfactory route to brain infection and conclude that the case for infection of olfactory neurons is weak, based on animal and human studies. Consistent brain infection after SARS-CoV-2 inoculation in mouse models is only seen when the virus entry proteins are expressed abnormally, and the timeline and progression of rare neuro-invasion in these and in other animal models points to alternative routes to the brain, other than along the olfactory projections. COVID-19 patients can be assured that loss of smell does not necessarily mean that the SARS-CoV-2 virus has gained access to and has infected their brains.


Subject(s)
Brain/virology , COVID-19/etiology , Olfactory Nerve/virology , Olfactory Receptor Neurons/virology , SARS-CoV-2/physiology , Virus Internalization , Animals , Disease Models, Animal , Humans
10.
Lupus ; 30(7): 1078-1085, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33858264

ABSTRACT

OBJECTIVE: To investigate the changes of olfactory function and odor-induced brain activation in patients with systemic lupus erythematosus (SLE) at early stages compared with healthy controls. MATERIALS AND METHODS: Olfactory function and odor-induced brain activation in 12 SLE patients at early stages and 12 age, gender and education matched healthy controls were evaluated using olfactory behavior test and odor-induced task-functional magnetic resonance imaging (task-fMRI). RESULTS: No significant differences in olfactory behavior scores (including olfactory threshold, olfactory identification, and olfactory memory) were found in the patients with SLE at early stages compared with the healthy controls, while significantly decreased odor-induced activations in olfactory-related brain regions were observed in the patients. In the SLE group, the patients with better performance in the olfactory threshold test had significantly lower levels of anti-dsDNA antibody. CONCLUSION: The current study demonstrated that significant alterations in odor-induced brain activations occurred prior to measurable olfactory decline in SLE at early stages, which provided a new method for early diagnosis of olfactory dysfunction in SLE.


Subject(s)
Brain/physiopathology , Lupus Erythematosus, Systemic/physiopathology , Olfaction Disorders/physiopathology , Olfactory Nerve/physiopathology , Adult , Antibodies, Antinuclear/blood , Brain/diagnostic imaging , Brain Mapping/methods , Case-Control Studies , Cognitive Dysfunction/complications , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/physiopathology , Early Diagnosis , Female , Humans , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/complications , Magnetic Resonance Imaging/methods , Neural Conduction/physiology , Neuropsychological Tests/standards , Olfaction Disorders/etiology , Prospective Studies , Sensory Thresholds/physiology , Severity of Illness Index
11.
Mol Cell Neurosci ; 102: 103450, 2020 01.
Article in English | MEDLINE | ID: mdl-31794879

ABSTRACT

Macrophage migration inhibitory factor (MIF) is an important regulator of innate immunity with key roles in neural regeneration and responses to pathogens, amongst a multitude of other functions. The expression of MIF and its binding partners has been characterised throughout the nervous system, with one key exception: the primary olfactory nervous system. Here, we showed in young mice (postnatal day 10) that MIF is expressed in the olfactory nerve by olfactory ensheathing glial cells (OECs) and by olfactory nerve fibroblasts. We also examined the expression of potential binding partners for MIF, and found that the serine protease HTRA1, known to be inhibited by MIF, was also expressed at high levels by OECs and olfactory fibroblasts in vivo and in vitro. We also demonstrated that MIF mediated segregation between OECs and J774a.1 cells (a monocyte/macrophage cell line) in co-culture, which suggests that MIF contributes to the fact that macrophages are largely absent from olfactory nerve fascicles. Phagocytosis assays of axonal debris demonstrated that MIF strongly stimulates phagocytosis by OECs, which indicates that MIF may play a role in the response of OECs to the continual turnover of olfactory axons that occurs throughout life.


