Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Viruses ; 13(8)2021 08 20.
Article in English | MEDLINE | ID: mdl-34452517

ABSTRACT

Patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019, suffer from respiratory and non-respiratory symptoms. Among these symptoms, the loss of smell has attracted considerable attention. The objectives of this study were to determine which cells are infected, what happens in the olfactory system after viral infection, and how these pathologic changes contribute to olfactory loss. For this purpose, Syrian golden hamsters were used. First, we verified the olfactory structures in the nasal cavity of Syrian golden hamsters, namely the main olfactory epithelium, the vomeronasal organ, and their cellular components. Second, we found angiotensin-converting enzyme 2 expression, a receptor protein of SARS-CoV-2, in both structures and infections of supporting, microvillar, and solitary chemosensory cells. Third, we observed pathological changes in the infected epithelium, including reduced thickness of the mucus layer, detached epithelia, indistinct layers of epithelia, infiltration of inflammatory cells, and apoptotic cells in the overall layers. We concluded that a structurally and functionally altered microenvironment influences olfactory function. We observed the regeneration of the damaged epithelium, and found multilayers of basal cells, indicating that they were activated and proliferating to reconstitute the injured epithelium.


Subject(s)
COVID-19/virology , Chemoreceptor Cells/virology , Olfactory Mucosa/virology , SARS-CoV-2 , Vomeronasal Organ/virology , Angiotensin-Converting Enzyme 2/metabolism , Animals , COVID-19/pathology , Chemoreceptor Cells/pathology , Male , Mesocricetus , Nasal Cavity/pathology , Nasal Cavity/virology , Olfactory Mucosa/metabolism , Olfactory Mucosa/pathology , Olfactory Receptor Neurons/metabolism , Olfactory Receptor Neurons/pathology , Olfactory Receptor Neurons/virology , Receptors, Coronavirus/metabolism , Regeneration , SARS-CoV-2/isolation & purification , Vomeronasal Organ/metabolism , Vomeronasal Organ/pathology
2.
Int J Mol Sci ; 22(12)2021 Jun 12.
Article in English | MEDLINE | ID: mdl-34204595

ABSTRACT

Among all the proposed pathogenic mechanisms to understand the etiology of Alzheimer's disease (AD), increased oxidative stress seems to be a robust and early disease feature where many of those hypotheses converge. However, despite the significant lines of evidence accumulated, an effective diagnosis and treatment of AD are not yet available. This limitation might be partially explained by the use of cellular and animal models that recapitulate partial aspects of the disease and do not account for the particular biology of patients. As such, cultures of patient-derived cells of peripheral origin may provide a convenient solution for this problem. Peripheral cells of neuronal lineage such as olfactory neuronal precursors (ONPs) can be easily cultured through non-invasive isolation, reproducing AD-related oxidative stress. Interestingly, the autofluorescence of key metabolic cofactors such as reduced nicotinamide adenine dinucleotide (NADH) can be highly correlated with the oxidative state and antioxidant capacity of cells in a non-destructive and label-free manner. In particular, imaging NADH through fluorescence lifetime imaging microscopy (FLIM) has greatly improved the sensitivity in detecting oxidative shifts with minimal intervention to cell physiology. Here, we discuss the translational potential of analyzing patient-derived ONPs non-invasively isolated through NADH FLIM to reveal AD-related oxidative stress. We believe this approach may potentially accelerate the discovery of effective antioxidant therapies and contribute to early diagnosis and personalized monitoring of this devastating disease.


Subject(s)
Alzheimer Disease/pathology , Microscopy, Fluorescence/methods , NAD/metabolism , Olfactory Receptor Neurons/pathology , Oxidative Stress , Animals , Antioxidants/metabolism , Humans
3.
Neurosci Lett ; 748: 135694, 2021 03 23.
Article in English | MEDLINE | ID: mdl-33600902

ABSTRACT

Patients with COVID-19 often complain of smell and taste disorders (STD). STD emerge early in the course of the disease, seem to be more common in SARS-CoV-2 infection than in other upper respiratory tract infections, and could in some cases persist for long after resolution of respiratory symptoms. Current evidence suggests that STD probably result from a loss of function of olfactory sensory neurons and taste buds, mainly caused by infection, inflammation, and subsequent dysfunction of supporting non-neuronal cells in the mucosa. However, the possible occurrence of other mechanisms leading to chemosensory dysfunction has also been hypothesized, and contrasting data have been reported regarding the direct infection of sensory neurons by SARS-CoV-2. In this mini-review, we summarize the currently available literature on pathogenesis, clinical manifestations, diagnosis, and outcomes of STD in COVID-19 and discuss possible future directions of research on this topic.


