Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 243
Filtrer
1.
Biosensors (Basel) ; 14(6)2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38920600

RÉSUMÉ

Development and optimisation of bioelectronic monitoring techniques like microelectrode array-based field potential measurement and impedance spectroscopy for the functional, label-free and non-invasive monitoring of in vitro neuronal networks is widely investigated in the field of biosensors. Thus, these techniques were individually used to demonstrate the capabilities of, e.g., detecting compound-induced toxicity in neuronal culture models. In contrast, extended application for investigating the effects of central nervous system infecting viruses are rarely described. In this context, we wanted to analyse the effect of herpesviruses on functional neuronal networks. Therefore, we developed a unique hybrid bioelectronic monitoring platform that allows for performing field potential monitoring and impedance spectroscopy on the same microelectrode. In the first step, a neuronal culture model based on primary hippocampal cells from neonatal rats was established with reproducible and stable synchronised electrophysiological network activity after 21 days of cultivation on microelectrode arrays. For a proof of concept, the pseudorabies model virus PrV Kaplan-ΔgG-GFP was applied and the effect on the neuronal networks was monitored by impedance spectroscopy and field potential measurement for 72 h in a multiparametric mode. Analysis of several bioelectronic parameters revealed a virus concentration-dependent degeneration of the neuronal network within 24-48 h, with a significant early change in electrophysiological activity, subsequently leading to a loss of activity and network synchronicity. In conclusion, we successfully developed a microelectrode array-based hybrid bioelectronic measurement platform for quantitative monitoring of pathologic effects of a herpesvirus on electrophysiological active neuronal networks.


Sujet(s)
Techniques de biocapteur , Spectroscopie diélectrique , Neurones , Animaux , Rats , Neurones/virologie , Réseau nerveux , Microélectrodes , Hippocampe/virologie , Herpèsvirus porcin de type 1 , Cellules cultivées , Maladie d'Aujeszky/virologie
2.
Front Immunol ; 12: 753683, 2021.
Article de Anglais | MEDLINE | ID: mdl-34899705

RÉSUMÉ

Human parechovirus type 3 (PeV-A3) infection has been recognized as an emerging etiologic factor causing severe nerve disease or sepsis in infants and young children. But the neuropathogenic mechanisms of PeV-A3 remain unknown. To understand the pathogenesis of PeV-A3 infection in the neuronal system, PeV-A3-mediated cytopathic effects were analyzed in human glioblastoma cells and neuroblastoma cells. PeV-A3 induced interferons and inflammatory cytokine expression in these neuronal cells. The pronounced cytopathic effects accompanied with activation of death signaling pathways of apoptosis, autophagy, and pyroptosis were detected. A new experimental disease model of parechovirus encephalitis was established. In the disease model, intracranial inoculation with PeV-A3 in C57BL/6 neonatal mice showed body weight loss, hindlimb paralysis, and approximately 20% mortality. PeV-A3 infection in the hippocampus and cortex regions of the neonatal mouse brain was revealed. Mechanistic assay supported the in vitro results, indicating detection of PeV-A3 replication, inflammatory cytokine expression, and death signaling transduction in mouse brain tissues. These in vitro and in vivo studies revealed that the activation of death signaling and inflammation responses is involved in PeV-A3-mediated neurological disorders. The present results might account for some of the PeV-A3-associated clinical manifestations.


Sujet(s)
Effet cytopathogène viral , Modèles animaux de maladie humaine , Encéphalite virale/métabolisme , Parechovirus/pathogénicité , Infections à Picornaviridae/métabolisme , Animaux , Animaux nouveau-nés , Apoptose , Autophagie , Lignée cellulaire tumorale , Cortex cérébral/virologie , Chlorocebus aethiops , Cytokines/biosynthèse , Cytokines/génétique , Encéphalite virale/anatomopathologie , Encéphalite virale/virologie , Glioblastome/anatomopathologie , Hippocampe/virologie , Humains , Inflammation , Interféron de type I/biosynthèse , Interféron de type I/génétique , Interféron de type I/pharmacologie , Interférons/biosynthèse , Interférons/génétique , Potentiel de membrane mitochondriale , Souris , Souris de lignée C57BL , Neuroblastome/anatomopathologie , Parechovirus/effets des médicaments et des substances chimiques , Parechovirus/physiologie , Infections à Picornaviridae/anatomopathologie , Infections à Picornaviridae/virologie , Pyroptose , Cellules Vero , Réplication virale/effets des médicaments et des substances chimiques , Interféron lambda
3.
Sci Rep ; 11(1): 23819, 2021 12 10.
Article de Anglais | MEDLINE | ID: mdl-34893671

RÉSUMÉ

Neurotropic viruses target the brain and contribute to neurologic diseases. C-type lectin receptors (CLRs) are pattern recognition receptors that recognize carbohydrate structures on endogenous molecules and pathogens. The myeloid CLR dendritic cell immunoreceptor (DCIR) is expressed by antigen presenting cells and mediates inhibitory intracellular signalling. To investigate the effect of DCIR on neurotropic virus infection, mice were infected experimentally with Theiler's murine encephalomyelitis virus (TMEV). Brain tissue of TMEV-infected C57BL/6 mice and DCIR-/- mice were analysed by histology, immunohistochemistry and RT-qPCR, and spleen tissue by flow cytometry. To determine the impact of DCIR deficiency on T cell responses upon TMEV infection in vitro, antigen presentation assays were utilised. Genetic DCIR ablation in C57BL/6 mice was associated with an ameliorated hippocampal integrity together with reduced cerebral cytokine responses and reduced TMEV loads in the brain. Additionally, absence of DCIR favoured increased peripheral cytotoxic CD8+ T cell responses following TMEV infection. Co-culture experiments revealed that DCIR deficiency enhances the activation of antigen-specific CD8+ T cells by virus-exposed dendritic cells (DCs), indicated by increased release of interleukin-2 and interferon-γ. Results suggest that DCIR deficiency has a supportive influence on antiviral immune mechanisms, facilitating virus control in the brain and ameliorates neuropathology during acute neurotropic virus infection.


