Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Brain Behav Immun ; 105: 67-81, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35803480

RESUMO

The epidemiological association between bacterial or viral maternal infections during pregnancy and increased risk for developing psychiatric disorders in offspring is well documented. Numerous rodent and non-human primate studies of viral- or, to a lesser extent, bacterial-induced maternal immune activation (MIA) have documented a series of neurological alterations that may contribute to understanding the pathophysiology of schizophrenia and autism spectrum disorders. Long-term neuronal and behavioral alterations are now ascribed to the effect of maternal proinflammatory cytokines rather than the infection itself. However, detailed electrophysiological alterations in brain areas relevant to psychiatric disorders, such as the dorsal hippocampus, are lacking in response to bacterial-induced MIA. This study determined if electrophysiological and morphological alterations converge in CA1 pyramidal cells (CA1 PC) from the dorsal hippocampus in bacterial-induced MIA offspring. A series of changes in the functional expression of K+ and Na+ ion channels altered the passive and active membrane properties and triggered hyperexcitability of CA1 PC. Contributing to the hyperexcitability, the somatic A-type potassium current (IA) was decreased in MIA CA1 PC. Likewise, the spontaneous glutamatergic and GABAergic inputs were dysregulated and biased toward increased excitation, thereby reshaping the excitation-inhibition balance. Consistent with these findings, the dendritic branching complexity of MIA CA1 PC was reduced. Together, these morphophysiological alterations modify CA1 PC computational capabilities and contribute to explaining cellular alterations that may underlie the cognitive symptoms of MIA-associated psychiatric disorders.


Assuntos
Imunidade , Neurônios , Canais de Potássio , Animais , Transtorno do Espectro Autista/imunologia , Região CA1 Hipocampal/citologia , Regulação para Baixo , Feminino , Neurônios/metabolismo , Canais de Potássio/metabolismo , Gravidez , Células Piramidais/imunologia , Esquizofrenia/imunologia
2.
Front Immunol ; 12: 749433, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34759929

RESUMO

The investigation of the molecular background of direct communication of neurons and immune cells in the brain is an important issue for understanding physiological and pathological processes in the nervous system. Direct contacts between brain-infiltrating immune cells and neurons, and the neuromodulatory effect of immune cell-derived regulatory peptides are well established. Several aspects of the role of immune and glial cells in the direct neuro-immune communication are also well known; however, there remain many questions regarding the molecular details of signaling from neurons to immune cells. Thus, we report here on the neuronal expression of genes encoding antimicrobial and immunomodulatory peptides, as well as proteins of immune cell-specific activation and communication mechanisms. In the present study, we analyzed the single-cell sequencing data of our previous transcriptomic work, obtained from electrophysiologically identified pyramidal cells and interneurons of the murine prefrontal cortex. We filtered out the genes that may be associated with the direct communication between immune cells and neurons and examined their expression pattern in the neuronal transcriptome. The expression of some of these genes by cortical neurons has not yet been reported. The vast majority of antimicrobial (~53%) and immune cell protein (~94%) transcripts was identified in the transcriptome of the 84 cells, owing to the high sensitivity of ultra-deep sequencing. Several of the antimicrobial and immune process-related protein transcripts showed cell type-specific or enriched expression. Individual neurons transcribed only a fraction of the investigated genes with low copy numbers probably due to the bursting kinetics of gene expression; however, the comparison of our data with available transcriptomic datasets from immune cells and neurons suggests the functional relevance of the reported findings. Accordingly, we propose further experimental and in silico studies on the neuronal expression of immune system-related genes and the potential role of the encoded proteins in neuroimmunological processes.


Assuntos
Córtex Pré-Frontal/imunologia , Células Piramidais/imunologia , Animais , Apresentação de Antígeno/genética , Peptídeos Antimicrobianos/genética , Linfócitos B/imunologia , Masculino , Camundongos Endogâmicos C57BL , Análise de Célula Única , Linfócitos T/imunologia , Transcriptoma
3.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-33619082