Subject(s)
COVID-19/complications , Olfaction Disorders/etiology , SARS-CoV-2/pathogenicity , Taste Disorders/etiology , COVID-19/immunology , COVID-19/virology , Humans , Mouth Mucosa/immunology , Mouth Mucosa/pathology , Olfaction Disorders/diagnosis , Olfaction Disorders/epidemiology , Olfaction Disorders/physiopathology , Olfactory Mucosa/immunology , Olfactory Mucosa/pathology , Olfactory Receptor Neurons/immunology , Olfactory Receptor Neurons/pathology , SARS-CoV-2/immunology , Smell/physiology , Taste/physiology , Taste Buds/immunology , Taste Buds/pathology , Taste Disorders/diagnosis , Taste Disorders/epidemiology , Taste Disorders/physiopathology
4.
ACS Chem Neurosci ; 12(4): 589-595, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33522795

ABSTRACT

Olfactory dysfunction is one of the most frequent and specific symptoms of coronavirus disease 2019 (COVID-19). Information on the damage and repair of the neuroepithelium and its impact on olfactory function after COVID-19 is still incomplete. While severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) causes the ongoing worldwide outbreak of COVID-19, little is known about the changes triggered by SARS-CoV-2 in the olfactory epithelium (OE) at the cellular level. Here, we report profiles of the OE after SARS-CoV-2 infection in golden Syrian hamsters, which is a reliable animal model of COVID-19. We observed severe damage in the OE as early as 3 days postinoculation and regionally specific damage and regeneration of the OE within the nasal cavity; the nasal septal region demonstrated the fastest recovery compared to other regions in the nasal turbinates. These findings suggest that anosmia related to SARS-CoV-2 infection may be fully reversible.


Subject(s)
Anosmia/physiopathology , COVID-19/pathology , Olfactory Mucosa/pathology , Olfactory Receptor Neurons/pathology , Regeneration , SARS-CoV-2 , Animals , Anosmia/etiology , COVID-19/complications , COVID-19/physiopathology , Disease Models, Animal , Mesocricetus , Nasal Cavity , Nasal Septum , Olfactory Mucosa/physiology , Olfactory Receptor Neurons/physiology , Organ Size , Turbinates
5.
J Neurochem ; 157(4): 930-943, 2021 05.
Article in English | MEDLINE | ID: mdl-32970861

ABSTRACT

Olfactory disorders have been increasingly reported in individuals infected with SARS-CoV-2, the virus causing the coronavirus disease 2019 (COVID-19). Losing the sense of smell has a strong impact on the quality of life, since it may lead to malnutrition, weight loss, food poisoning, depression, and exposure to dangerous chemicals. Individuals who suffer from anosmia (inability to smell) also cannot sense the flavor of food, which is a combination of taste and smell. Interestingly, infected individuals have reported sudden loss of smell with no congested nose, as is frequently observed in common colds or other upper respiratory tract infections. These observations suggest that SARS-CoV-2 infection leads to olfactory loss through a distinct mechanism, which is still unclear. This article provides an overview of olfactory loss and the recent findings relating to COVID-19. Possible mechanisms of SARS-CoV-2-induced olfactory loss are also discussed.


Subject(s)
COVID-19/complications , Olfaction Disorders/etiology , Virus Diseases/complications , Humans , Olfaction Disorders/pathology , Olfactory Receptor Neurons/pathology
6.
Brain Behav Immun ; 89: 579-586, 2020 10.
Article in English | MEDLINE | ID: mdl-32629042

ABSTRACT

Anosmia is one of the most prevalent symptoms of SARS-CoV-2 infection during the COVID-19 pandemic. However, the cellular mechanism behind the sudden loss of smell has not yet been investigated. The initial step of odour detection takes place in the pseudostratified olfactory epithelium (OE) mainly composed of olfactory sensory neurons surrounded by supporting cells known as sustentacular cells. The olfactory neurons project their axons to the olfactory bulb in the central nervous system offering a potential pathway for pathogens to enter the central nervous system by bypassing the blood brain barrier. In the present study, we explored the impact of SARS-CoV-2 infection on the olfactory system in golden Syrian hamsters. We observed massive damage of the OE as early as 2 days post nasal instillation of SARS-CoV-2, resulting in a major loss of cilia necessary for odour detection. These damages were associated with infection of a large proportion of sustentacular cells but not of olfactory neurons, and we did not detect any presence of the virus in the olfactory bulbs. We observed massive infiltration of immune cells in the OE and lamina propria of infected animals, which may contribute to the desquamation of the OE. The OE was partially restored 14 days post infection. Anosmia observed in COVID-19 patient is therefore likely to be linked to a massive and fast desquamation of the OE following sustentacular cells infection with SARS-CoV-2 and subsequent recruitment of immune cells in the OE and lamina propria.