Sujet(s)
Infections à cardiovirus/virologie , Hippocampe/métabolisme , Hippocampe/virologie , Lectines de type C/métabolisme , Theilovirus/physiologie , Animaux , Cellules présentatrices d'antigène/immunologie , Cellules présentatrices d'antigène/métabolisme , Marqueurs biologiques , Biopsie , Cytokines/métabolisme , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Modèles animaux de maladie humaine , Prédisposition aux maladies , Hippocampe/anatomopathologie , Immunohistochimie , Immunomodulation , Lectines de type C/génétique , Souris , Souris knockout , Maladies neuro-inflammatoires/immunologie , Maladies neuro-inflammatoires/métabolisme , Maladies neuro-inflammatoires/anatomopathologie , Maladies neuro-inflammatoires/virologie , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , Sous-populations de lymphocytes T/anatomopathologie , Charge virale
4.
Int J Mol Sci ; 22(13)2021 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-34209576

RÉSUMÉ

Neurotropic viruses target the brain and contribute to neurologic diseases. Caspase recruitment domain containing family member 9 (CARD9) controls protective immunity in a variety of infectious disorders. To investigate the effect of CARD9 in neurotropic virus infection, CARD9-/- and corresponding C57BL/6 wild-type control mice were infected with Theiler's murine encephalomyelitis virus (TMEV). Brain tissue was analyzed by histology, immunohistochemistry and molecular analyses, and spleens by flow cytometry. To determine the impact of CARD9 deficiency on T cell responses in vitro, antigen presentation assays were utilized. Genetic ablation of CARD9 enhanced early pro-inflammatory cytokine responses and accelerated infiltration of T and B cells in the brain, together with a transient increase in TMEV-infected cells in the hippocampus. CARD9-/- mice showed an increased loss of neuronal nuclear protein+ mature neurons and doublecortin+ neuronal precursor cells and an increase in ß-amyloid precursor protein+ damaged axons in the hippocampus. No effect of CARD9 deficiency was found on the initiation of CD8+ T cell responses by flow cytometry and co-culture experiments using virus-exposed dendritic cells or microglia-enriched glial cell mixtures, respectively. The present study indicates that CARD9 is dispensable for the initiation of early antiviral responses and TMEV elimination but may contribute to the modulation of neuroinflammation, thereby reducing hippocampal injury following neurotropic virus infection.


Sujet(s)
Protéines adaptatrices de signalisation CARD/déficit , Prédisposition aux maladies , Encéphalite virale/étiologie , Hippocampe/virologie , Infections à Picornaviridae/étiologie , Picornaviridae/physiologie , Animaux , Marqueurs biologiques , Modèles animaux de maladie humaine , Encéphalite virale/anatomopathologie , Prédisposition génétique à une maladie , Hippocampe/métabolisme , Hippocampe/anatomopathologie , Interactions hôte-pathogène/génétique , Interactions hôte-pathogène/immunologie , Immunohistochimie , Sous-populations de lymphocytes/immunologie , Sous-populations de lymphocytes/métabolisme , Souris , Souris knockout , Infections à Picornaviridae/anatomopathologie , Charge virale
5.
Neurochem Res ; 46(9): 2427-2438, 2021 Sep.
Article de Anglais | MEDLINE | ID: mdl-34165668

RÉSUMÉ

Nucleoprotein (P40) is one of the most important proteins of Borna disease virus 1 (BoDV-1), but which proteins it would bind to in the pathogenesis of BoDV-1-infected hosts is unknown. We used lentivirus LV5-P40 overexpressing P40 to infect primary hippocampal neurons and characterized the interactome of P40 with co-immunoprecipitation (Co-IP) followed by mass spectrometry (MS) analysis. These interacting protein partners revealed the pathogenesis of BoDV-1-infected hosts. We also show for the first time that P40 interacts with 5HT2CR in rat neurons, which may be the molecular basis leading to neuropsychiatric diseases such as anxiety disorders and behavioral abnormalities after BoDV-1 infection of hosts.


Sujet(s)
Maladie de Borna/étiologie , Virus de la maladie de Borna/pathogénicité , Neurones/métabolisme , Récepteur de la sérotonine de type 5-HT2C/métabolisme , Protéines virales/métabolisme , Animaux , Virus de la maladie de Borna/composition chimique , Embryon de mammifère , Femelle , Hippocampe/cytologie , Hippocampe/virologie , Lentivirus/génétique , Neurones/virologie , Grossesse , Liaison aux protéines , Rat Sprague-Dawley , Protéines virales/génétique
6.
PLoS One ; 15(11): e0241667, 2020.
Article de Anglais | MEDLINE | ID: mdl-33137166

RÉSUMÉ

OBJECTIVE: HIV-associated CNS dysfunction is a significant problem among people with HIV (PWH), who now live longer due to viral suppression from combined anti-retroviral therapy (ART). Over the course of infection, HIV generates toxic viral proteins and induces inflammatory cytokines that have toxic effects on neurons in the CNS. Among these viral proteins, HIV Nef has been found in neurons of postmortem brain specimens from PWH. However, the source of Nef and its impact on neuronal cell homeostasis are still elusive. METHODS AND RESULTS: Here, in using a simian immunodeficiency virus (SIV) infected rhesus macaque model of neuroHIV, we find SIV Nef reactivity in the frontal cortex, hippocampus and cerebellum of SIV-infected animals using immunohistochemistry (IHC). Interestingly, SIV-infected macaques treated with ART also showed frequent Nef positive cells in the cerebellum and hippocampus. Using dual quantitative RNAscope and IHC, we observed cells that were positive for Nef, but were not for SIV RNA, suggesting that Nef protein is present in cells that are not actively infected with SIV. Using cell specific markers, we observed Nef protein in microglia/macrophages and astrocytes. Importantly, we also identified a number of NeuN-positive neurons, which are not permissive to SIV infection, but contained Nef protein. Further characterization of Nef-positive neurons showed caspase 3 activation, indicating late stage apoptosis in the CNS neurons. CONCLUSIONS: Our results suggest that regardless of ART status, Nef is expressed in the brain of SIV infected macaques and may contribute to neurological complications seen in PWH.