RESUMO

Encephalitis associated with antibodies against the neuronal gamma-aminobutyric acid A receptor (GABAA-R) is a rare form of autoimmune encephalitis. The pathogenesis is still unknown but autoimmune mechanisms were surmised. Here we identified a strongly expanded B cell clone in the cerebrospinal fluid of a patient with GABAA-R encephalitis. We expressed the antibody produced by it and showed by enzyme-linked immunosorbent assay (ELISA) and immunohistochemistry that it recognizes the GABAA-R. Patch-clamp recordings revealed that it tones down inhibitory synaptic transmission and causes increased excitability of hippocampal CA1 pyramidal neurons. Thus, the antibody likely contributed to clinical disease symptoms. Hybridization to a protein array revealed the cross-reactive protein LIM-domain-only protein 5 (LMO5), which is related to cell-cycle regulation and tumor growth. We confirmed LMO5 recognition by immunoprecipitation and ELISA and showed that cerebrospinal fluid samples from two other patients with GABAA-R encephalitis also recognized LMO5. This suggests that cross-reactivity between GABAA-R and LMO5 is frequent in GABAA-R encephalitis and supports the hypothesis of a paraneoplastic etiology.


Assuntos
Antígenos de Neoplasias/imunologia , Autoanticorpos/imunologia , Reações Cruzadas/imunologia , Suscetibilidade a Doenças , Encefalite/etiologia , Receptores de GABA-A/imunologia , Autoantígenos/imunologia , Doenças Autoimunes do Sistema Nervoso/etiologia , Doenças Autoimunes do Sistema Nervoso/metabolismo , Autoimunidade , Linfócitos B/imunologia , Linfócitos B/metabolismo , Biomarcadores , Suscetibilidade a Doenças/imunologia , Encefalite/metabolismo , Encefalite/patologia , Humanos , Células Piramidais/imunologia , Células Piramidais/metabolismo
4.
J Neuroinflammation ; 17(1): 69, 2020 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-32087716

RESUMO

BACKGROUND: Interferon-γ (IFN-γ, a type II IFN) is present in the central nervous system (CNS) under various conditions. Evidence is emerging that, in addition to its immunological role, IFN-γ modulates neuronal morphology, function, and development in several brain regions. Previously, we have shown that raising levels of IFN-ß (a type I IFN) lead to increased neuronal excitability of neocortical layer 5 pyramidal neurons. Because of shared non-canonical signaling pathways of both cytokines, we hypothesized a similar neocortical role of acutely applied IFN-γ. METHODS: We used semi-quantitative RT-PCR, immunoblotting, and immunohistochemistry to analyze neuronal expression of IFN-γ receptors and performed whole-cell patch-clamp recordings in layer 5 pyramidal neurons to investigate sub- and suprathreshold excitability, properties of hyperpolarization-activated cyclic nucleotide-gated current (Ih), and inhibitory neurotransmission under the influence of acutely applied IFN-γ. RESULTS: We show that IFN-γ receptors are present in the membrane of rat's neocortical layer 5 pyramidal neurons. As expected from this and the putative overlap in IFN type I and II alternative signaling pathways, IFN-γ diminished Ih, mirroring the effect of type I IFNs, suggesting a likewise activation of protein kinase C (PKC). In contrast, IFN-γ did neither alter subthreshold nor suprathreshold neuronal excitability, pointing to augmented inhibitory transmission by IFN-γ. Indeed, IFN-γ increased electrically evoked inhibitory postsynaptic currents (IPSCs) on neocortical layer 5 pyramidal neurons. Furthermore, amplitudes of spontaneous IPSCs and miniature IPSCs were elevated by IFN-γ, whereas their frequency remained unchanged. CONCLUSIONS: The expression of IFN-γ receptors on layer 5 neocortical pyramidal neurons together with the acute augmentation of inhibition in the neocortex by direct application of IFN-γ highlights an additional interaction between the CNS and immune system. Our results strengthen our understanding of the role of IFN-γ in neocortical neurotransmission and emphasize its impact beyond its immunological properties, particularly in the pathogenesis of neuropsychiatric disorders.


Assuntos
Interferon gama/metabolismo , Neocórtex/metabolismo , Neuroimunomodulação/fisiologia , Células Piramidais/metabolismo , Receptores de Interferon/metabolismo , Animais , Interferon gama/farmacologia , Masculino , Neocórtex/efeitos dos fármacos , Neocórtex/imunologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/imunologia , Ratos , Ratos Wistar
5.
Front Immunol ; 10: 1752, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31396238

RESUMO

Recent studies suggest that mild hypoxia-induced neonatal seizures can trigger an acute neuroinflammatory response leading to long-lasting changes in brain excitability along with associated cognitive and behavioral deficits. The cellular elements and signaling pathways underlying neuroinflammation in this setting remain incompletely understood but could yield novel therapeutic targets. Here we show that brief global hypoxia-induced neonatal seizures in mice result in transient cytokine production, a selective expansion of microglia and long-lasting changes to the neuronal structure of pyramidal neurons in the hippocampus. Treatment of neonatal mice after hypoxia-seizures with the novel anti-inflammatory compound candesartan cilexetil suppressed acute seizure-damage and mitigated later-life aggravated seizure responses and hippocampus-dependent learning deficits. Together, these findings improve our understanding of the effects of neonatal seizures and identify potentially novel treatments to protect against short and long-lasting harmful effects.