Subject(s)
Coronavirus Infections/pathology , Olfactory Bulb/pathology , Olfactory Mucosa/pathology , Pneumonia, Viral/pathology , Animals , Betacoronavirus , COVID-19 , Cilia/pathology , Coronavirus Infections/physiopathology , Mesocricetus , Olfaction Disorders/pathology , Olfaction Disorders/physiopathology , Olfactory Bulb/virology , Olfactory Mucosa/virology , Olfactory Receptor Neurons/pathology , Olfactory Receptor Neurons/virology , Pandemics , Pneumonia, Viral/physiopathology , SARS-CoV-2
7.
Med Hypotheses ; 143: 109881, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32474381

ABSTRACT

Since the outbreak of Coronavirus Disease 2019 (COVID-19), loss of smell has increasingly been reported as a frequent clinical sign. Understanding the underlying mechanism and the prognostic value of this symptom will help better manage patients. SARS-CoV-2, as SARS-CoV-1, may likely spread to the central nervous system (CNS) via the olfactory nerve, a known gateway for respiratory neurotropic viruses. We hypothesise that sudden loss of smell due to COVID-19 is the consequence of a protective host defence mechanism involving apoptosis of olfactory receptor neurons. Sacrificing smelling over neuroprotection is a logical strategy, even more so as olfaction is the only sense with the ability to regenerate in adults. Induced apoptosis of olfactory neurons has been shown in mice, successfully preventing neuroinvasion. On the other hand, adult olfactory neurogenesis has been shown to be regulated in part by the immune system, allowing to restore olfactory function. Understanding anosmia as part of a defence mechanism would support the concept of sudden anosmia as being a positive prognostic factor in the short term. Also, it may orient research to investigate the risk of future neurodegenerative disease linked to persisting coronavirus in neurons.


Subject(s)
Betacoronavirus , Coronavirus Infections/complications , Olfaction Disorders/etiology , Pandemics , Pneumonia, Viral/complications , Animals , Apoptosis , Betacoronavirus/immunology , Betacoronavirus/pathogenicity , COVID-19 , Coronavirus Infections/immunology , Coronavirus Infections/physiopathology , Humans , Mice , Models, Immunological , Models, Neurological , Olfaction Disorders/immunology , Olfaction Disorders/physiopathology , Olfactory Receptor Neurons/pathology , Olfactory Receptor Neurons/physiology , Phenotype , Pneumonia, Viral/immunology , Pneumonia, Viral/physiopathology , Prognosis , SARS-CoV-2
8.
Prion ; 14(1): 31-41, 2020 12.
Article in English | MEDLINE | ID: mdl-31950869

ABSTRACT

Adult neurogenesis, analogous to early development, is comprised of several, often concomitant, processes including proliferation, differentiation, and formation of synaptic connections. However, due to continual, asynchronous turn-over, newly-born adult olfactory sensory neurons (OSNs) must integrate into existing circuitry. Additionally, OSNs express high levels of cellular prion protein (PrPC), particularly in the axon, which implies a role in this cell type. The cellular prion has been shown to be important for proper adult OSN neurogenesis primarily by stabilizing mature olfactory neurons within this circuitry. However, the role of PrPC on each specific adult neurogenic processes remains to be investigated in detail. To tease out the subtle effects of prion protein expression level, a large population of regenerating neurons must be investigated. The thyroid drug methimazole (MTZ) causes nearly complete OSN loss in rodents and is used as a model of acute olfactory injury, providing a mechanism to induce synchronized OSN regeneration. This study investigated the effect of PrPC on adult neurogenesis after acute nasotoxic injury. Altered PrPC levels affected olfactory sensory epithelial (OSE) regeneration, cell proliferation, and differentiation. Attempts to investigate the role of PrPC level on axon regeneration did not support previous studies, and glomerular targeting did not recover to vehicle-treated levels, even by 20 weeks. Together, these studies demonstrate that the cellular prion protein is critical for regeneration of neurons, whereby increased PrPC levels promote early neurogenesis, and that lack of PrPC delays the regeneration of this tissue after acute injury.