Sujet(s)
Cervelet/métabolisme , Produits du gène nef/génétique , Hippocampe/métabolisme , Syndrome d'immunodéficience acquise du singe/métabolisme , Animaux , Cervelet/virologie , Produits du gène nef/métabolisme , Hippocampe/virologie , Macaca mulatta , Mâle , Syndrome d'immunodéficience acquise du singe/virologie , Virus de l'immunodéficience simienne/génétique , Virus de l'immunodéficience simienne/métabolisme
7.
J Neuroinflammation ; 17(1): 278, 2020 Sep 20.
Article de Anglais | MEDLINE | ID: mdl-32951602

RÉSUMÉ

BACKGROUND: Tick-borne encephalitis virus (TBEV) is an important human pathogen that can cause the serious illness tick-borne encephalitis (TBE). Patients with clinical symptoms can suffer from severe meningoencephalitis with sequelae that include cognitive disorders and paralysis. While less than 30% of patients with clinical symptoms develop meningoencephalitis, the number of seropositive individuals in some regions indicates a much higher prevalence of TBEV infections, either with no or subclinical symptoms. The functional relevance of these subclinical TBEV infections and their influence on brain functions, such as learning and memory, has not been investigated so far. METHODS: To compare the effect of low and high viral replication in the brain, wildtype and Irf-7-/- mice were infected with Langat virus (LGTV), which belongs to the TBEV-serogroup. The viral burden was analyzed in the olfactory bulb and the hippocampus. Open field, elevated plus maze, and Morris water maze experiments were performed to determine the impact on anxiety-like behavior, learning, and memory formation. Spine density of hippocampal neurons and activation of microglia and astrocytes were analyzed. RESULTS: In contrast to susceptible Irf-7-/- mice, wildtype mice showed no disease signs upon LGTV infection. Detection of viral RNA in the olfactory bulb revealed CNS infections in wildtype and Irf-7-/- mice. Very low levels of viral replication were detectable in the hippocampus of wildtype mice. Although wildtype mice develop no disease signs, they showed reduced anxiety-like behavior and impaired memory formation, whereas Irf-7-/- mice were not affected. This impairment was associated with a significant decrease in spine density of neurons in the hippocampal CA1 region of wildtype mice. Microglia activation and astrogliosis were detected in the hippocampus. CONCLUSION: In this study, we demonstrate that subclinical infections by viruses from the TBEV-serogroup affected anxiety-like behavior. Virus replication in the olfactory bulb induced far-reaching effects on hippocampal neuron morphology and impaired hippocampus-dependent learning and memory formation.


Sujet(s)
Virus de l'encéphalite à tiques (sous-groupe)/isolement et purification , Encéphalites à tiques/anatomopathologie , Hippocampe/anatomopathologie , Hippocampe/virologie , Neurones/anatomopathologie , Neurones/virologie , Animaux , Femelle , Mâle , Apprentissage du labyrinthe/physiologie , Souris , Souris de lignée C57BL , Souris knockout
8.
Neurosci Lett ; 736: 135295, 2020 09 25.
Article de Anglais | MEDLINE | ID: mdl-32800922

RÉSUMÉ

Herpes simplex virus type 1 (HSV-1) is the main etiological agent of acute and sporadic encephalitis. Proteins of the suppressor of cytokine signaling (SOCS) family have shown to regulate the inflammation during HSV-1 infection in the brain. However, the effects of SOCS2 and SOCS3 in viral encephalitis remain unclear. The aim of the current study is to investigate the potential association between SOCS2, SOCS3, cytokines, and hippocampal damage, especially neuronal apoptosis, during acute intracranial HSV-1 infection in mice. Male C57BL/6 mice were infected by intracranial route with 102 plaque-forming units (PFU) inoculum of purified HSV-1. At three days post-infection (3 d.p.i.), mice were euthanized and their hippocampi were collected for histopathological analysis, immunohistochemical reaction against active caspase-3 and quantification of SOCS2, SOCS3 and cytokines (tumoral necrosis factor (TNF), interleukin (IL) 1ß, IL-6, IL-10; interferon (IFN) -α, IFN-ß, IFN-γ) mRNA expression. Infected mice exhibited neuronal loss and hemorrhagic focus in Cornu Ammonis (CA) region. The apoptotic index was higher in infected mice compared to controls. HSV-1 infection was associated with increased hippocampal expression of TNF, IL1-ß, IL-6 and IFNα/IFNß and decreased expression of IL-10, IFN-γ, SOCS2 and SOCS3. Our results suggest that down regulation of SOCS2 and SOCS3 contributes to a pro-inflammatory environment associated with hippocampal damage and neuronal apoptosis during acute HSV-1 infection in mice.


Sujet(s)
Encéphalite à herpès simplex/métabolisme , Hippocampe/virologie , Inflammation/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Protéines SOCS/métabolisme , Animaux , Apoptose/physiologie , Chlorocebus aethiops , Cytokines/métabolisme , Hippocampe/métabolisme , Mâle , Souris , Neurones/métabolisme , Neurones/virologie , Cellules Vero
9.
Neurotox Res ; 38(3): 626-639, 2020 Oct.
Article de Anglais | MEDLINE | ID: mdl-32683649

RÉSUMÉ

Major depression is a leading cause of morbidity and disease burden in modern society. Current drug treatment is only effective in a fraction of patients as underlying mechanisms of depression are not fully understood. ProBDNF, a precursor of brain-derived neurotrophic factor (BDNF), and its receptor p75NTR are highly upregulated in patients with major depression and in animal models of depression induced by chronic stress. Here, we hypothesise that proBDNF may be a pathogenic factor triggering depression. C57BL/6 mice were injected in the bilateral gluteus maximus muscle with AAV-proBDNF or AAV-EGFP. Four weeks after the injection, AAV-proBDNF injected animals developed depression-like behaviours, which were evident for 4-8 weeks and then returned to the control level after 12 weeks. In the second experiment, mice were divided into three groups; one group was treated with sheep anti-proBDNF antibody after AAV-proBDNF injection whereas the other two groups received PBS injection after the AAV-proBDNF or AAV-EGFP delivery. The group that was injected with AAV-proBDNF showed a time-dependent increase in immobility time in the tail suspension test and forced swim test, reduced sucrose consumption and decreased grooming time after sucrose spraying. Treatment with sheep anti-proBDNF antibody alleviated the depressive-like symptoms. Peripheral AAV-proBDNF delivery also resulted in a reduction of density and length of dendritic spines in the dentate gyrus and amygdala. Thus, we conclude that peripheral proBDNF is a primary pathogenic factor triggering depression-like behavioural changes in mice likely by reducing dendritic spine plasticity.