Assuntos
Anti-Inflamatórios/farmacologia , Benzimidazóis/farmacologia , Compostos de Bifenilo/farmacologia , Hipocampo/imunologia , Doenças do Recém-Nascido , Células Piramidais/imunologia , Convulsões , Tetrazóis/farmacologia , Animais , Modelos Animais de Doenças , Humanos , Recém-Nascido , Doenças do Recém-Nascido/imunologia , Doenças do Recém-Nascido/terapia , Camundongos , Microglia/imunologia , Convulsões/tratamento farmacológico , Convulsões/imunologia
6.
EBioMedicine ; 43: 424-434, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-31085101

RESUMO

BACKGROUND: The delivery of therapeutic proteins to selected sites within the central nervous system (CNS) parenchyma is a major challenge in the treatment of various neurodegenerative disorders. As brain-derived neurotrophic factor (BDNF) is reduced in the brain of people with Alzheimer's disease (AD) and its administration has shown promising therapeutic effects in mouse model of the disease, we generated a novel platform for T cell-based BDNF delivery into the brain parenchyma. METHODS: We generated amyloid beta-protein (Aß)-specific CD4 T cells (Aß-T cells), genetically engineered to express BDNF, and injected them intracerebroventricularly into the 5XFAD mouse model of AD. FINDINGS: The BDNF-secreting Aß-T cells migrated efficiently to amyloid plaques, where they significantly increased the levels of BDNF, its receptor TrkB, and various synaptic proteins known to be reduced in AD. Furthermore, the injected mice demonstrated reduced levels of beta-secretase 1 (BACE1)-a protease essential in the cleavage process of the amyloid precursor protein-and ameliorated amyloid pathology and inflammation within the brain parenchyma. INTERPRETATION: A T cell-based delivery of proteins into the brain can serve as a platform to modulate neurotoxic inflammation and to promote neuronal repair in neurodegenerative diseases.


Assuntos
Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/imunologia , Peptídeos beta-Amiloides/metabolismo , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Animais , Biomarcadores , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/patologia , Fator Neurotrófico Derivado do Encéfalo/genética , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linhagem Celular , Citocinas/metabolismo , Modelos Animais de Doenças , Epitopos de Linfócito T/imunologia , Feminino , Expressão Gênica , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos , Placa Amiloide/genética , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Células Piramidais/imunologia , Células Piramidais/metabolismo , Células Piramidais/patologia , Especificidade do Receptor de Antígeno de Linfócitos T , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
7.
J Cell Mol Med ; 23(3): 2103-2114, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30663210

RESUMO

We engineered and employed a chaperone-like amyloid-binding protein Nucleobindin 1 (NUCB1) to stabilize human islet amyloid polypeptide (hIAPP) protofibrils for use as immunogen in mice. We obtained multiple monoclonal antibody (mAb) clones that were reactive against hIAPP protofibrils. A secondary screen was carried out to identify clones that cross-reacted with amyloid beta-peptide (Aß42) protofibrils, but not with Aß40 monomers. These mAbs were further characterized in several in vitro assays, in immunohistological studies of a mouse model of Alzheimer's disease (AD) and in AD patient brain tissue. We show that mAbs obtained by immunizing mice with the NUCB1-hIAPP complex cross-react with Aß42, specifically targeting protofibrils and inhibiting their further aggregation. In line with conformation-specific binding, the mAbs appear to react with an intracellular antigen in diseased tissue, but not with amyloid plaques. We hypothesize that the mAbs we describe here recognize a secondary or quaternary structural epitope that is common to multiple amyloid protofibrils. In summary, we report a method to create mAbs that are conformation-sensitive and sequence-independent and can target more than one type of protofibril species.