Subject(s)
Nerve Regeneration/physiology , Olfactory Receptor Neurons/pathology , Prion Proteins/metabolism , Acute Disease , Animals , Axons/drug effects , Axons/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Male , Methimazole/toxicity , Mice, Transgenic , Nerve Regeneration/drug effects , Neurogenesis/drug effects , Olfactory Mucosa/drug effects , Olfactory Mucosa/pathology , Olfactory Receptor Neurons/drug effects
9.
Anat Rec (Hoboken) ; 303(3): 626-633, 2020 03.
Article in English | MEDLINE | ID: mdl-30632702

ABSTRACT

The olfactory epithelium (OE) is the peripheral organ for the sense of smell, housing primary sensory neurons that project axons from the nose to the brain. Due to the presence of a basal stem cell niche, the adult mammalian OE is a dynamic tissue capable of replacing neurons following their loss. Nonetheless, certain conditions, such as blunt head trauma, can result in persistent olfactory loss, thought to be due to shearing of olfactory nerve filaments at the skull base, degeneration, and failures in proper regeneration/reinnervation. The identification of new treatment strategies aimed at preventing degeneration of olfactory neurons is, therefore, needed. In considering potential therapies, we have focused on N-acetylcysteine (NAC), a glutathione substrate shown to be neuroprotective, with a record of safe clinical use. Here, we have tested the use of NAC in an animal model of olfactory degeneration. Administered acutely, we found that NAC (100 mg/kg, twice daily) resulted in a reduction of olfactory neuronal loss from the OE of the nose following surgical ablation of the olfactory bulb. At 1 week postlesion, we identified 54 ± 8.1 mature neurons per 0.5 mm epithelium in NAC-treated animals vs. 28 ± 4.2 in vehicle-treated controls (P = 0.02). Furthermore, in an olfactory cell culture model, we have identified significant alterations in the expression of several genes involved in oxidative stress pathways following NAC exposure. Our results provide evidence supporting the potential therapeutic utility for NAC acutely following head trauma-induced olfactory loss. Anat Rec, 303:626-633, 2020. © 2019 American Association for Anatomy.


Subject(s)
Acetylcysteine/therapeutic use , Nerve Degeneration/drug therapy , Neuroprotective Agents/therapeutic use , Olfactory Mucosa/drug effects , Olfactory Receptor Neurons/drug effects , Acetylcysteine/pharmacology , Animals , Cell Survival/drug effects , Mice , Nerve Degeneration/pathology , Neuroprotective Agents/pharmacology , Olfactory Bulb/injuries , Olfactory Mucosa/pathology , Olfactory Receptor Neurons/pathology
10.
Stem Cell Reports ; 12(6): 1354-1365, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31155504

ABSTRACT

Stem cell-based therapies have been proposed as a strategy to replace damaged tissues, especially in the nervous system. A primary sensory modality, olfaction, is impaired in 12% of the US population, but lacks treatment options. We report here the development of a novel mouse model of inducible hyposmia and demonstrate that purified tissue-specific stem cells delivered intranasally engraft to produce olfactory neurons, achieving recovery of function. Adult mice were rendered hyposmic by conditional deletion of the ciliopathy-related IFT88 gene in the olfactory sensory neuron lineage and following experimentally induced olfactory injury, received either vehicle or stem cell infusion intranasally. Engraftment-derived olfactory neurons were identified histologically, and functional improvements were measured via electrophysiology and behavioral assay. We further explored mechanisms in culture that promote expansion of engraftment-competent adult olfactory basal progenitor cells. These findings provide a basis for translational research on propagating adult tissue-specific sensory progenitor cells and testing their therapeutic potential.


Subject(s)
Ciliopathies , Neural Stem Cells , Olfaction Disorders , Olfactory Receptor Neurons , Smell , Stem Cell Transplantation , Animals , Benzilates , Ciliopathies/genetics , Ciliopathies/metabolism , Ciliopathies/pathology , Ciliopathies/therapy , Mice, Transgenic , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neural Stem Cells/transplantation , Olfaction Disorders/genetics , Olfaction Disorders/metabolism , Olfaction Disorders/pathology , Olfaction Disorders/therapy , Olfactory Receptor Neurons/metabolism , Olfactory Receptor Neurons/pathology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
11.
Biochem Biophys Res Commun ; 513(3): 635-641, 2019 06 04.
Article in English | MEDLINE | ID: mdl-30981508