Subject(s)
High-Temperature Requirement A Serine Peptidase 1/metabolism , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Neuroglia/metabolism , Olfactory Nerve/metabolism , Animals , Cell Line , Cells, Cultured , Fibroblasts/metabolism , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Nerve Regeneration , Olfactory Nerve/cytology , Olfactory Nerve/physiology , Phagocytosis , Protein Binding
12.
Acta Neurochir (Wien) ; 163(9): 2453-2457, 2021 09.
Article in English | MEDLINE | ID: mdl-34291382

ABSTRACT

BACKGROUND: Olfactory function preservation is a desirable objective in anterior skull base (ASB) surgery. The "infracerebral-supraolfactory nerve" corridor is presented. METHOD: The technique for preserving the olfactory nerves (OlfNs) in anterior ASB meningioma removal involves the following points: deep knowledge of the ASB vascular and meningeal anatomy, precise preoperative planning, wide and sharp dissection of the OlfNs away from the frontal lobes, gravity-aided frontal lobe retraction, Gelfoam-assisted hemostasis on nervous structures, and access to the lesion through an infracerebral-supraolfactory nerve corridor. CONCLUSIONS: This technique may be a valid option for patients affected by anterior skull base meningiomas with intact preoperative olfactory function.


Subject(s)
Meningeal Neoplasms , Meningioma , Skull Base Neoplasms , Humans , Meningeal Neoplasms/diagnostic imaging , Meningeal Neoplasms/surgery , Meningioma/diagnostic imaging , Meningioma/surgery , Neurosurgical Procedures , Olfactory Nerve/diagnostic imaging , Olfactory Nerve/surgery , Skull Base/diagnostic imaging , Skull Base/surgery , Skull Base Neoplasms/diagnostic imaging , Skull Base Neoplasms/surgery
13.
Glia ; 68(9): 1757-1774, 2020 09.
Article in English | MEDLINE | ID: mdl-32057144

ABSTRACT

Olfactory ensheathing cells (OECs) are unique glial cells with axonal growth-promoting properties in the olfactory epithelium and olfactory bulb, covering the entire length of the olfactory nerve. The proliferation of OECs is necessary for the formation of the presumptive olfactory nerve layer (ONL) during development and OECs transplantation. However, the molecular mechanism underlying the regulation of OEC proliferation in the ONL still remains unknown. In the present study, we examined the role of sphingosine 1-phosphate (S1P) and S1P receptors (S1PRs) on OEC proliferation. Initially, reverse transcription-PCR (RT-PCR), western blot and immunostaining revealed that S1PRs were highly expressed in the OECs in vitro and in vivo. Furthermore, we found that S1P treatment promoted the proliferation of primary cultured OECs mediated by S1PR1. Mechanistically, yes-associated protein (YAP) was required for S1P-induced OEC proliferation through RhoA signaling. Finally, conditional knockout of YAP in OECs reduced OEC proliferation in ONL, which impaired the axonal projection and growth of olfactory sensory neurons, and olfactory functions. Taken together, these results reveal a previously unrecognized function of S1P/RhoA/YAP pathway in the proliferation of OECs, contributing to the formation of ONL and the projection, growth, and function of olfactory sensory neurons during development.


Subject(s)
Neuroglia , Olfactory Nerve , Cell Proliferation , Cells, Cultured , Lysophospholipids , Olfactory Bulb , Sphingosine/analogs & derivatives
14.
Glia ; 68(3): 631-645, 2020 03.
Article in English | MEDLINE | ID: mdl-31696993

ABSTRACT

Astrocytes constitute the main glial component of the mammalian blood brain barrier (BBB). However, in the olfactory bulb (OB), the olfactory nerve layer (ONL) is almost devoid of astrocytes, raising the question which glial cells are part of the BBB. We used mice expressing EGFP in astrocytes and tdTomato in olfactory ensheathing cells (OECs), a specialized type of glial cells in the ONL, to unequivocally identify both glial cell types and investigate their contribution to the BBB in the olfactory bulb. OECs were located exclusively in the ONL, while somata of astrocytes were located in deeper layers and extended processes in the inner sublamina of the ONL. These processes surrounded blood vessels and contained aquaporin-4, an astrocytic protein enriched at the BBB. In the outer sublamina of the ONL, in contrast, blood vessels were surrounded by aquaporin-4-negative processes of OECs. Transcardial perfusion of blood vessels with lanthanum and subsequent visualization by electron microscopy showed that blood vessels enwrapped by OECs possessed intact tight junctions. In acute olfactory bulb preparations, injection of fluorescent glucose 6-NBDG into blood vessels resulted in labeling of OECs, indicating glucose transport from the perivascular space into OECs. In addition, Ca2+ transients in OECs in the outer sublamina evoked vasoconstriction, whereas Ca2+ signaling in OECs of the inner sublamina had no effect on adjacent blood vessels. Our results demonstrate that the BBB in the inner sublamina of the ONL contains astrocytes, while in the outer ONL OECs are part of the BBB.


