Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
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.
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
3.
ACS Chem Neurosci ; 11(15): 2152-2155, 2020 08 05.
Article in English | MEDLINE | ID: mdl-32673476

ABSTRACT

It has become clear since the pandemic broke out that SARS-CoV-2 virus causes reduction of smell and taste in a significant fraction of COVID-19 patients. The olfactory dysfunction often occurs early in the course of the disease, and sometimes it is the only symptom in otherwise asymptomatic carriers. The cellular mechanisms for these specific olfactory disturbances in COVID-19 are now beginning to be elucidated. Several very recent papers contributed to explaining the key cellular steps occurring in the olfactory epithelium leading to anosmia/hyposmia (collectively known as dysosmia) initiated by SARS-CoV-2 infection. In this Viewpoint, we discuss current progress in research on olfactory dysfunction in COVID-19 and we also propose an updated model of the SARS-CoV-2-induced dysosmia. The emerging central role of sustentacular cells and inflammatory processes in the olfactory epithelium are particularly considered. The proposed model of anosmia in COVID-19 does not answer unequivocally whether the new coronavirus exploits the olfactory route to rapidly or slowly reach the brain in COVID-19 patients. To answer this question, new systematic studies using an infectious virus and appropriate animal models are needed.


Subject(s)
Betacoronavirus , Coronavirus Infections/complications , Olfaction Disorders/etiology , Olfaction Disorders/virology , Olfactory Receptor Neurons/cytology , Olfactory Receptor Neurons/virology , Pneumonia, Viral/complications , Animals , COVID-19 , Coronavirus Infections/metabolism , Humans , Olfaction Disorders/metabolism , Olfactory Receptor Neurons/metabolism , Pandemics , Pneumonia, Viral/metabolism , SARS-CoV-2 , Smell/physiology
4.
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
5.
ACS Chem Neurosci ; 11(9): 1200-1203, 2020 05 06.
Article in English | MEDLINE | ID: mdl-32283006

ABSTRACT

The novel SARS-CoV-2 virus has very high infectivity, which allows it to spread rapidly around the world. Attempts at slowing the pandemic at this stage depend on the number and quality of diagnostic tests performed. We propose that the olfactory epithelium from the nasal cavity may be a more appropriate tissue for detection of SARS-CoV-2 virus at the earliest stages, prior to onset of symptoms or even in asymptomatic people, as compared to commonly used sputum or nasopharyngeal swabs. Here we emphasize that the nasal cavity olfactory epithelium is the likely site of enhanced binding of SARS-CoV-2. Multiple non-neuronal cell types present in the olfactory epithelium express two host receptors, ACE2 and TMPRSS2 proteases, that facilitate SARS-CoV-2 binding, replication, and accumulation. This may be the underlying mechanism for the recently reported cases of smell dysfunction in patients with COVID-19. Moreover, the possibility of subsequent brain infection should be considered which begins in olfactory neurons. In addition, we discuss the possibility that olfactory receptor neurons may initiate rapid immune responses at early stages of the disease. We emphasize the need to undertake research focused on additional aspects of SARS-CoV-2 actions in the nervous system, especially in the olfactory pathway.


Subject(s)
Betacoronavirus/isolation & purification , Brain/virology , Coronavirus Infections/diagnosis , Early Diagnosis , Mass Screening/methods , Olfactory Mucosa/virology , Pneumonia, Viral/diagnosis , Smell , Angiotensin-Converting Enzyme 2 , Animals , Betacoronavirus/growth & development , Betacoronavirus/immunology , Brain/immunology , Brain/physiopathology , COVID-19 , Coronavirus Infections/immunology , Coronavirus Infections/physiopathology , Coronavirus Infections/transmission , Humans , Immunity, Innate , Mass Screening/standards , Mice , Olfactory Mucosa/cytology , Olfactory Mucosa/immunology , Olfactory Mucosa/metabolism , Olfactory Receptor Neurons/immunology , Olfactory Receptor Neurons/metabolism , Olfactory Receptor Neurons/virology , Pandemics , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/immunology , Pneumonia, Viral/physiopathology , Pneumonia, Viral/transmission , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology , SARS-CoV-2 , Serine Endopeptidases/metabolism , Virus Replication
6.
Sci Rep ; 10(1): 5259, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32210257