ABSTRACT

Clinical data show that part of patients with sinonasal diseases suffered from olfactory dysfunction, especially with allergic rhinitis (AR) and chronic rhinosinusitis (CRS). However, the mechanisms responsible for AR-induced olfactory loss are still largely unknown. Because of the difficulty to obtain human olfactory mucosa, an AR-induced olfactory loss animal model needs to be constructed to clarify the mechanism. The AR mouse model was induced by intraperitoneal sensitizing with ovalbumin (OVA) followed by intranasal challenge lasted from 1 to 12 weeks. For groups with recovery, mice were housed for another 4-week long without any treatment after the last intranasal challenge. Olfactory function, olfactory receptor neurons (ORNs) density and leukocytes infiltration were examined at different time points. Olfactory loss occurs immediately after 1-week intranasal challenge and deteriorates almost to anosmia after 8th week, and after that olfactory loss become irreversible. Nasal inflammation induces significant infiltration of leukocytes into olfactory epithelium (OE), which negatively correlated with the density of ORNs and mouse olfaction in a time dependent manner. The neutrophilic subtype dominates in number amongst the total infiltrated leukocytes, indicating its pivotal role in nasal inflammation-induced olfactory dysfunction. In this study, we constructed a persistent AR-induced olfactory loss mouse model, losing the ability to recover from dysfunction if the disease duration more than eight weeks, which implies that timely treatments are necessary. Meanwhile, this mouse model could provide an easy and reliable system to clarify the mechanisms of AR-induced irreversible olfactory dysfunction.


Subject(s)
Olfaction Disorders/etiology , Rhinitis, Allergic/complications , Animals , Disease Models, Animal , Leukocytes/pathology , Male , Mice , Mice, Inbred BALB C , Olfaction Disorders/physiopathology , Olfactory Receptor Neurons/pathology , Ovalbumin , Rhinitis, Allergic/chemically induced , Rhinitis, Allergic/physiopathology , Smell
12.
Artif Cells Nanomed Biotechnol ; 47(1): 564-570, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30857434

ABSTRACT

Developing a biomaterial that promotes regeneration of both respiratory epithelium (RE) and olfactory neuroepithelium (ON) improves the surgical outcome of endoscopic sinus surgery. Although chitosan (CS) inhibits mucociliary differentiation of RE, it has been reported to regenerate ON. In addition, hyaluronic acid (HA) has been demonstrated to promote regeneration of RE. Whether the composite CS + HA would simultaneously benefit RE and ON remains unexplored. Human nasal respiratory epithelial cells (RECs) and olfactory neuroepithelial cells (ONCs) are respectively obtained from the RE and the ON. They are cultured in vitro and divided into groups undergoing four treatments, control, CS, HA, and CS + HA and assessed by scanning electron microscope, immunocytochemistry, and Western blots following indicated growth conditions. RECs keep polygonal morphology with mucociliary differentiation in the CS + HA group. The levels of E-cadherin, zonula occludens-1, mucin 5AC, and forkhead box protein J1 are significantly higher in the CS + HA group than in the CS alone group. In addition, ONCs express lower cytokeratin 18 (CK18) and higher olfactory marker protein (OMP) in the CS + HA group than in HA alone group. ONCs express more signal transduction apparatuses, adenylate cyclase 3, in CS and CS + HA groups than in HA and controls. Chitosan-hyaluronan plays a part in promoting differentiation of ORNs and facilitating mucociliary differentiation of RECs. This composite is a promising biomaterial for the sinonasal application.


Subject(s)
Cell Differentiation/drug effects , Chitosan/pharmacology , Hyaluronic Acid/pharmacology , Nasal Mucosa/drug effects , Olfactory Receptor Neurons/drug effects , Cells, Cultured , Chronic Disease , Humans , Nasal Mucosa/pathology , Olfactory Receptor Neurons/pathology , Rhinitis/pathology , Signal Transduction/drug effects
13.
Sci Rep ; 8(1): 17300, 2018 11 23.
Article in English | MEDLINE | ID: mdl-30470811