Assuntos
Peptídeos beta-Amiloides/imunologia , Amiloide/imunologia , Anticorpos Monoclonais/imunologia , Fragmentos de Peptídeos/imunologia , Doença de Alzheimer/imunologia , Doença de Alzheimer/metabolismo , Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/metabolismo , Especificidade de Anticorpos/imunologia , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/patologia , Epitopos/química , Epitopos/imunologia , Epitopos/metabolismo , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/imunologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Camundongos , Nucleobindinas/imunologia , Nucleobindinas/metabolismo , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Conformação Proteica , Células Piramidais/imunologia , Células Piramidais/metabolismo
8.
Biol Psychiatry ; 85(11): 891-903, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30219209

RESUMO

BACKGROUND: Although several studies have linked adolescent cannabis use to long-term cognitive dysfunction, there are negative reports as well. The fact that not all users develop cognitive impairment suggests a genetic vulnerability to adverse effects of cannabis, which are attributed to action of Δ9-tetrahydrocannabinol (Δ9-THC), a cannabis constituent and partial agonist of brain cannabinoid receptor 1. As both neurons and glial cells express cannabinoid receptor 1, genetic vulnerability could influence Δ9-THC-induced signaling in a cell type-specific manner. METHODS: Here we use an animal model of inducible expression of dominant-negative disrupted in schizophrenia 1 (DN-DISC1) selectively in astrocytes to evaluate the molecular mechanisms, whereby an astrocyte genetic vulnerability could interact with adolescent Δ9-THC exposure to impair recognition memory in adulthood. RESULTS: Selective expression of DN-DISC1 in astrocytes and adolescent treatment with Δ9-THC synergistically affected recognition memory in adult mice. Similar deficits in recognition memory were observed following knockdown of endogenous Disc1 in hippocampal astrocytes in mice treated with Δ9-THC during adolescence. At the molecular level, DN-DISC1 and Δ9-THC synergistically activated the nuclear factor-κB-cyclooxygenase-2 pathway in astrocytes and decreased immunoreactivity of parvalbumin-positive presynaptic inhibitory boutons around pyramidal neurons of the hippocampal CA3 area. The cognitive abnormalities were prevented in DN-DISC1 mice exposed to Δ9-THC by simultaneous adolescent treatment with the cyclooxygenase-2 inhibitor, NS398. CONCLUSIONS: Our data demonstrate that individual vulnerability to cannabis can be exclusively mediated by astrocytes. Results of this work suggest that genetic predisposition within astrocytes can exaggerate Δ9-THC-produced cognitive impairments via convergent inflammatory signaling, suggesting possible targets for preventing adverse effects of cannabis within susceptible individuals.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Dronabinol/efeitos adversos , Memória/efeitos dos fármacos , NF-kappa B/metabolismo , Proteínas do Tecido Nervoso/genética , Reconhecimento Psicológico/efeitos dos fármacos , Fatores Etários , Animais , Astrócitos/metabolismo , Região CA3 Hipocampal/imunologia , Feminino , Técnicas de Silenciamento de Genes , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/biossíntese , Nitrobenzenos/farmacologia , Parvalbuminas/metabolismo , Terminações Pré-Sinápticas/efeitos dos fármacos , Células Piramidais/imunologia , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia
9.
Cereb Cortex ; 29(9): 3982-3992, 2019 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-30395185

RESUMO

Epidemiological and experimental studies suggest that maternal immune activation (MIA) leads to developmental brain disorders, but whether the pathogenic mechanism impacts neurons already at birth is not known. We now report that MIA abolishes in mice the oxytocin-mediated delivery γ-aminobutyric acid (GABA) shift from depolarizing to hyperpolarizing in CA3 pyramidal neurons, and this is restored by the NKCC1 chloride importer antagonist bumetanide. Furthermore, MIA hippocampal pyramidal neurons at birth have a more exuberant apical arbor organization and increased apical dendritic length than age-matched controls. The frequency of spontaneous glutamatergic postsynaptic currents is also increased in MIA offspring, as well as the pairwise correlation of the synchronized firing of active cells in CA3. These alterations produced by MIA persist, since at P14-15 GABA action remains depolarizing, produces excitatory action, and network activity remains elevated with a higher frequency of spontaneous glutamatergic postsynaptic currents. Therefore, the pathogenic actions of MIA lead to important morphophysiological and network alterations in the hippocampus already at birth.