Sujet(s)
Dependovirus/métabolisme , Dépression/métabolisme , Trouble dépressif majeur/virologie , Stress psychologique/métabolisme , Animaux , Modèles animaux de maladie humaine , Hippocampe/métabolisme , Hippocampe/virologie , Souris de lignée C57BL , Muscles/virologie , Précurseurs de protéines/métabolisme
10.
PLoS Pathog ; 16(6): e1008381, 2020 06.
Article de Anglais | MEDLINE | ID: mdl-32525948

RÉSUMÉ

HIV invades the brain during acute infection. Yet, it is unknown whether long-lived infected brain cells release productive virus that can egress from the brain to re-seed peripheral organs. This understanding has significant implication for the brain as a reservoir for HIV and most importantly HIV interplay between the brain and peripheral organs. Given the sheer number of astrocytes in the human brain and their controversial role in HIV infection, we evaluated their infection in vivo and whether HIV infected astrocytes can support HIV egress to peripheral organs. We developed two novel models of chimeric human astrocyte/human peripheral blood mononuclear cells: NOD/scid-IL-2Rgc null (NSG) mice (huAstro/HuPBMCs) whereby we transplanted HIV (non-pseudotyped or VSVg-pseudotyped) infected or uninfected primary human fetal astrocytes (NHAs) or an astrocytoma cell line (U138MG) into the brain of neonate or adult NSG mice and reconstituted the animals with human peripheral blood mononuclear cells (PBMCs). We also transplanted uninfected astrocytes into the brain of NSG mice and reconstituted with infected PBMCs to mimic a biological infection course. As expected, the xenotransplanted astrocytes did not escape/migrate out of the brain and the blood brain barrier (BBB) was intact in this model. We demonstrate that astrocytes support HIV infection in vivo and egress to peripheral organs, at least in part, through trafficking of infected CD4+ T cells out of the brain. Astrocyte-derived HIV egress persists, albeit at low levels, under combination antiretroviral therapy (cART). Egressed HIV evolved with a pattern and rate typical of acute peripheral infection. Lastly, analysis of human cortical or hippocampal brain regions of donors under cART revealed that astrocytes harbor between 0.4-5.2% integrated HIV gag DNA and 2-7% are HIV gag mRNA positive. These studies establish a paradigm shift in the dynamic interaction between the brain and peripheral organs which can inform eradication of HIV reservoirs.


Sujet(s)
Astrocytes , Barrière hémato-encéphalique , Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , Hippocampe , Libération de particules virales , Animaux , Antirétroviraux/pharmacologie , Astrocytes/métabolisme , Astrocytes/anatomopathologie , Astrocytes/virologie , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/anatomopathologie , Barrière hémato-encéphalique/virologie , Lignée cellulaire tumorale , Infections à VIH/génétique , Infections à VIH/métabolisme , Infections à VIH/anatomopathologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Hippocampe/métabolisme , Hippocampe/anatomopathologie , Hippocampe/virologie , Humains , Souris , Souris de lignée NOD , Souris knockout , Souris SCID
11.
J Neurovirol ; 26(4): 590-601, 2020 08.
Article de Anglais | MEDLINE | ID: mdl-32572834

RÉSUMÉ

Although neuropsychological studies of human immunodeficiency virus (HIV)-infected patients have demonstrated heterogeneity in neurocognitive impairment and neuroimaging studies have reported diverse brain regions affected by HIV, it remains unclear whether individual differences in neurocognitive impairment are underpinned by their neural bases. Here, we investigated spatial distribution patterns of correlation between neurocognitive function and regional gray matter (GM) volume across patients with HIV. Thirty-one combination antiretroviral therapy-treated HIV-infected Japanese male patients and 33 age- and sex-matched healthy controls were included in the analysis after strict exclusion criteria, especially for substance use. Fifteen neurocognitive tests were used, and volumetric magnetic resonance imaging was performed. We used voxel-based morphometry to compare GM volume between groups and identify regional GM volumes that correlated with neurocognitive tests across patients. Using the Frascati criteria, 10 patients were diagnosed with asymptomatic neurocognitive impairment, while the others were not diagnosed with HIV-associated neurocognitive disorders. Patients showed a significantly lower performance in five neurocognitive tests as well as significantly reduced GM volume relative to controls, with volume-reduced regions spread diffusely across the whole brain. Different aspects of neurocognitive impairment (i.e., figural copy, finger tapping, and Pegboard) were associated with different GM regions. Our findings suggest a biological background constituting heterogeneity of neurocognitive impairment in HIV infection and support the clinical importance of considering individual differences for tailor-made medicine for people living with HIV.


Sujet(s)
Agents antiVIH/usage thérapeutique , Dysfonctionnement cognitif/physiopathologie , Substance grise/physiopathologie , Infections à VIH/physiopathologie , Adulte , Thérapie antirétrovirale hautement active , Maladies asymptomatiques , Attention/effets des médicaments et des substances chimiques , Études cas-témoins , Dysfonctionnement cognitif/imagerie diagnostique , Dysfonctionnement cognitif/traitement médicamenteux , Dysfonctionnement cognitif/virologie , Fonction exécutive/effets des médicaments et des substances chimiques , Substance grise/imagerie diagnostique , Substance grise/effets des médicaments et des substances chimiques , Substance grise/virologie , Gyrus du cingulum/imagerie diagnostique , Gyrus du cingulum/effets des médicaments et des substances chimiques , Gyrus du cingulum/physiopathologie , Gyrus du cingulum/virologie , Infections à VIH/imagerie diagnostique , Infections à VIH/traitement médicamenteux , Infections à VIH/virologie , Hippocampe/imagerie diagnostique , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/physiopathologie , Hippocampe/virologie , Humains , Imagerie par résonance magnétique , Mâle , Mémoire/effets des médicaments et des substances chimiques , Tests de l'état mental et de la démence , Adulte d'âge moyen , Aptitudes motrices/effets des médicaments et des substances chimiques , Neuroimagerie/méthodes , Lobe occipital/imagerie diagnostique , Lobe occipital/effets des médicaments et des substances chimiques , Lobe occipital/physiopathologie , Lobe occipital/virologie , Lobe pariétal/imagerie diagnostique , Lobe pariétal/effets des médicaments et des substances chimiques , Lobe pariétal/physiopathologie , Lobe pariétal/virologie , Cortex préfrontal/imagerie diagnostique , Cortex préfrontal/effets des médicaments et des substances chimiques , Cortex préfrontal/physiopathologie , Cortex préfrontal/virologie , Indice de gravité de la maladie , Parole/effets des médicaments et des substances chimiques
12.
PLoS One ; 15(3): e0230436, 2020.
Article de Anglais | MEDLINE | ID: mdl-32210460