ABSTRACT

Exercise (Ex) and caloric restriction (CR) reduce oxidative stress and improve organ function. For instance, voluntary Ex or CR is known to reduce age-related cochlear damage in male C57BL/6J mice. However, the effect of Ex and CR on the olfactory system is unknown. In this study, we confirmed the positive effect of Ex and CR on age-related cochlear damage, but found that Ex and CR affected negatively cell dynamics in the olfactory epithelium (OE) by reducing the number of mature olfactory sensory neurons (OSNs) and increasing the number of proliferative basal cells and apoptotic OSNs in the dorsal zone of the olfactory epithelium (OE), which contains neurons expressing NADPH quinone oxido-reductase 1 (NQO1). In addition, these interventions resulted in lower odor-induced c-fos expression in areas of the olfactory bulb receiving projections from dorsal-zone OSNs than in areas receiving ventral-zone projections. Further, we observed substantial oxidative stress in NQO1-positive cells and apoptotic OSNs in the dorsal zone in Ex and CR animals. These results suggest that, in contrast to their positive effects in other organs, Ex and CR facilitate oxidative stress and negatively impact structure and function in dorsal-zone OSNs, probably in association with NQO1 bioactivation.


Subject(s)
Caloric Restriction , NAD(P)H Dehydrogenase (Quinone)/metabolism , Olfactory Mucosa/pathology , Olfactory Receptor Neurons/pathology , Physical Conditioning, Animal , Animals , Male , Mice , Mice, Inbred C57BL , Olfactory Mucosa/metabolism , Olfactory Receptor Neurons/metabolism , Time Factors
14.
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
15.
J Neurosci ; 38(31): 6806-6824, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29934351

ABSTRACT

The olfactory epithelium (OE) of vertebrates is a highly regenerative neuroepithelium that is maintained under normal conditions by a population of stem and progenitor cells, globose basal cells (GBCs), which also contribute to epithelial reconstitution after injury. However, aging of the OE often leads to neurogenic exhaustion, the disappearance of both GBCs and olfactory sensory neurons (OSNs). Aneuronal tissue may remain as olfactory, with an uninterrupted sheet of apically arrayed microvillar-capped sustentacular cell, or may undergo respiratory metaplasia. We have generated a transgenic mouse model for neurogenic exhaustion using olfactory marker protein-driven Tet-off regulation of the A subunit of Diphtheria toxin such that the death of mature OSNs is accelerated. At as early as 2 months of age, the epithelium of transgenic mice, regardless of sex, recapitulates what is seen in the aged OE of humans and rodents. Areas of the epithelium completely lack neurons and GBCs; whereas the horizontal basal cells, a reserve stem cell population, show no evidence of activation. Surprisingly, other areas that were olfactory undergo respiratory metaplasia. The impact of accelerated neuronal death and reduced innervation on the olfactory bulb (OB) was also examined. Constant neuronal turnover leaves glomeruli shrunken and affects the dopaminergic interneurons in the periglomerular layer. Moreover, the acceleration of OSN death can be reversed in those areas where some GBCs persist. However, the projection onto the OB recovers incompletely and the reinnervated glomeruli are markedly altered. Therefore, the capacity for OE regeneration is tempered when GBCs disappear.SIGNIFICANCE STATEMENT A large percentage of humans lose or suffer a significant decline in olfactory function as they age. Therefore, quality of life suffers and safety and nutritional status are put at risk. With age, the OE apparently becomes incapable of fully maintaining the neuronal population of the epithelium despite its well known capacity for recovering from most forms of injury when younger. Efforts to identify the mechanism by which olfactory neurogenesis becomes exhausted with age require a powerful model for accelerating age-related tissue pathology. The current OMP-tTA;TetO-DTA transgenic mouse model, in which olfactory neurons die when they reach maturity and accelerated death can be aborted to assess the capacity for structural recovery, satisfies that need.


Subject(s)
Aging/physiology , Nerve Regeneration/physiology , Neural Stem Cells/cytology , Neurogenesis , Olfactory Mucosa/cytology , Olfactory Receptor Neurons/cytology , Aged , Aged, 80 and over , Animals , Diphtheria Toxin/genetics , Diphtheria Toxin/toxicity , Female , Humans , Inflammation , Male , Mice , Mice, Transgenic , Middle Aged , Nerve Degeneration/chemically induced , Olfaction Disorders/etiology , Olfaction Disorders/pathology , Olfactory Mucosa/growth & development , Olfactory Receptor Neurons/pathology , Peptide Fragments/genetics , Peptide Fragments/toxicity , Severity of Illness Index
16.
Neurotox Res ; 34(3): 353-362, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29667127