Assuntos
Região CA3 Hipocampal/crescimento & desenvolvimento , Região CA3 Hipocampal/imunologia , Potenciais da Membrana , Gravidez/imunologia , Células Piramidais/imunologia , Ácido gama-Aminobutírico/imunologia , Animais , Região CA3 Hipocampal/efeitos dos fármacos , Dendritos/efeitos dos fármacos , Dendritos/imunologia , Feminino , Ácido Glutâmico/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Poli I-C/administração & dosagem , Células Piramidais/citologia , Células Piramidais/efeitos dos fármacos , Membro 2 da Família 12 de Carreador de Soluto/imunologia
10.
PLoS One ; 12(7): e0181409, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28746384

RESUMO

Antibodies against brain proteins were identified in the plasma of cancer patients and are defined to cause paraneoplastic neurological syndromes. The profiles of brain-directed antibodies in non-small cell lung cancer (NSCLC) are largely unknown. Here, for the first time, we compared autoantibodies against brain proteins in NSCLC (n = 18) against those present in age-matched non-cancer control subjects (n = 18) with a similar life-style, habit, and medical history. Self-recognizing immunoglobulin (IgG) are primarily directed against cells in the cortex (P = 0.008), hippocampus (P = 0.003-0.05), and cerebellum (P = 0.02). More specifically, IgG targets were prominent in the pyramidal, Purkinje, and granule cell layers. Furthermore, autoimmune IgG signals were localized to neurons (81%), astrocytes (48%), and endothelial (29%) cells. While cancer sera yielded overall higher intensity signals, autoantigens of 100, 65, 45, 37, and 30 kDa molecular weights were the most represented. Additionally, a group of 100 kDa proteins seem more prevalent in female adenocarcinoma patients (4/5, 80%). In conclusion, our results revealed autoantigen specificity in NSCLC, which implicitly depends on patient's demographics and disease history. Patients at risk for lung cancer but with no active disease revealed that the immune profile in NSCLC is disease-dependent.


Assuntos
Autoanticorpos/imunologia , Encéfalo/imunologia , Carcinoma Pulmonar de Células não Pequenas/imunologia , Neoplasias Pulmonares/imunologia , Adenocarcinoma/sangue , Adenocarcinoma/imunologia , Idoso , Idoso de 80 Anos ou mais , Animais , Antígenos Nucleares/imunologia , Astrócitos/imunologia , Autoanticorpos/sangue , Autoantígenos/química , Autoantígenos/imunologia , Western Blotting , Carcinoma Pulmonar de Células não Pequenas/sangue , Feminino , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Imuno-Histoquímica , Neoplasias Pulmonares/sangue , Masculino , Pessoa de Meia-Idade , Peso Molecular , Proteínas do Tecido Nervoso/imunologia , Neurônios/imunologia , Células de Purkinje/imunologia , Células Piramidais/imunologia , Ratos Sprague-Dawley
11.
Mol Med Rep ; 16(2): 1627-1634, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28627606

RESUMO

Ischemic preconditioning (IPC) is induced by exposure to brief durations of transient ischemia, which results in ischemic tolerance to a subsequent longer or lethal period of ischemia. In the present study, the effects of IPC (2 min of transient cerebral ischemia) were examined on immunoreactivity of platelet­derived growth factor (PDGF)­BB and on neuroprotection in the gerbil hippocampal CA1 region following lethal transient cerebral ischemia (LTCI; 5 min of transient cerebral ischemia). IPC was subjected to a 2­min sublethal ischemia and a LTCI was given 5­min transient ischemia. The animals in all of the groups were given recovery times of 1, 2 and 5 days and change in PDGF­BB immunoreactivity was examined as was the neuronal damage/death in the hippocampus induced by LTCI. LTCI induced a significant loss of pyramidal neurons in the hippocampal CA1 region 5 days after LTCI, and significantly decreased PDGF­BB immunoreactivity in the CA1 pyramidal neurons from day 1 after LTCI. Conversely, IPC effectively protected the CA1 pyramidal neurons from LTCI and increased PDGF­BB immunoreactivity in the CA1 pyramidal neurons post­LTCI. In conclusion, the results demonstrated that LTCI significantly altered PDGF­BB immunoreactivity in pyramidal neurons in the hippocampal CA1 region, whereas IPC increased the immunoreactivity. These findings indicated that PDGF­BB may be associated with IPC­mediated neuroprotection.