ABSTRACT

Sindbis virus (SINV) is an alphavirus that causes age-dependent encephalomyelitis in mice. Within 7-8 days after infection infectious virus is cleared from neurons through the antiviral effects of antibody and interferon-gamma (IFNγ), but RNA persists. To better understand changes in viral RNA associated with immune-mediated clearance we developed recombinant strains of SINV that have genomic and subgenomic viral RNAs tagged with the Broccoli RNA aptamer that binds and activates a conditional fluorophore for live cell imaging of RNA. Treatment of SINV-Broccoli-infected cells with antibody to the SINV E2 glycoprotein had cell type-specific effects. In BHK cells, antibody increased levels of intracellular viral RNA and changed the primary location of genomic RNA from the perinuclear region to the plasma membrane without improving cell viability. In undifferentiated and differentiated AP7 (dAP7) neuronal cells, antibody treatment decreased levels of viral RNA. Occasional dAP7 cells escaped antibody-mediated clearance by not expressing cell surface E2 or binding antibody to the plasma membrane. IFNγ decreased viral RNA levels only in dAP7 cells and synergized with antibody for RNA clearance and improved cell survival. Therefore, analysis of aptamer-tagged SINV RNAs identified cell type- and neuronal maturation-dependent responses to immune mediators of virus clearance.


Subject(s)
Antibodies, Viral/pharmacology , Aptamers, Nucleotide/analysis , Fibroblasts/virology , Glycoproteins/immunology , Interferon-gamma/pharmacology , Neurons/virology , RNA, Viral/analysis , Sindbis Virus/genetics , Single-Cell Analysis/methods , Time-Lapse Imaging/methods , Viral Nonstructural Proteins/analysis , Viral Proteins/immunology , Animals , Antibodies, Viral/immunology , Antibody Specificity , Cell Differentiation , Cell Line , Cell Line, Transformed , Cricetinae , Fibroblasts/ultrastructure , Luminescent Proteins , Mesocricetus , Neurons/ultrastructure , Olfactory Receptor Neurons/ultrastructure , Olfactory Receptor Neurons/virology , Rats , Recombinant Proteins/analysis , Sindbis Virus/immunology , Subcellular Fractions/chemistry , Subcellular Fractions/ultrastructure , Red Fluorescent Protein
7.
J Neurochem ; 155(2): 137-153, 2020 09.
Article in English | MEDLINE | ID: mdl-31811775

ABSTRACT

The olfactory mucosa, where the first step of odor detection occurs, is a privileged pathway for environmental toxicants and pathogens toward the central nervous system. Indeed, some pathogens can infect olfactory sensory neurons including their axons projecting to the olfactory bulb allowing them to bypass the blood-brain barrier and reach the central nervous system (CNS) through the so-called olfactory pathway. The respiratory syncytial virus (RSV) is a major respiratory tract pathogen but there is growing evidence that RSV may lead to CNS impairments. However, the mechanisms involved in RSV entering into the CNS have been poorly described. In this study, we wanted to explore the capacity of RSV to reach the CNS via the olfactory pathway and to better characterize RSV cellular tropism in the nasal cavity. We first explored the distribution of RSV infectious sites in the nasal cavity by in vivo bioluminescence imaging and a tissue clearing protocol combined with deep-tissue imaging and 3D image analyses. This whole tissue characterization was confirmed with immunohistochemistry and molecular biology approaches. Together, our results provide a novel 3D atlas of mouse nasal cavity anatomy and show that RSV can infect olfactory sensory neurons giving access to the central nervous system by entering the olfactory bulb. Cover Image for this issue: doi: 10.1111/jnc.14765.