RÉSUMÉ

OBJECTIVES: Our aim was to compare neuropsychological and psychiatric outcomes across three encephalitis aetiological groups: Herpes simplex virus (HSV), other infections or autoimmune causes (Other), and encephalitis of unknown cause (Unknown). METHODS: Patients recruited from NHS hospitals underwent neuropsychological and psychiatric assessment in the short-term (4 months post-discharge), medium-term (9-12 months after the first assessment), and long-term (>1-year). Healthy control subjects were recruited from the general population and completed the same assessments. RESULTS: Patients with HSV were most severely impaired on anterograde and retrograde memory tasks. In the short-term, they also showed executive, IQ, and naming deficits, which resolved in the long-term. Patients with Other or Unknown causes of encephalitis showed moderate memory impairments, but no significant impairment on executive tests. Memory impairment was associated with hippocampal/medial temporal damage on magnetic resonance imaging (MRI), and naming impairment with left temporal and left frontal abnormalities. Patients reported more subjective cognitive complaints than healthy controls, with tiredness a significant problem, and there were high rates of depression and anxiety in the HSV and the Other encephalitis groups. These subjective, self-reported complaints, depression, and anxiety persisted even after objectively measured neuropsychological performance had improved. CONCLUSIONS: Neuropsychological and psychiatric outcomes after encephalitis vary according to aetiology. Memory and naming are severely affected in HSV, and less so in other forms. Neuropsychological functioning improves over time, particularly in those with more severe short-term impairments, but subjective cognitive complaints, depression, and anxiety persist, and should be addressed in rehabilitation programmes.


Sujet(s)
Troubles de la cognition/physiopathologie , Dysfonctionnement cognitif/physiopathologie , Encéphalite/physiopathologie , Troubles de la mémoire/physiopathologie , Troubles de la cognition/imagerie diagnostique , Troubles de la cognition/virologie , Dysfonctionnement cognitif/imagerie diagnostique , Dysfonctionnement cognitif/virologie , Dépression/physiopathologie , Encéphalite/imagerie diagnostique , Encéphalite/psychologie , Encéphalite/virologie , Femelle , Hippocampe/imagerie diagnostique , Hippocampe/physiopathologie , Hippocampe/virologie , Humains , Mâle , Mémoire/physiologie , Troubles de la mémoire/imagerie diagnostique , Troubles de la mémoire/virologie , Adulte d'âge moyen , Tests neuropsychologiques , Simplexvirus/pathogénicité , Vision/physiologie
13.
Biomed Res Int ; 2020: 2702175, 2020.
Article de Anglais | MEDLINE | ID: mdl-32185196

RÉSUMÉ

Chemokine CC motif ligand 2 (CCL2) is one of the most recognized proinflammatory chemokines, and the expression of CCL2 in the cerebrospinal fluid of patients infected with HIV-1 is significantly higher than that of healthy people. As such, it is seen as an important cause of HIV-associated neurocognitive disorder (HAND). Our previous investigation has confirmed the pathological role of CCL2 in mediating brain damage leading to cognitive dysfunction. Currently, however, research on therapeutic drugs for the central nervous system targeting CCL2 is very limited. Our present study used brain stereotactic technology to induce cognitive impairment in rats by injecting CCL2 (5 ng) into the bilateral hippocampus. To investigate the protective effect and mechanism of Tanshinone IIA (25, 50, 75 mg/kg/d) on CCL2-induced learning memory and cognitive impairment in rats, we performed the Morris water maze (MWM) and novel object recognition tests (NORT) on the rats. The results showed that Tanshinone IIA significantly alleviated CCL2-induced learning memory and cognitive dysfunction. Further studies on the hippocampal tissue of the rats revealed that Tanshinone IIA treatment significantly increased the activity of SOD and GSH-Px while the level of MDA decreased compared to the model group. Additionally, the relative expression of apoptosis-associated genes caspase-3, caspase-8, and caspase-9 and inflammation-associated genes IL-1ß and IL-6 in Tanshinone IIA-treated rats was lower than that in model rats. Finally, we confirmed hippocampal neuron loss and apoptosis by Nissl staining and TdT-mediated dUTP Nick end labeling (TUNEL). Taken together, these data imply that Tanshinone IIA can ameliorate CCL2-induced learning memory and cognitive impairment by impacting oxidative stress, inflammation, and apoptosis. Tanshinone IIA may be a potential therapeutic agent for the treatment of HAND.