ABSTRACT

Chloroform-induced olfactory mucosal degeneration has been reported in adult rats following gavage. We used fixed-point chloroform infusions on different postnatal days (PNDs) to investigate the effects of early olfactory bilateral deprivation on the main olfactory bulbs in Sprague Dawley rats. The experimental groups included rats infused with chloroform (5 µl) or saline (sham, 5 µl) on PNDs 3 and 8, and rats not receiving infusions (control) (n = 6 in all groups). Rats receiving chloroform on PND 3 showed significant hypoevolutism when compared to those in other groups (P < 0.05). There was a complete disappearance and a significant reduction in the size of olfactory glomeruli in the PND 3 and 8 groups, respectively, when compared to the respective sham groups. Rats receiving chloroform on PND 3 had significant memory impairment (P < 0.01) and increased levels of learned helplessness (P < 0.05), as measured using the Morris water maze and tail suspension tests, respectively. GABAA receptor alpha5 subunit (α5GABAAR) expression in hippocampal neurons was significantly lower in rats receiving chloroform on PND 3 than in rats in other groups (P < 0.01), as measured using immunohistochemistry and polymerase chain reaction. There was thus a critical period for the preservation of regenerative ability in olfactory receptor neurons, during which damage and olfactory deprivation led to altered rhinencephalon structure and disappearance of olfactory glomeruli, which induced hypoevolutism. Olfactory deprivation after the critical period had no significant effect on olfactory receptor neuron regeneration, leading to reduced developmental and behavioral effects in Sprague Dawley rats.


Subject(s)
Hippocampus/metabolism , Olfaction Disorders/pathology , Olfactory Mucosa/pathology , Olfactory Receptor Neurons/pathology , Receptors, GABA-A/metabolism , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Chloroform/toxicity , Depression/etiology , Female , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Hindlimb Suspension , Male , Maze Learning/drug effects , Olfaction Disorders/chemically induced , Olfactory Mucosa/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/genetics , Sensory Deprivation , Solvents/toxicity
17.
Neuroscience ; 371: 106-118, 2018 02 10.
Article in English | MEDLINE | ID: mdl-29203228

ABSTRACT

Despite the regenerative capacity of the olfactory bulb (OB), head trauma causes olfactory disturbances in up to 30% of patients. While models of olfactory nerve transection, olfactory receptor neuron (ORN) ablation, or direct OB impact have been used to examine OB recovery, these models are severe and not ideal for study of OB synaptic repair. We posited that a mild fluid percussion brain injury (mFPI), delivered over mid-dorsal cortex, would produce diffuse OB deafferentation without confounding pathology. Wild type FVB/NJ mice were subjected to mFPI and OB probed for ORN axon degeneration and onset of reactive synaptogenesis. OB extracts revealed 3 d postinjury elevation of calpain-cleaved 150-kDa αII-spectrin, an indicator of axon damage, in tandem with reduced olfactory marker protein (OMP), a protein specific to intact ORN axons. Moreover, mFPI also produced a 3-d peak in GFAP+ astrocyte and IBA1+ microglial reactivity, consistent with postinjury inflammation. OB glomeruli showed disorganized ORN axons, presynaptic degeneration, and glial phagocytosis at 3 and 7 d postinjury, all indicative of deafferentation. At 21 d after mFPI, normal synaptic structure re-emerged along with OMP recovery, supporting ORN afferent reinnervation. Robust 21 d postinjury upregulation of GAP-43 was consistent with the time course of ORN axon sprouting and synapse regeneration reported after more severe olfactory insult. Together, these findings define a cycle of synaptic degeneration and recovery at a site remote to non-contusive brain injury. We show that mFPI models diffuse ORN axon damage, useful for the study of time-dependent reactive synaptogenesis in the deafferented OB.


Subject(s)
Axons/pathology , Axons/physiology , Brain Concussion/pathology , Brain Concussion/physiopathology , Olfactory Bulb/pathology , Olfactory Bulb/physiopathology , Animals , Astrocytes/pathology , Astrocytes/physiology , Disease Models, Animal , GAP-43 Protein/metabolism , Male , Mice , Microglia/pathology , Microglia/physiology , Nerve Regeneration/physiology , Neuronal Plasticity/physiology , Olfactory Marker Protein/metabolism , Olfactory Receptor Neurons/pathology , Olfactory Receptor Neurons/physiology , Random Allocation , Spectrin/metabolism , Synapses/pathology , Synapses/physiology , Time Factors
18.
Sci Rep ; 7(1): 11605, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28912588