Assuntos
Região CA1 Hipocampal/metabolismo , Gerbillinae/metabolismo , Ataque Isquêmico Transitório/metabolismo , Proteínas Proto-Oncogênicas c-sis/metabolismo , Animais , Becaplermina , Região CA1 Hipocampal/imunologia , Região CA1 Hipocampal/patologia , Morte Celular/fisiologia , Hipocampo/metabolismo , Hipocampo/patologia , Imuno-Histoquímica , Ataque Isquêmico Transitório/patologia , Precondicionamento Isquêmico/métodos , Locomoção , Masculino , Neuroproteção , Proteínas Proto-Oncogênicas c-sis/imunologia , Células Piramidais/imunologia , Células Piramidais/metabolismo , Células Piramidais/patologia
12.
Nat Med ; 23(6): 714-722, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28504723

RESUMO

Impaired learning and cognitive function often occurs during systemic infection or inflammation. Although activation of the innate immune system has been linked to the behavioral and cognitive effects that are associated with infection, the underlying mechanisms remain poorly understood. Here we mimicked viral immune activation with poly(I:C), a synthetic analog of double-stranded RNA, and longitudinally imaged postsynaptic dendritic spines of layer V pyramidal neurons in the mouse primary motor cortex using two-photon microscopy. We found that peripheral immune activation caused dendritic spine loss, impairments in learning-dependent dendritic spine formation and deficits in multiple learning tasks in mice. These observed synaptic alterations in the cortex were mediated by peripheral-monocyte-derived cells and did not require microglial function in the central nervous system. Furthermore, activation of CX3CR1highLy6Clow monocytes impaired motor learning and learning-related dendritic spine plasticity through tumor necrosis factor (TNF)-α-dependent mechanisms. Taken together, our results highlight CX3CR1high monocytes and TNF-α as potential therapeutic targets for preventing infection-induced cognitive dysfunction.


Assuntos
Comportamento Animal , Espinhas Dendríticas/imunologia , Aprendizagem , Monócitos/imunologia , Córtex Motor/imunologia , Plasticidade Neuronal/imunologia , Células Piramidais/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Receptor 1 de Quimiocina CX3C , Espinhas Dendríticas/patologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imuno-Histoquímica , Microscopia Intravital , Camundongos , Microscopia , Poli I-C , Polinucleotídeos/farmacologia , Células Piramidais/patologia , Receptores de Quimiocinas/metabolismo
13.
Neuropathology ; 37(5): 398-406, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28493345

RESUMO

The apoptosis of pyramidal neurons in CA2 and CA3 subregions of the hippocampus is induced after infection with Mu-3 virus (Mu-3), a neuropathogenic strain of the JHM virus (JHMV), at 4-5 days post-inoculation (dpi). The viral antigens in the hippocampus are mainly found in the CD11b-positive cells distributed in the stratum oriens located outside the pyramidal layer, and only a few pyramidal neurons are infected. Furthermore, the apoptotic cells, indicated as showing caspase 3 (Cas3) activation, consist of a high number of uninfected cells. Therefore, it is considered that the apoptotic lesions occur through the indirect effects of infection, and not as a result of direct infection with Mu-3, similar to the reported neuronal apoptosis in the hippocampus after other types of infection. The apoptosis in the pyramidal neurons is accompanied by various types of proinflammatory cytokines depending on the causative agents. Thus, the local expression of proinflammatory cytokines was studied, revealing no correlation in the distribution of cytokine expression with the subregions showing apoptosis. However, the anti-inflammatory cytokine IL-10 was produced by pyramidal neurons of CA2 and CA3 at 3 dpi when there is no destructive change or viral invasion in the hippocampus.


Assuntos
Apoptose/imunologia , Infecções por Coronavirus/imunologia , Interleucina-10/biossíntese , Células Piramidais/imunologia , Células Piramidais/virologia , Animais , Camundongos , Vírus da Hepatite Murina , Células Piramidais/patologia
14.
J Alzheimers Dis ; 52(1): 243-69, 2016 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-27031470

RESUMO

Alzheimer's disease (AD) is a degenerative and irreversible disorder whose progressiveness is dependent on age. It is histopathologically characterized by the massive accumulation of insoluble forms of tau and amyloid-ß (Aß) asneurofibrillary tangles and neuritic plaques, respectively. Many studies have documented that these two polypeptides suffer several posttranslational modifications employing postmortem tissue sections from brains of patients with AD. In order to elucidate the molecular mechanisms underlying the posttranslational modifications of key players in this disease, including Aß and tau, several transgenic mouse models have been developed. One of these models is the 3×Tg-AD transgenic mouse, carrying three transgenes encoding APPSWE, S1M146V, and TauP301L proteins. To further characterize this transgenicmouse, we determined the accumulation of fibrillar Aß as a function of age in relation to the hyperphosphorylation patterns of TauP301L at both its N- and C-terminus in the hippocampal formation by immunofluorescence and confocal microscopy. Moreover, we searched for the expression of activated protein kinases and mediators of inflammation by western blot of wholeprotein extracts from hippocampal tissue sections since 3 to 28 months as well. Our results indicate that the presence of fibrillar Aß deposits correlates with a significant activation of astrocytes and microglia in subiculum and CA1 regions of hippocampus. Accordingly, we also observed a significant increase in the expression of TNF-α associated to neuritic plaques and glial cells. Importantly, there is an overexpression of the stress activated protein kinases SAPK/JNK and Cdk-5 in pyramidal neurons, which might phosphorylate several residues at the C-terminus of TauP301L. Therefore, the accumulation of Aß oligomers results in an inflammatory environment that upregulates kinases involved in hyperphosphorylation of TauP301L polypeptide.