Sujet(s)
Abiétanes/pharmacologie , Chimiokine CCL2/métabolisme , Dysfonctionnement cognitif/traitement médicamenteux , Dysfonctionnement cognitif/étiologie , Infections à VIH/complications , Troubles de la mémoire/traitement médicamenteux , Troubles de la mémoire/étiologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Protéines régulatrices de l'apoptose/métabolisme , Cognition/effets des médicaments et des substances chimiques , Dysfonctionnement cognitif/métabolisme , Dysfonctionnement cognitif/virologie , Modèles animaux de maladie humaine , Infections à VIH/métabolisme , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Hippocampe/virologie , Méthode TUNEL/méthodes , Inflammation/traitement médicamenteux , Inflammation/étiologie , Inflammation/métabolisme , Inflammation/virologie , Mâle , Mémoire/effets des médicaments et des substances chimiques , Troubles de la mémoire/métabolisme , Troubles de la mémoire/virologie , Rats , Rat Sprague-Dawley
14.
Proc Natl Acad Sci U S A ; 116(51): 25982-25990, 2019 12 17.
Article de Anglais | MEDLINE | ID: mdl-31792184

RÉSUMÉ

Retrotransposons compose a staggering 40% of the mammalian genome. Among them, endogenous retroviruses (ERV) represent sequences that closely resemble the proviruses created from exogenous retroviral infection. ERVs make up 8 to 10% of human and mouse genomes and range from evolutionarily ancient sequences to recent acquisitions. Studies in Drosophila have provided a causal link between genomic retroviral elements and cognitive decline; however, in mammals, the role of ERVs in learning and memory remains unclear. Here we studied 2 independent murine models for ERV activation: muMT strain (lacking B cells and antibody production) and intracerebroventricular injection of streptozotocin (ICVI-STZ). We conducted behavioral assessments (contextual fear memory and spatial learning), as well as gene and protein analysis (RNA sequencing, PCR, immunohistochemistry, and western blot assays). Mice lacking mitochondrial antiviral-signaling protein (MAVS) and mice lacking stimulator of IFN genes protein (STING), 2 downstream sensors of ERV activation, provided confirmation of ERV impact. We found that muMT mice and ICVI-STZ mice induced hippocampal ERV activation, as shown by increased gene and protein expression of the Gag sequence of the transposable element intracisternal A-particle. ERV activation was accompanied by significant hippocampus-related memory impairment in both models. Notably, the deficiency of the MAVS pathway was protective against ICVI-STZ-induced cognitive pathology. Overall, our results demonstrate that ERV activation is associated with cognitive impairment in mice. Moreover, they provide a molecular target for strategies aimed at attenuating retroviral element sensing, via MAVS, to treat dementia and neuropsychiatric disorders.


Sujet(s)
Rétrovirus endogènes/génétique , Hippocampe/virologie , Troubles de la mémoire/génétique , Troubles de la mémoire/métabolisme , Troubles de la mémoire/virologie , Protéines adaptatrices de la transduction du signal/génétique , Animaux , Comportement animal , Encéphale/anatomopathologie , Dysfonctionnement cognitif , Éléments transposables d'ADN , Modèles animaux de maladie humaine , Rétrovirus endogènes/physiologie , Régulation de l'expression des gènes , Produits du gène gag , Hippocampe/effets des médicaments et des substances chimiques , Apprentissage , Mâle , Protéines membranaires/métabolisme , Mémoire , Troubles de la mémoire/psychologie , Souris , Souris de lignée C57BL , Souris knockout , Streptozocine/pharmacologie
15.
Stem Cells ; 37(11): 1467-1480, 2019 11.
Article de Anglais | MEDLINE | ID: mdl-31381841

RÉSUMÉ

We previously reported that Herpes simplex virus type-1 (HSV-1) infection of cultured neurons triggered intracellular accumulation of amyloid-ß protein (Aß) markedly impinging on neuronal functions. Here, we demonstrated that HSV-1 affects in vitro and in vivo adult hippocampal neurogenesis by reducing neural stem/progenitor cell (NSC) proliferation and their neuronal differentiation via intracellular Aß accumulation. Specifically, cultured NSCs were more permissive for HSV-1 replication than mature neurons and, once infected, they exhibited reduced proliferation (assessed by 5'-bromo-deoxyuridine incorporation, Ki67 immunoreactivity, and Sox2 mRNA expression) and impaired neuronal differentiation in favor of glial phenotype (evaluated by immunoreactivity for the neuronal marker MAP2, the glial marker glial fibrillary astrocyte protein, and the expression of the proneuronal genes Mash1 and NeuroD1). Similarly, impaired adult neurogenesis was observed in the subgranular zone of hippocampal dentate gyrus of an in vivo model of recurrent HSV-1 infections, that we recently set up and characterized, with respect to mock-infected mice. The effects of HSV-1 on neurogenesis did not depend on cell death and were due to Aß accumulation in infected NSCs. Indeed, they were: (a) reverted, in vitro, by the presence of either ß/γ-secretase inhibitors preventing Aß production or the specific 4G8 antibody counteracting the action of intracellular Aß; (b) not detectable, in vivo, in HSV-1-infected amyloid precursor protein knockout mice, unable to produce and accumulate Aß. Given the critical role played by adult neurogenesis in hippocampal-dependent memory and learning, our results suggest that multiple virus reactivations in the brain may contribute to Alzheimer's disease phenotype by also targeting NSCs. Stem Cells 2019;37:1467-1480.


Sujet(s)
Peptides bêta-amyloïdes/métabolisme , Herpèsvirus humain de type 1/pathogénicité , Hippocampe/métabolisme , Hippocampe/virologie , Peptides bêta-amyloïdes/génétique , Précurseur de la protéine bêta-amyloïde/génétique , Précurseur de la protéine bêta-amyloïde/métabolisme , Animaux , Technique de Western , Cellules cultivées , Immunohistochimie , Mâle , Souris , Souris de lignée C57BL , Cellules souches neurales/cytologie , Cellules souches neurales/métabolisme , Neurogenèse/génétique , Neurogenèse/physiologie , RT-PCR
16.
mBio ; 10(4)2019 07 02.
Article de Anglais | MEDLINE | ID: mdl-31266862