ABSTRACT

The olfactory mucosa (OM) is exposed to environmental agents and therefore vulnerable to inflammation. To examine the effects of environmental toxin-initiated OM inflammation on the olfactory bulb (OB), we induced persistent rhinitis in mice and analyzed the spatial and temporal patterns of histopathological changes in the OM and OB. Mice received unilateral intranasal administration of lipopolysaccharide (LPS) or saline three times per week, and were immunohistologically analyzed at 1, 3, 7, 14 and 21 days after the first administration. LPS administration induced an inflammatory response in the OM, including the infiltration of Ly-6G-, CD11b-, Iba-1- and CD3-positive cells, the production of interleukin-1ß by CD11b- and Iba-1-positive cells, and loss of olfactory sensory neurons (OSNs). In the OB, we observed activation of microglia and astrocytes and decreased expression of tyrosine hydroxylase in periglomerular cells, vesicular glutamate transporter 1, a presynaptic protein, in mitral and tufted projection neurons, and 5T4 in granule cells. Thus, the OM inflammation exerted a detrimental effect, not only on OSNs, but also on OB neurons, which might lead to neurodegeneration.


Subject(s)
Gliosis/etiology , Gliosis/metabolism , Lipopolysaccharides/adverse effects , Olfactory Bulb/metabolism , Rhinitis/complications , Rhinitis/etiology , Synapses/metabolism , Animals , Cell Count , Fluorescent Antibody Technique , Gliosis/pathology , Male , Mice , Microscopy, Fluorescence , Nerve Degeneration , Olfactory Bulb/pathology , Olfactory Mucosa/metabolism , Olfactory Mucosa/pathology , Olfactory Receptor Neurons/metabolism , Olfactory Receptor Neurons/pathology , Synapses/pathology
19.
J Vis Exp ; (125)2017 07 13.
Article in English | MEDLINE | ID: mdl-28745638

ABSTRACT

Insects have evolved sophisticated olfactory reception systems to sense exogenous chemical signals. These chemical signals are transduced by Olfactory Receptor Neurons (ORNs) housed in hair-like structures, called chemosensilla, of the antennae. On the ORNs' membranes, Odorant Receptors (ORs) are believed to be involved in odor coding. Thus, being able to identify genes localized to the ORNs is necessary to recognize OR genes, and provides a fundamental basis for further functional in situ studies. The RNA expression levels of specific ORs in insect antennae are very low, and preserving insect tissue for histology is challenging. Thus, it is difficult to localize an OR to a specific type of sensilla using RNA in situ hybridization. In this paper, a detailed and highly effective RNA in situ hybridization protocol particularly for lowly expressed OR genes of insects, is introduced. In addition, a specific OR gene was identified by conducting double-color fluorescent in situ hybridization experiments using a co-expressing receptor gene, Orco, as a marker.


Subject(s)
Arthropod Antennae/pathology , Grasshoppers/metabolism , RNA/metabolism , Receptors, Odorant/genetics , Animals , Arthropod Antennae/metabolism , Immunohistochemistry , In Situ Hybridization, Fluorescence , Microscopy, Confocal , Olfactory Receptor Neurons/metabolism , Olfactory Receptor Neurons/pathology , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/metabolism , Receptors, Odorant/metabolism , Video Recording
20.
Cell Metab ; 26(1): 198-211.e5, 2017 Jul 05.
Article in English | MEDLINE | ID: mdl-28683287

ABSTRACT

Olfactory inputs help coordinate food appreciation and selection, but their role in systemic physiology and energy balance is poorly understood. Here we demonstrate that mice upon conditional ablation of mature olfactory sensory neurons (OSNs) are resistant to diet-induced obesity accompanied by increased thermogenesis in brown and inguinal fat depots. Acute loss of smell perception after obesity onset not only abrogated further weight gain but also improved fat mass and insulin resistance. Reduced olfactory input stimulates sympathetic nerve activity, resulting in activation of ß-adrenergic receptors on white and brown adipocytes to promote lipolysis. Conversely, conditional ablation of the IGF1 receptor in OSNs enhances olfactory performance in mice and leads to increased adiposity and insulin resistance. These findings unravel a new bidirectional function for the olfactory system in controlling energy homeostasis in response to sensory and hormonal signals.


Subject(s)
Obesity/metabolism , Obesity/physiopathology , Olfactory Receptor Neurons/metabolism , Smell , Thermogenesis , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/physiopathology , Animals , Diet, High-Fat/adverse effects , Energy Metabolism , Insulin Resistance , Insulin-Like Growth Factor I/metabolism , Lipolysis , Mice , Obesity/etiology , Olfactory Receptor Neurons/pathology , Receptors, Adrenergic, beta/metabolism , Weight Gain
SELECTION OF CITATIONS
SEARCH DETAIL
...