Subject(s)
Olfactory Mucosa/innervation , Olfactory Mucosa/virology , Olfactory Receptor Neurons/virology , Respiratory Syncytial Viruses , Animals , Central Nervous System/diagnostic imaging , Central Nervous System/virology , Central Nervous System Diseases/diagnostic imaging , Central Nervous System Diseases/virology , Female , Head/anatomy & histology , Imaging, Three-Dimensional , Mice , Mice, Inbred BALB C , Nasal Mucosa/virology , Olfactory Bulb/virology , Olfactory Mucosa/diagnostic imaging , RNA, Viral/isolation & purification , Tropism , Virus Replication
8.
Proc Natl Acad Sci U S A ; 116(25): 12428-12436, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31160464

ABSTRACT

The nervous system regulates host immunity in complex ways. Vertebrate olfactory sensory neurons (OSNs) are located in direct contact with pathogens; however, OSNs' ability to detect danger and initiate immune responses is unclear. We report that nasal delivery of rhabdoviruses induces apoptosis in crypt OSNs via the interaction of the OSN TrkA receptor with the viral glycoprotein in teleost fish. This signal results in electrical activation of neurons and very rapid proinflammatory responses in the olfactory organ (OO), but dampened inflammation in the olfactory bulb (OB). CD8α+ cells infiltrate the OO within minutes of nasal viral delivery, and TrkA blocking, but not caspase-3 blocking, abrogates this response. Infiltrating CD8α+ cells were TCRαß T cells with a nonconventional phenotype that originated from the microvasculature surrounding the OB and not the periphery. Nasal delivery of viral glycoprotein (G protein) recapitulated the immune responses observed with the whole virus, and antibody blocking of viral G protein abrogated these responses. Ablation of crypt neurons in zebrafish resulted in increased susceptibility to rhabdoviruses. These results indicate a function for OSNs as a first layer of pathogen detection in vertebrates and as orchestrators of nasal-CNS antiviral immune responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Infectious hematopoietic necrosis virus/immunology , Olfactory Receptor Neurons/physiology , Receptor, trkA/metabolism , Animals , Apoptosis , Caspase 3/metabolism , Nasal Mucosa/immunology , Nasal Mucosa/virology , Olfactory Receptor Neurons/cytology , Olfactory Receptor Neurons/virology , Oncorhynchus mykiss
9.
J Neural Transm (Vienna) ; 124(6): 721-738, 2017 06.
Article in English | MEDLINE | ID: mdl-28477284

ABSTRACT

Braak and Del Tredici have proposed that typical Parkinson disease (PD) has its origins in the olfactory bulb and gastrointestinal tract. However, the role of the olfactory system has insufficiently been explored in the pathogeneses of PD and Alzheimer disease (AD) in laboratory models. Here, we demonstrate applications of a new method to process mouse heads for microscopy by sectioning, mounting, and staining whole skulls ('holocranohistochemistry'). This technique permits the visualization of the olfactory system from the nasal cavity to mitral cells and dopamine-producing interneurons of glomeruli in the olfactory bulb. We applied this method to two specific goals: first, to visualize PD- and AD-linked gene expression in the olfactory system, where we detected abundant, endogenous α-synuclein and tau expression in the olfactory epithelium. Furthermore, we observed amyloid-ß plaques and proteinase-K-resistant α-synuclein species, respectively, in cranial nerve-I of APP- and human SNCA-over-expressing mice. The second application of the technique was to the modeling of gene-environment interactions in the nasal cavity of mice. We tracked the infection of a neurotropic respiratory-enteric-orphan virus from the nose pad into cranial nerves-I (and -V) and monitored the ensuing brain infection. Given its abundance in the olfactory epithelia, we questioned whether α-synuclein played a role in innate host defenses to modify the outcome of infections. Indeed, Snca-null mice were more likely to succumb to viral encephalitis versus their wild-type littermates. Moreover, using a bacterial sepsis model, Snca-null mice were less able to control infection after intravenous inoculation with Salmonella typhimurium. Together, holocranohistochemistry enabled new discoveries related to α-synuclein expression and its function in mice. Future studies will address: the role of Mapt and mutant SNCA alleles in infection paradigms; the contribution of xenobiotics in the initiation of idiopathic PD; and the safety to the host when systemically targeting α-synuclein by immunotherapy.