Assuntos
Envelhecimento/imunologia , Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/metabolismo , Hipocampo/imunologia , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/patologia , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Modelos Animais de Doenças , Feminino , Hipocampo/patologia , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neuroglia/imunologia , Neuroglia/patologia , Fosforilação/imunologia , Placa Amiloide/imunologia , Placa Amiloide/patologia , Presenilina-1/genética , Presenilina-1/metabolismo , Células Piramidais/imunologia , Células Piramidais/patologia , Proteínas tau/genética
15.
Acta Neuropathol ; 131(2): 235-246, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26724934

RESUMO

To successfully treat Alzheimer's disease (AD), pathophysiological events in preclinical stages need to be identified. Preclinical AD refers to the stages that exhibit amyloid deposition in the brain but have normal cognitive function, which are replicated in young adult APPswe/PS1deltaE9 (deltaE9) mice. By long-term in vivo two-photon microscopy, we demonstrate impaired adaptive spine plasticity in these transgenic mice illustrated by their failure to increase dendritic spine density and form novel neural connections when housed in enriched environment (EE). Decrease of amyloid plaques by reducing BACE1 activity restores the gain of spine density upon EE in deltaE9 mice, but not the remodeling of neural networks. On the other hand, anti-inflammatory treatment with pioglitazone or interleukin 1 receptor antagonist in deltaE9 mice successfully rescues the impairments in increasing spine density and remodeling of neural networks during EE. Our data suggest that neuroinflammation disrupts experience-dependent structural plasticity of dendritic spines in preclinical stages of AD.


Assuntos
Doença de Alzheimer/imunologia , Espinhas Dendríticas/imunologia , Neuroimunomodulação/imunologia , Plasticidade Neuronal/imunologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/metabolismo , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/patologia , Modelos Animais de Doenças , Feminino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neuroimunomodulação/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Pioglitazona , Células Piramidais/efeitos dos fármacos , Células Piramidais/imunologia , Células Piramidais/patologia , Receptores Tipo I de Interleucina-1/antagonistas & inibidores , Receptores Tipo I de Interleucina-1/metabolismo , Córtex Somatossensorial/efeitos dos fármacos , Córtex Somatossensorial/imunologia , Córtex Somatossensorial/patologia , Tiazolidinedionas/farmacologia
16.
Sci Rep ; 6: 19106, 2016 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-26742695

RESUMO

Maternal immune activation (MIA) resulting from prenatal exposure to infectious pathogens or inflammatory stimuli is increasingly recognized to play an important etiological role in neuropsychiatric disorders with neurodevelopmental features. MIA in pregnant rodents induced by injection of the synthetic double-stranded RNA, Poly I:C, a mimic of viral infection, leads to a wide spectrum of behavioral abnormalities as well as structural and functional defects in the brain. Previous MIA studies using poly I:C prenatal treatment suggested that neurophysiological alterations occur in the hippocampus. However, these investigations used only juvenile or adult animals. We postulated that MIA-induced alterations could occur earlier at neonatal/early postnatal stages. Here we examined the neurophysiological properties of cultured pyramidal-like hippocampal neurons prepared from neonatal (P0-P2) offspring of pregnant rats injected with poly I:C. Offspring neurons from poly I:C-treated mothers exhibited significantly lower intrinsic excitability and stronger spike frequency adaptation, compared to saline. A similar lower intrinsic excitability was observed in CA1 pyramidal neurons from hippocampal slices of two weeks-old poly I:C offspring. Cultured hippocampal neurons also displayed lower frequency of spontaneous firing, higher charge transfer of IPSCs and larger amplitude of miniature IPSCs. Thus, maternal immune activation leads to strikingly early neurophysiological abnormalities in hippocampal neurons.