RÉSUMÉ

HIV causes neurodegeneration and dementia in AIDS patients, but its function in milder cognitive impairments in virologically suppressed patients on antiretroviral therapy is unknown. Such patients are immunocompetent, have low peripheral and brain HIV burdens, and show minimal brain neuropathology. Using the model of HIV-related memory impairment in EcoHIV-infected conventional mice, we investigated the neurobiological and cognitive consequences of efficient EcoHIV expression in the mouse brain after intracerebral infection. HIV integrated and persisted in an expressed state in brain tissue, was detectable in brain monocytic cells, and caused neuroinflammatory responses and lasting spatial, working, and associative memory impairment. Systemic antiretroviral treatment prevented direct brain infection and memory dysfunction indicating the requirement for HIV expression in the brain for disease. Similarly inoculated murine leukemia virus used as a control replicated in mouse brain but not in monocytic cells and was cognitively benign, linking the disease to HIV-specific functions. Memory impairment correlated in real time with hippocampal dysfunction shown by defective long-term potentiation in hippocampal slices ex vivo and with diffuse synaptodendritic injury in the hippocampus reflected in significant reduction in microtubule-associated protein 2 and synapsin II staining. In contrast, there was no evidence of overt neuronal loss in this region as determined by neuron-specific nuclear protein quantification, TUNEL assay, and histological observations. Our results reveal a novel capacity of HIV to induce neuronal dysfunction and memory impairment independent of neurotoxicity, distinct from the neurotoxicity of HIV infection in dementia.IMPORTANCE HIV neuropathogenesis has been attributed in large measure to neurotoxicity of viral proteins and inflammatory factors produced by infected monocytic cells in the brain. We show here that HIV expression in mouse brain causes lasting memory impairment by a mechanism involving injury to hippocampal synaptodendritic arbors and neuronal function but not overt neuronal loss in the region. Our results mirror the observation of minimal neurodegeneration in cognitively impaired HIV patients on antiretroviral therapy and demonstrate that HIV is nonneurotoxic in certain brain abnormalities that it causes. If neurons comprising the cognition-related networks survive HIV insult, at least for some time, there is a window of opportunity for disease treatment.


Sujet(s)
Démence associée au SIDA/physiopathologie , Dysfonctionnement cognitif/physiopathologie , Hippocampe/anatomopathologie , Hippocampe/virologie , Troubles de la mémoire/complications , Troubles de la mémoire/physiopathologie , Animaux , Comportement animal , Modèles animaux de maladie humaine , Mâle , Souris , Charge virale
17.
Elife ; 82019 06 17.
Article de Anglais | MEDLINE | ID: mdl-31205000

RÉSUMÉ

Multiple lines of evidence suggest that functionally intact cerebello-hippocampal interactions are required for appropriate spatial processing. However, how the cerebellum anatomically and physiologically engages with the hippocampus to sustain such communication remains unknown. Using rabies virus as a retrograde transneuronal tracer in mice, we reveal that the dorsal hippocampus receives input from topographically restricted and disparate regions of the cerebellum. By simultaneously recording local field potential from both the dorsal hippocampus and anatomically connected cerebellar regions, we additionally suggest that the two structures interact, in a behaviorally dynamic manner, through subregion-specific synchronization of neuronal oscillations in the 6-12 Hz frequency range. Together, these results reveal a novel neural network macro-architecture through which we can understand how a brain region classically associated with motor control, the cerebellum, may influence hippocampal neuronal activity and related functions, such as spatial navigation.


Sujet(s)
Cervelet/physiologie , Hippocampe/physiologie , Réseau nerveux/physiologie , Voies nerveuses/physiologie , Animaux , Cervelet/anatomie et histologie , Cervelet/virologie , Stimulation électrique , Hippocampe/anatomie et histologie , Hippocampe/virologie , Mâle , Souris de lignée C57BL , Réseau nerveux/anatomie et histologie , Réseau nerveux/virologie , Voies nerveuses/anatomie et histologie , Voies nerveuses/virologie , Neurones/physiologie , Neurones/virologie , Rage (maladie)/physiopathologie , Rage (maladie)/virologie , Virus de la rage/physiologie , Navigation spatiale/physiologie
18.
J Neurovirol ; 25(4): 560-577, 2019 08.
Article de Anglais | MEDLINE | ID: mdl-31102185

RÉSUMÉ

Poor antiretroviral penetration may contribute to human immunodeficiency virus (HIV) persistence within the brain and to neurocognitive deficits in opiate abusers. To investigate this problem, HIV-1 Tat protein and morphine effects on blood-brain barrier (BBB) permeability and drug brain penetration were explored using a conditional HIV-1 Tat transgenic mouse model. Tat and morphine effects on the leakage of fluorescently labeled dextrans (10-, 40-, and 70-kDa) into the brain were assessed. To evaluate effects on antiretroviral brain penetration, Tat+ and Tat- mice received three antiretroviral drugs (dolutegravir, abacavir, and lamivudine) with or without concurrent morphine exposure. Antiretroviral and morphine brain and plasma concentrations were determined by LC-MS/MS. Morphine exposure, and, to a lesser extent, Tat, significantly increased tracer leakage from the vasculature into the brain. Despite enhanced BBB breakdown evidenced by increased tracer leakiness, morphine exposure led to significantly lower abacavir concentrations within the striatum and significantly less dolutegravir within the hippocampus and striatum (normalized to plasma). P-glycoprotein, an efflux transporter for which these drugs are substrates, expression and function were significantly increased in the brains of morphine-exposed mice compared to mice not exposed to morphine. These findings were consistent with lower antiretroviral concentrations in brain tissues examined. Lamivudine concentrations were unaffected by Tat or morphine exposure. Collectively, our investigations indicate that Tat and morphine differentially alter BBB integrity. Morphine decreased brain concentrations of specific antiretroviral drugs, perhaps via increased expression of the drug efflux transporter, P-glycoprotein.


Sujet(s)
Agents antiVIH/pharmacocinétique , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Morphine/effets indésirables , Produits du gène tat du virus de l'immunodéficience humaine/biosynthèse , Glycoprotéine P/génétique , Glycoprotéine P/métabolisme , Animaux , Transport biologique/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/virologie , Perméabilité capillaire , Corps strié/effets des médicaments et des substances chimiques , Corps strié/métabolisme , Corps strié/virologie , Dextrane/pharmacocinétique , Didéoxynucléosides/pharmacocinétique , Femelle , Fluorescéine-5-isothiocyanate/analogues et dérivés , Fluorescéine-5-isothiocyanate/pharmacocinétique , Infections à VIH/métabolisme , Infections à VIH/psychologie , Infections à VIH/virologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , Composés hétérocycliques 3 noyaux/pharmacocinétique , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Hippocampe/virologie , Lamivudine/pharmacocinétique , Souris , Souris transgéniques , Modèles biologiques , Troubles neurocognitifs/métabolisme , Troubles neurocognitifs/psychologie , Troubles neurocognitifs/virologie , Oxazines , Pipérazines , Pyridones , Produits du gène tat du virus de l'immunodéficience humaine/génétique
19.
J Neurovirol ; 25(4): 480-495, 2019 08.
Article de Anglais | MEDLINE | ID: mdl-31028692