Subject(s)
Brain/metabolism , Brain/virology , Encephalitis, Viral/virology , Olfactory Mucosa/anatomy & histology , Olfactory Mucosa/metabolism , Reoviridae Infections/virology , alpha-Synuclein/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/diagnostic imaging , Brain/pathology , Disease Models, Animal , Encephalitis, Viral/immunology , Encephalitis, Viral/mortality , Encephalitis, Viral/pathology , Female , Head , Humans , Immunohistochemistry , Male , Mammalian orthoreovirus 3 , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Neural Pathways/anatomy & histology , Neural Pathways/diagnostic imaging , Neural Pathways/metabolism , Neural Pathways/pathology , Olfactory Mucosa/pathology , Olfactory Receptor Neurons/metabolism , Olfactory Receptor Neurons/virology , Reoviridae Infections/immunology , Salmonella Infections/immunology , Salmonella Infections/pathology , Salmonella typhimurium , Tissue Preservation/methods , alpha-Synuclein/genetics
10.
PLoS One ; 11(7): e0159033, 2016.
Article in English | MEDLINE | ID: mdl-27428110

ABSTRACT

BACKGROUND: Viral infection is a common cause of olfactory dysfunction. The complexities of studying post-viral olfactory loss in humans have impaired further progress in understanding the underlying mechanism. Recently, evidence from clinical studies has implicated Parainfluenza virus 3 as a causal agent. An animal model of post viral olfactory disorders (PVOD) would allow better understanding of disease pathogenesis and represent a major advance in the field. OBJECTIVE: To develop a mouse model of PVOD by evaluating the effects of Sendai virus (SeV), the murine counterpart of Parainfluenza virus, on olfactory function and regenerative ability of the olfactory epithelium. METHODS: C57BL/6 mice (6-8 months old) were inoculated intranasally with SeV or ultraviolet (UV)-inactivated virus (UV-SeV). On days 3, 10, 15, 30 and 60 post-infection, olfactory epithelium was harvested and analyzed by histopathology and immunohistochemical detection of S-phase nuclei. We also measured apoptosis by TUNEL assay and viral load by real-time PCR. The buried food test (BFT) was used to measure olfactory function of mice at day 60. In parallel, cultured murine olfactory sensory neurons (OSNs) infected with SeV or UV-SeV were tested for odorant-mixture response by measuring changes in intracellular calcium concentrations indicated by fura-4 AM assay. RESULTS: Mice infected with SeV suffered from olfactory dysfunction, peaking on day 15, with no loss observed with UV-SeV. At 60 days, four out of 12 mice infected with SeV still had not recovered, with continued normal function in controls. Viral copies of SeV persisted in both the olfactory epithelium (OE) and the olfactory bulb (OB) for at least 60 days. At day 10 and after, both unit length labeling index (ULLI) of apoptosis and ULLI of proliferation in the SeV group was markedly less than the UV-SeV group. In primary cultured OSNs infected by SeV, the percentage of cells responding to mixed odors was markedly lower in the SeV group compared to UV-SeV (P = 0.007). CONCLUSION: We demonstrate that SeV impairs olfaction, persists in OE and OB tissue, reduces their regenerative ability, and impairs the normal physiological function of OSNs without gross cytopathology. This mouse model shares key features of human post-viral olfactory loss, supporting its future use in studies of PVOD. Further testing and development of this model should allow us to clarify the pathophysiology of PVOD.


Subject(s)
Olfaction Disorders/pathology , Olfaction Disorders/virology , Olfactory Mucosa/pathology , Olfactory Receptor Neurons/pathology , Respirovirus Infections/complications , Sendai virus/physiology , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Odorants/analysis , Olfaction Disorders/physiopathology , Olfactory Mucosa/physiopathology , Olfactory Mucosa/virology , Olfactory Receptor Neurons/virology , Respirovirus Infections/pathology , Respirovirus Infections/physiopathology , Smell
11.
J Virol ; 90(12): 5785-96, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27053560