Subject(s)
Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Olfactory Bulb/metabolism , Olfactory Nerve/pathology , Animals , Astrocytes/metabolism , Mice , Neuroglia/metabolism , Neurons/metabolism , Olfactory Bulb/pathology , Olfactory Nerve/metabolism
15.
J Neurophysiol ; 123(4): 1283-1294, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31891524

ABSTRACT

Sensory processing deficits are increasingly recognized as core symptoms of autism spectrum disorders (ASDs). However the molecular and circuit mechanisms that lead to sensory deficits are unknown. We show that two molecularly disparate mouse models of autism display similar deficits in sensory-evoked responses in the mouse olfactory system. We find that both Cntnap2- and Shank3-deficient mice of both sexes exhibit reduced response amplitude and trial-to-trial reliability during repeated odor presentation. Mechanistically, we show that both mouse models have weaker and fewer synapses between olfactory sensory nerve (OSN) terminals and olfactory bulb tufted cells and weaker synapses between OSN terminals and inhibitory periglomerular cells. Consequently, deficits in sensory processing provide an excellent candidate phenotype for analysis in ASDs.NEW & NOTEWORTHY The genetics of autism spectrum disorder (ASD) are complex. How the many risk genes generate the similar sets of symptoms that define the disorder is unknown. In particular, little is understood about the functional consequences of these genetic alterations. Sensory processing deficits are important aspects of the ASD diagnosis and may be due to unreliable neural circuits. We show that two mouse models of autism, Cntnap2- and Shank3-deficient mice, display reduced odor-evoked response amplitudes and reliability. These data suggest that altered sensory-evoked responses may constitute a circuit phenotype in ASDs.


Subject(s)
Autism Spectrum Disorder/physiopathology , Olfaction Disorders/physiopathology , Olfactory Bulb/physiopathology , Olfactory Nerve/physiopathology , Olfactory Perception/physiology , Perceptual Disorders/physiopathology , Synaptic Potentials/physiology , Animals , Calcium , Disease Models, Animal , Female , Male , Membrane Proteins/deficiency , Mice , Mice, Knockout , Microfilament Proteins/deficiency , Microscopy, Fluorescence, Multiphoton , Nerve Tissue Proteins/deficiency , Patch-Clamp Techniques , Phenotype
16.
J Neurosci Res ; 98(12): 2376-2383, 2020 12.
Article in English | MEDLINE | ID: mdl-32869376

ABSTRACT

Manifestation of neurological symptoms in certain patients of coronavirus disease-2019 (COVID-19) has warranted for their virus-induced etiogenesis. SARS-CoV-2, the causative agent of COVID-19, belongs to the genus of betacoronaviruses which also includes SARS-CoV-1 and MERS-CoV; causative agents for severe acute respiratory syndrome (SARS) in 2002 and Middle East respiratory syndrome (MERS) in 2012, respectively. Studies demonstrating the neural invasion of SARS-CoV-2 in vivo are still scarce, although such characteristics of certain other betacoronaviruses are well demonstrated in the literature. Based on the recent evidence for the presence of SARS-CoV-2 host cell entry receptors in specific components of the human nervous and vascular tissue, a neural (olfactory and/or vagal), and a hematogenous-crossing the blood-brain barrier, routes have been proposed. The neurological symptoms in COVID-19 may also arise as a consequence of the "cytokine storm" (characteristically present in severe disease) induced neuroinflammation, or co-morbidities. There is also a possibility that, there may be multiple routes of SARS-CoV-2 entry into the brain, or multiple mechanisms can be involved in the pathogenesis of the neurological symptoms. In this review article, we have discussed the possible routes of SARS-CoV-2 brain entry based on the emerging evidence for this virus, and that available for other betacoronaviruses in literature.