RÉSUMÉ

The age of the HIV-infected population is increasing. Although many studies document gray matter volume (GMV) changes following HIV infection, GMV also declines with age. Findings have been inconsistent concerning interactions between HIV infection and age on brain structure. Effects of age, substance use, and inadequate viral suppression may confound identification of GMV serostatus effects using quantitative structural measures. In a cross-sectional study of HIV infection, including 97 seropositive and 84 seronegative, demographically matched participants, ages 30-70, we examined serostatus and age effects on GMV and neuropsychological measures. Ninety-eight percent of seropositive participants were currently treated with anti-retroviral therapies and all were virally suppressed. Gray, white, and CSF volumes were estimated using high-resolution T1-weighted MRI. Linear regression modeled effects of serostatus, age, education, comorbidities, and magnetic field strength on brain structure, using both a priori regions and voxel-based morphometry. Although seropositive participants exhibited significant bilateral decreases in striatal GMV, no serostatus effects were detected in the thalamus, hippocampus, or cerebellum. Age was associated with cortical, striatal, thalamic, hippocampal, and cerebellar GMV reductions. Effects of age and serostatus on striatal GMV were additive. Although no main effects of serostatus on neuropsychological performance were observed, serostatus moderated the relationship between pegboard performance and striatal volume. Both HIV infection and age were associated with reduced striatal volume. The lack of interaction of these two predictors suggests that HIV infection is associated with premature, but not accelerated, brain age. In serostatus groups matched on demographic and clinical variables, there were no observed differences in neuropsychological performance. Striatal GMV measures may be promising biomarker for use in studies of treated HIV infection.


Sujet(s)
Vieillissement/anatomopathologie , Corps strié/anatomopathologie , Substance grise/anatomopathologie , Infections à VIH/anatomopathologie , Hippocampe/anatomopathologie , Lobe temporal/anatomopathologie , Thalamus/anatomopathologie , Adulte , Facteurs âges , Sujet âgé , Vieillissement/effets des médicaments et des substances chimiques , Agents antiVIH/usage thérapeutique , Thérapie antirétrovirale hautement active , Cartographie cérébrale , Études cas-témoins , Corps strié/imagerie diagnostique , Corps strié/effets des médicaments et des substances chimiques , Corps strié/virologie , Femelle , Substance grise/imagerie diagnostique , Substance grise/effets des médicaments et des substances chimiques , Substance grise/virologie , Infections à VIH/imagerie diagnostique , Infections à VIH/traitement médicamenteux , Infections à VIH/virologie , Hippocampe/imagerie diagnostique , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/virologie , Humains , Imagerie par résonance magnétique , Mâle , Adulte d'âge moyen , Neuroimagerie , Tests neuropsychologiques , Lobe temporal/imagerie diagnostique , Lobe temporal/effets des médicaments et des substances chimiques , Lobe temporal/virologie , Thalamus/imagerie diagnostique , Thalamus/effets des médicaments et des substances chimiques , Thalamus/virologie , Substance blanche/imagerie diagnostique , Substance blanche/effets des médicaments et des substances chimiques , Substance blanche/anatomopathologie , Substance blanche/virologie
20.
J Virol ; 93(13)2019 07 01.
Article de Anglais | MEDLINE | ID: mdl-31019048

RÉSUMÉ

Fatal neurological syndromes can occur after measles virus (MeV) infection of the brain. The mechanisms controlling MeV spread within the central nervous system (CNS) remain poorly understood. We analyzed the role of type I interferon (IFN-I) receptor (IFNAR) signaling in the control of MeV infection in a murine model of brain infection. Using organotypic brain cultures (OBC) from wild-type and IFNAR-knockout (IFNARKO) transgenic mice ubiquitously expressing the human SLAM (CD150) receptor, the heterogeneity of the permissiveness of different CNS cell types to MeV infection was characterized. In the absence of IFNAR signaling, MeV propagated significantly better in explant slices. In OBC from IFNAR-competent mice, while astrocytes and microglia were infected on the day of explant preparation, they became refractory to infection with time, in contrast to neurons and oligodendrocytes, which remained permissive to infection. This selective loss of permissiveness to MeV infection was not observed in IFNARKO mouse OBC. Accordingly, the development of astrogliosis related to the OBC procedure was exacerbated in the presence of IFNAR signaling. In the hippocampus, this astrogliosis was characterized by a change in the astrocyte phenotype and by an increase of IFN-I transcripts. A proteome analysis showed the upregulation of 84 out of 111 secreted proteins. In the absence of IFNAR, only 27 secreted proteins were upregulated, and none of these were associated with antiviral activities. Our results highlight the essential role of the IFN-I response in astrogliosis and in the permissiveness of astrocytes and microglia that could control MeV propagation throughout the CNS.IMPORTANCE Measles virus (MeV) can infect the central nervous system (CNS), with dramatic consequences. The mechanisms controlling MeV invasion of the CNS remain ill-defined since most previous data were obtained from postmortem analysis. Here, we highlight for the first time the crucial role of the type I interferon (IFN-I) response not only in the control of CNS invasion but also in the early permissiveness of glial cells to measles virus infection.


Sujet(s)
Astrocytes/virologie , Virus de la rougeole/métabolisme , Rougeole/métabolisme , Microglie/virologie , Récepteur à l'interféron alpha-bêta/métabolisme , Transduction du signal/physiologie , Animaux , Antiviraux/pharmacologie , Astrocytes/anatomopathologie , Encéphale/virologie , Système nerveux central/virologie , Cytokines , Femelle , Hippocampe/anatomopathologie , Hippocampe/virologie , Humains , Mâle , Rougeole/anatomopathologie , Rougeole/virologie , Souris , Souris knockout , Neurones/virologie , Oligodendroglie/virologie , Récepteur à l'interféron alpha-bêta/génétique , Transduction du signal/génétique , Membre-1 de la famille des molécules de signalisation de l'activation des lymphocytes/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...