ABSTRACT

UNLABELLED: Venezuelan and western equine encephalitis viruses (VEEV and WEEV; Alphavirus; Togaviridae) are mosquito-borne pathogens causing central nervous system (CNS) disease in humans and equids. Adult CD-1 mice also develop CNS disease after infection with VEEV and WEEV. Adult CD-1 mice infected by the intranasal (i.n.) route, showed that VEEV and WEEV enter the brain through olfactory sensory neurons (OSNs). In this study, we injected the mouse footpad with recombinant WEEV (McMillan) or VEEV (subtype IC strain 3908) expressing firefly luciferase (fLUC) to simulate mosquito infection and examined alphavirus entry in the CNS. Luciferase expression served as a marker of infection detected as bioluminescence (BLM) by in vivo and ex vivo imaging. BLM imaging detected WEEV and VEEV at 12 h postinoculation (hpi) at the injection site (footpad) and as early as 72 hpi in the brain. BLM from WEEV.McM-fLUC and VEEV.3908-fLUC injections was initially detected in the brain's circumventricular organs (CVOs). No BLM activity was detected in the olfactory neuroepithelium or OSNs. Mice were also injected in the footpad with WEEV.McM expressing DsRed (Discosoma sp.) and imaged by confocal fluorescence microscopy. DsRed imaging supported our BLM findings by detecting WEEV in the CVOs prior to spreading along the neuronal axis to other brain regions. Taken together, these findings support our hypothesis that peripherally injected alphaviruses enter the CNS by hematogenous seeding of the CVOs followed by centripetal spread along the neuronal axis. IMPORTANCE: VEEV and WEEV are mosquito-borne viruses causing sporadic epidemics in the Americas. Both viruses are associated with CNS disease in horses, humans, and mouse infection models. In this study, we injected VEEV or WEEV, engineered to express bioluminescent or fluorescent reporters (fLUC and DsRed, respectively), into the footpads of outbred CD-1 mice to simulate transmission by a mosquito. Reporter expression serves as detectable bioluminescent and fluorescent markers of VEEV and WEEV replication and infection. Bioluminescence imaging, histological examination, and confocal fluorescence microscopy were used to identify early entry sites of these alphaviruses in the CNS. We observed that specific areas of the brain (circumventricular organs [CVOs]) consistently showed the earliest signs of infection with VEEV and WEEV. Histological examination supported VEEV and WEEV entering the brain of mice at specific sites where the blood-brain barrier is naturally absent.


Subject(s)
Blood-Brain Barrier/virology , Brain/virology , Encephalitis Virus, Venezuelan Equine/physiology , Encephalitis Virus, Western Equine/physiology , Encephalomyelitis, Venezuelan Equine/virology , Virus Internalization , Adult , Animals , Blood-Brain Barrier/physiopathology , Brain/pathology , Disease Models, Animal , Encephalitis Virus, Venezuelan Equine/genetics , Encephalitis Virus, Venezuelan Equine/growth & development , Encephalitis Virus, Western Equine/genetics , Encephalitis Virus, Western Equine/growth & development , Humans , Luciferases , Luminescent Measurements , Mice , Olfactory Receptor Neurons/virology , Optical Imaging/methods , Viral Load
12.
ACS Chem Neurosci ; 7(4): 464-9, 2016 Apr 20.
Article in English | MEDLINE | ID: mdl-27058872

ABSTRACT

In 1935, the olfactory route was hypothesized to be a portal for virus entry into the central nervous system (CNS). This hypothesis was based on experiments in which nasophayngeal infection with poliovirus in monkeys was prevented from spreading to their CNS via transection of olfactory tracts between the olfactory neuroepithelium (ONE) of the nasal cavity and the olfactory bulb (OB). Since then, numerous neurotropic viruses have been observed to enter the CNS via retrograde transport along axons of olfactory sensory neurons whose cell bodies reside in the ONE. Importantly, this route of infection can occur even after subcutaneous inoculation of arboviruses that can cause encephalitis in humans. While the olfactory route is now accepted as an important pathway for viral entry into the CNS, it is unclear whether it provides a way for infection to spread to other brain regions. More recently, studies of antiviral innate and adaptive immune responses within the olfactory bulb suggest it provides early virologic control. Here we will review the data demonstrating that neurotropic viruses gain access to the CNS initially via the olfactory route with emphasis on findings that suggest the OB is a critical immunosensory effector organ that effectively clears virus.