Subject(s)
Betacoronavirus/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Coronavirus Infections/metabolism , Nervous System Diseases/metabolism , Olfactory Nerve/metabolism , Pneumonia, Viral/metabolism , Animals , Blood-Brain Barrier/virology , Brain/virology , COVID-19 , Coronavirus Infections/complications , Coronavirus Infections/transmission , Humans , Nervous System Diseases/etiology , Olfactory Nerve/virology , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/transmission , SARS-CoV-2
17.
Chem Senses ; 45(3): 203-209, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32010939

ABSTRACT

Estrogen has been shown to affect differentiation and proliferation as a mitogen in various neural systems. Olfactory receptor cells are unique within the nervous system, and have the ability to regenerate even after an individual has reached maturity. Olfactory receptor cells also regenerate after experimentally induced degeneration. The purpose of this study is to observe the influence of estrogen depletion induced by ovariectomy on olfactory nerve regeneration. Female mice underwent bilateral ovariectomy at 8 weeks of age and received intraperitoneal administration of methimazole 1 week later. At 2, 4, and 6 weeks after methimazole administration, the olfactory mucosa was analyzed histochemically to determine olfactory epithelium (OE) thickness, olfactory marker protein distribution, and Ki-67 immunoreactivity. Furthermore, 2 weeks after ovariectomy, trkA protein distribution in the OE and nerve growth factor (NGF) levels in the olfactory bulb were determined by immunohistochemistry and enzyme-linked immunosorbent assay, respectively. Our results showed that in ovariectomized mice OMP, Ki-67, and trkA-immunopositive cells expression decreased at 2 weeks after methimazole injection, a time point at which regeneration is underway. At this same time point, although NGF production in the olfactory bulb had increased before methimazole administration, no differences were observed between the ovx and control groups. These results suggest that estrogen depletion induces a suppressive effect on regeneration of olfactory neurons, and that estrogen may have a potential use in the treatment of sensorineural olfactory dysfunction.


Subject(s)
Nerve Regeneration , Olfactory Nerve , Ovariectomy , Animals , Estrogens/pharmacology , Female , Mice , Mice, Inbred BALB C , Nerve Regeneration/drug effects , Olfactory Bulb/drug effects , Olfactory Bulb/pathology , Olfactory Mucosa/drug effects , Olfactory Mucosa/pathology , Olfactory Nerve/drug effects , Olfactory Nerve/surgery
18.
Brain Behav Evol ; 95(3-4): 139-161, 2020.
Article in English | MEDLINE | ID: mdl-33171468

ABSTRACT

The volume of the olfactory bulbs (OBs) relative to the brain has been used previously as a proxy for olfactory capabilities in many vertebrate taxa, including fishes. Although this gross approach has predictive power, a more accurate assessment of the number of afferent olfactory inputs and the convergence of this information at the level of the telencephalon is critical to our understanding of the role of olfaction in the behaviour of fishes. In this study, we used transmission electron microscopy to assess the number of first-order axons within the olfactory nerve (ON) and the number of second-order axons in the olfactory peduncle (OP) in established model species within cartilaginous (brownbanded bamboo shark, Chiloscyllium punctatum [CP]) and bony (common goldfish, Carassius auratus [CA]) fishes. The total number of axons varied from a mean of 18.12 ± 7.50 million in the ON to a mean of 0.38 ± 0.21 million in the OP of CP, versus 0.48 ± 0.16 million in the ON and 0.09 ± 0.02 million in the OP of CA. This resulted in a convergence ratio of approximately 50:1 and 5:1, respectively, for these two species. Based on astroglial ensheathing, axon type (unmyelinated [UM] and myelinated [M]) and axon size, we found no differentiated tracts in the OP of CP, whereas a lateral and a medial tract (both of which could be subdivided into two bundles or areas) were identified for CA, as previously described. Linear regression analyses revealed significant differences not only in axon density between species and locations (nerves and peduncles), but also in axon type and axon diameter (p < 0.05). However, UM axon diameter was larger in the OPs than in the nerve in both species (p = 0.005), with no significant differences in UM axon diameter in the ON (p = 0.06) between species. This study provides an in-depth analysis of the neuroanatomical organisation of the ascending olfactory pathway in two fish taxa and a quantitative anatomical comparison of the summation of olfactory information. Our results support the assertion that relative OB volume is a good indicator of the level of olfactory input and thereby a proxy for olfactory capabilities.