Subject(s)
Central Nervous System Viral Diseases/pathology , Inflammation/prevention & control , Olfactory Bulb/pathology , Olfactory Bulb/virology , Animals , Anti-Inflammatory Agents/therapeutic use , Central Nervous System Viral Diseases/complications , Humans , Inflammation/etiology , Neuroectodermal Tumors, Primitive, Peripheral , Olfactory Receptor Neurons/pathology , Olfactory Receptor Neurons/virology , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
13.
Int Immunopharmacol ; 22(1): 242-7, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24998164

ABSTRACT

Upper airway viral infection in patients with airway allergy often exacerbates olfactory dysfunction, but the mechanism for this exacerbation remains unclear. Here, we examined the effects of respiratory syncytial virus (RSV) infection, in the presence or absence of airway allergy, on olfactory receptor neurons (ORNs) and their progenitors in mice. Immunohistological analyses revealed that cockroach allergen (CRA)-induced airway allergy alone did not affect the number of OMP(+) mature ORNs and SOX2(+) ORN progenitors. Intranasal RSV line 19 infection in allergy-free mice resulted in a transient decrease in SOX2(+) ORN progenitors without affecting OMP(+) ORNs. In contrast, the RSV-induced decrease in SOX2(+) ORN progenitors was exacerbated and prolonged in allergic mice, which resulted in eventual loss of OMP(+) ORNs. In the allergic mice, reduction of RSV in the olfactory epithelium was delayed as compared with allergy-free mice. These results suggest that ORN progenitors were impaired by RSV infection and that airway allergy exacerbated damage to ORN progenitors by reducing viral clearance.


Subject(s)
Hypersensitivity/immunology , Nasal Mucosa/immunology , Olfactory Receptor Neurons/physiology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Allergens/immunology , Animals , Cell Differentiation/immunology , Cockroaches , Female , Hypersensitivity/complications , Mice , Mice, Inbred BALB C , Nasal Mucosa/virology , Olfactory Receptor Neurons/virology , Respiratory Syncytial Virus Infections/complications , SOXB1 Transcription Factors/metabolism , Viral Load
14.
J Virol ; 85(20): 10682-90, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21849461

ABSTRACT

Venezuelan equine encephalitis (VEE) virus is a mosquito-borne alphavirus associated with sporadic outbreaks in human and equid populations in the Western Hemisphere. After the bite of an infected mosquito, the virus initiates a biphasic disease: a peripheral phase with viral replication in lymphoid and myeloid tissues, followed by a neurotropic phase with infection of central nervous system (CNS) neurons, causing neuropathology and in some cases fatal encephalitis. The mechanisms allowing VEE virus to enter the CNS are currently poorly understood. Previous data have shown that the virus gains access to the CNS by infecting olfactory sensory neurons in the nasal mucosa of mice. However, at day 5 after inoculation, the infection of the brain is multifocal, indicating that virus particles are able to cross the blood-brain barrier (BBB). To better understand the role of the BBB during VEE virus infection, we used a well-characterized mouse model system. Using VEE virus replicon particles (VRP), we modeled the early events of neuroinvasion, showing that the replication of VRP in the nasal mucosa induced the opening of the BBB, allowing peripherally administered VRP to invade the brain. Peripheral VEE virus infection was characterized by a biphasic opening of the BBB. Further, inhibition of BBB opening resulted in a delayed viral neuroinvasion and pathogenesis. Overall, these results suggest that VEE virus initially enters the CNS through the olfactory pathways and initiates viral replication in the brain, which induces the opening of the BBB, allowing a second wave of invading virus from the periphery to enter the brain.


Subject(s)
Blood-Brain Barrier/physiopathology , Blood-Brain Barrier/virology , Encephalitis Virus, Venezuelan Equine/pathogenicity , Encephalomyelitis, Venezuelan Equine/pathology , Encephalomyelitis, Venezuelan Equine/virology , Animals , Disease Models, Animal , Mice , Mice, Inbred BALB C , Olfactory Receptor Neurons/virology , Rodent Diseases/pathology , Rodent Diseases/virology
15.
J Vis Exp ; (51)2011 May 22.
Article in English | MEDLINE | ID: mdl-21633336

ABSTRACT

Development of a precise olfactory circuit relies on accurate projection of olfactory sensory neuron (OSN) axons to their synaptic targets in the olfactory bulb (OB). The molecular mechanisms of OSN axon growth and targeting are not well understood. Manipulating gene expression and subsequent visualizing of single OSN axons and their terminal arbor morphology have thus far been challenging. To study gene function at the single cell level within a specified time frame, we developed a lentiviral based technique to manipulate gene expression in OSNs in vivo. Lentiviral particles are delivered to OSNs by microinjection into the olfactory epithelium (OE). Expression cassettes are then permanently integrated into the genome of transduced OSNs. Green fluorescent protein expression identifies infected OSNs and outlines their entire morphology, including the axon terminal arbor. Due to the short turnaround time between microinjection and reporter detection, gene function studies can be focused within a very narrow period of development. With this method, we have detected GFP expression within as few as three days and as long as three months following injection. We have achieved both over-expression and shRNA mediated knock-down by lentiviral microinjection. This method provides detailed morphologies of OSN cell bodies and axons at the single cell level in vivo, and thus allows characterization of candidate gene function during olfactory development.


Subject(s)
Lentivirus/genetics , Olfactory Receptor Neurons/chemistry , Olfactory Receptor Neurons/physiology , Animals , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Mice , Mice, Inbred C57BL , Olfactory Receptor Neurons/virology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics
16.
Microbes Infect ; 8(7): 1806-12, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16815072

ABSTRACT

A role for the US3 protein kinase of herpes simplex virus (HSV) in regulating virus-induced neuronal apoptosis was investigated in an experimental mouse system, in which wild-type HSV invades the central nervous system (CNS) via the olfactory and vomeronasal systems upon intranasal infection. Wild-type HSV-2 strain 186 infected a fraction of olfactory and vomeronasal chemosensory neurons without inducing apoptosis and was transmitted to the CNS, precipitating lethal encephalitis. In sharp contrast, an US3-disrupted mutant, L1BR1, induced neuronal apoptosis in these peripheral conduits upon infection, blocking viral transmission to the CNS and causing no signs of disease. An US3-repaired mutant, L1B(-)11, behaved similarly to the wild-type virus. Only 5 p.f.u. of L1BR1 was sufficient to compromise mice when the mutant virus was introduced directly into the olfactory bulb, a viral entry site of the CNS. These results suggest that the US3 protein kinase of HSV regulates virus-induced neuronal apoptosis in peripheral conduits and determines the neuroinvasive phenotype of HSV. Furthermore, virus-induced neuronal apoptosis of peripheral nervous system cells may be a protective host response that blocks viral transmission to the CNS.


Subject(s)
Apoptosis , Neurons, Afferent/cytology , Neurons, Afferent/virology , Olfactory Receptor Neurons/virology , Protein Serine-Threonine Kinases/physiology , Simplexvirus/enzymology , Viral Proteins/physiology , Vomeronasal Organ/virology , Animals , Body Weight , Brain/virology , Disease Models, Animal , Encephalitis/virology , Female , Gene Deletion , Herpes Simplex/immunology , Herpes Simplex/pathology , Immunohistochemistry , Mice , Mice, Inbred BALB C , Olfactory Nerve/virology , Olfactory Receptor Neurons/cytology , Protein Serine-Threonine Kinases/genetics , Simplexvirus/genetics , Simplexvirus/immunology , Simplexvirus/pathogenicity , Survival Analysis , Viral Proteins/genetics , Vomeronasal Organ/cytology
17.
J Neurovirol ; 11(2): 129-37, 2005 Apr.
Article in English | MEDLINE | ID: mdl-16036791

ABSTRACT

Olfactory receptor neurons are unique in their anatomical structure and function. Each neuron is directly exposed to the external environment at the site of its dendritic nerve terminals where it is exposed to macromolecules. These molecules can be incorporated into by olfactory receptor neurons and transported transsynaptically to the central nervous system. Certain neurotropic pathogens such as herpes simplex virus and Borna disease virus make use of this physiological mechanism to invade the brain. Here the authors review the olfactory transmission of infectious agents and the resulting hazards to human and animal health.