Subject(s)
Axons/ultrastructure , Goldfish/anatomy & histology , Olfactory Bulb/cytology , Olfactory Nerve/cytology , Olfactory Pathways/cytology , Sharks/anatomy & histology , Animals , Microscopy, Electron, Transmission , Olfactory Bulb/ultrastructure , Olfactory Cortex/cytology , Olfactory Nerve/ultrastructure , Olfactory Pathways/ultrastructure
19.
J Bioenerg Biomembr ; 51(1): 65-75, 2019 02.
Article in English | MEDLINE | ID: mdl-30604088

ABSTRACT

Most animals depend upon olfaction to find food, mates, and to avoid predators. An animal's olfactory circuit helps it sense its olfactory environment and generate critical behavioral responses. The general architecture of the olfactory circuit, which is conserved across species, is made up of a few different neuronal types including first-order receptor neurons, second- and third-order neurons, and local interneurons. Each neuronal type differs in their morphology, physiology, and neurochemistry. However, several recent studies have suggested that there is intrinsic diversity even among neurons of the same type and that this diversity is important for neural function. In this review, we first examine instances of intrinsic diversity observed among individual types of olfactory neurons. Next, we review potential genetic and experience-based plasticity mechanisms that underlie this diversity. Finally, we consider the implications of intrinsic neuronal diversity for circuit function. Overall, we hope to highlight the importance of intrinsic diversity as a previously underestimated property of circuit function.


Subject(s)
Olfactory Nerve/cytology , Animals , Humans , Interneurons , Neuronal Plasticity , Olfactory Receptor Neurons
20.
Mol Cell Neurosci ; 92: 27-39, 2018 10.
Article in English | MEDLINE | ID: mdl-29940213

ABSTRACT

Olfactory ensheathing cells (OECs) migrate from olfactory epithelium towards olfactory bulb (OB), contributing to formation of the presumptive olfactory nerve layer during development. However, it remains unclear that molecular mechanism of regulation of OEC migration in OB. In the present study, we found that OECs highly expressed the receptors of semaphorin 3A (Sema3A) in vitro and in vivo, whereas Sema3A displayed a gradient expression pattern with higher in inner layer of OB and lower in outer layer of OB. Furthermore, the collapse assays, Boyden chamber migration assays and single-cell migration assays showed that Sema3A induced the collapse of leading front of OECs and inhibited OEC migration. Thirdly, the leading front of OECs exhibited adaptation in a protein synthesis-independent manner, and endocytosis-dependent manner during Sema3A-induced OEC migration. Finally, Sema3A-induced collapse of leading front was required the decrease of focal adhesion and a retrograde F-actin flow in a cofilin activation-dependent manner. Taken together, these results demonstrate that Sema3A as an inhibitive migratory factor for OEC migration through cofilin activation is involved in the formation of olfactory nerve layer.


Subject(s)
Cell Movement , Olfactory Nerve/cytology , Semaphorin-3A/metabolism , Actin Depolymerizing Factors/metabolism , Actins/metabolism , Animals , Cells, Cultured , Male , Neurogenesis , Neuroglia/cytology , Neuroglia/metabolism , Olfactory Nerve/metabolism , Rats , Rats, Sprague-Dawley , Semaphorin-3A/genetics
SELECTION OF CITATIONS
SEARCH DETAIL