Subject(s)
Central Nervous System Viral Diseases/virology , Olfactory Receptor Neurons/virology , Virus Diseases/virology , Animals , Borna disease virus/pathogenicity , Borna disease virus/physiology , Humans , Influenza A virus/pathogenicity , Influenza A virus/physiology , Lyssavirus/pathogenicity , Lyssavirus/physiology , Nervous System/virology , Olfactory Mucosa/virology , Paramyxovirinae/pathogenicity , Prion Diseases/etiology , Prions/pathogenicity , Prions/physiology , Simplexvirus/pathogenicity , Simplexvirus/physiology , Synapses/virology
18.
J Gen Virol ; 83(Pt 9): 2109-2116, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12185263

ABSTRACT

Olfactory receptor neurons (ORNs) were infected upon intranasal inoculation with the R404BP strain of neurovirulent influenza A virus. Virus-infected neurons and a small fraction of neighbouring uninfected neurons displayed apoptotic neurodegeneration substantiated by the immunohistochemistry for activated caspase-3 molecules and the terminal deoxynucleotidyl transferase-mediated dUTP nick end-labelling method. However, virus infection was restricted within the peripheral neuroepithelium and all mice survived the infection. Virus-infected ORNs revealed upregulated expression of the Fas ligand molecules, activating the c-Jun N-terminal kinase signal transduction pathway. In addition, Iba1-expressing activated microglia/macrophages appeared to partake in phagocytic activities, eventually clearing apoptotic bodies. These results raise the possibility that induction of apoptosis in olfactory receptor neurons at an early stage of infection may provide protective effects against invasion of the neurovirulent virus from the peripheral to the CNS.


Subject(s)
Apoptosis , Brain/virology , Central Nervous System Viral Diseases/physiopathology , Influenza A virus , Nerve Degeneration , Olfactory Receptor Neurons/physiopathology , Animals , Calcium-Binding Proteins/analysis , Calcium-Binding Proteins/metabolism , Central Nervous System Viral Diseases/virology , Fas Ligand Protein , Female , Influenza A virus/genetics , Membrane Glycoproteins/analysis , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Microfilament Proteins , Microglia/metabolism , Olfactory Mucosa/metabolism , Olfactory Receptor Neurons/virology , Proto-Oncogene Proteins c-jun/analysis , Proto-Oncogene Proteins c-jun/metabolism , Recombination, Genetic , Signal Transduction
19.
Science ; 279(5348): 237-42, 1998 Jan 09.
Article in English | MEDLINE | ID: mdl-9422698

ABSTRACT

Candidate mammalian odorant receptors were first cloned some 6 years ago. The physiological function of these receptors in initiating transduction in olfactory receptor neurons remains to be established. Here, a recombinant adenovirus was used to drive expression of a particular receptor gene in an increased number of sensory neurons in the rat olfactory epithelium. Electrophysiological recording showed that increased expression of a single gene led to greater sensitivity to a small subset of odorants.


Subject(s)
Aldehydes/pharmacology , Odorants , Olfactory Receptor Neurons/physiology , Receptors, Odorant/physiology , Adenoviridae/genetics , Adenoviridae/physiology , Aldehydes/metabolism , Animals , Electrophysiology , Female , Gene Expression , Genetic Vectors , Green Fluorescent Proteins , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Male , Olfactory Receptor Neurons/virology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Receptors, Odorant/genetics , Receptors, Odorant/metabolism , Recombinant Proteins
20.
Ann N Y Acad Sci ; 855: 751-61, 1998 Nov 30.
Article in English | MEDLINE | ID: mdl-9929681

ABSTRACT

This communication describes our ongoing studies of the interaction of the mouse host and vesicular stomatitis virus (VSV). When VSV is applied to the nasal neuroepithelium, it initially replicates in olfactory receptor neurons, and is transmitted along the olfactory nerve to the central nervous system (CNS) within 12 hours. In the olfactory bulb, the virus replicates invasively through the layers of the olfactory bulb, reaching the olfactory ventricle by day 4-5 post infection, and the hindbrain by day 8 post infection. In mice, infection may result in a 50% mortality rate. The crucial host innate and specific immune responses responsible for restricting viral propagation and caudal spread of the virus will be discussed. The efficacy of interleukin-12 (IL-12) treatment for enhanced viral clearance and promotion of host recovery are described along with the implications for treatment of human encephalitis. The hosts' response to infection is also regulated by the sex of the host, and the age at infection. The role of specific mucosal humoral immunity and systemic cellular immunity in prevention of infection are described.


Subject(s)
Olfactory Bulb/virology , Olfactory Receptor Neurons/virology , Vesicular stomatitis Indiana virus/physiology , Virus Replication , Animals , Brain/virology , Humans , Immunity, Mucosal , Mice , Olfactory Bulb/immunology , Olfactory Receptor Neurons/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...