Assuntos
Antígenos Virais/farmacologia , Hipocampo/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Poli I-C/farmacologia , Células Piramidais/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Comportamento Animal/efeitos dos fármacos , Feminino , Hipocampo/imunologia , Hipocampo/patologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/imunologia , Efeitos Tardios da Exposição Pré-Natal/patologia , Cultura Primária de Células , Células Piramidais/imunologia , Células Piramidais/patologia , Ratos
17.
PLoS One ; 10(6): e0129555, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26090827

RESUMO

Type 1 narcolepsy is caused by deficiency of hypothalamic orexin/hypocretin. An autoimmune basis is suspected, but no specific antibodies, either causative or as biomarkers, have been identified. However, the AS03 adjuvanted split virion H1N1 (H1N1-AS03) vaccine, created to protect against the 2009 Pandemic, has been implicated as a trigger of narcolepsy particularly in children. Sera and CSFs from 13 H1N1-AS03-vaccinated patients (12 children, 1 young adult) with type 1 narcolepsy were tested for autoantibodies to known neuronal antigens including the N-methyl-D-aspartate receptor (NMDAR) and contactin-associated protein 2 (CASPR2), both associated with encephalopathies that include disordered sleep, to rodent brain tissue including the lateral hypothalamus, and to live hippocampal neurons in culture. When sufficient sample was available, CSF levels of melanin-concentrating hormone (MCH) were measured. Sera from 44 H1N1-ASO3-vaccinated children without narcolepsy were also examined. None of these patients' CSFs or sera was positive for NMDAR or CASPR2 antibodies or binding to neurons; 4/13 sera bound to orexin-neurons in rat brain tissue, but also to other neurons. MCH levels were a marginally raised (n = 8; p = 0.054) in orexin-deficient narcolepsy patients compared with orexin-normal children (n = 6). In the 44 H1N1-AS03-vaccinated healthy children, there was no rise in total IgG levels or in CASPR2 or NMDAR antibodies three weeks following vaccination. In conclusion, there were no narcolepsy-specific autoantibodies identified in type 1 narcolepsy sera or CSFs, and no evidence for a general increase in immune reactivity following H1N1-AS03 vaccination in the healthy children. Antibodies to other neuronal specific membrane targets, with their potential for directing use of immunotherapies, are still an important goal for future research.


Assuntos
Autoanticorpos/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/efeitos adversos , Narcolepsia/imunologia , Neurônios/imunologia , Adolescente , Animais , Autoanticorpos/sangue , Autoantígenos/imunologia , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/patologia , Criança , Pré-Escolar , Modelos Animais de Doenças , Feminino , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Vacinas contra Influenza/imunologia , Masculino , Narcolepsia/sangue , Narcolepsia/líquido cefalorraquidiano , Orexinas/líquido cefalorraquidiano , Ligação Proteica/imunologia , Células Piramidais/imunologia , Ratos , Adulto Jovem
18.
PLoS One ; 10(3): e0121364, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25774787

RESUMO

Antibodies to glutamic acid decarboxylase (GAD-ab) associate to different neurological syndromes. It is unknown if the diversity in syndrome association represents epitopes in different immunodominant domains or co-existence of antibodies to other proteins of the inhibitory synapsis. We examined the serum and CSF of 106 patients with anti-GAD related syndromes (39 cerebellar ataxia, 32 stiff-person syndrome [SPS], 18 epilepsy, and 17 limbic encephalitis [LE]). GAD65-ab titres were quantified by ELISA. Immunoblot was used to determine if the antibody-targeted epitopes of GAD65 and GAD67 were linear. A cell-based assay (CBA) with HEK293 cells expressing the GAD65 N-terminal, central catalytic domain, or C-terminal was used to investigate the immunodominant domains. Antibodies to GAD67, gamma-aminobutyric acid A receptor (GABAaR), glycine receptor (GlyR), GABAaR-associated protein (GABARAP), and gephyrin were determined with CBA. GAD-ab internalization was investigated using cultured rat hippocampal neurons. CSF GAD65-ab titres were higher in patients with cerebellar ataxia and LE compared to those with SPS (p = 0.02). GAD67-ab were identified in 81% of sera and 100% of CSF. GAD65-ab recognized linear epitopes in 98% of the patients and GAD67-ab in 42% (p<0.001). The GAD65 catalytic domain was recognized by 93% of sera, and the three domains by 22% of sera and 74% of CSF (p<0.001). Six patients had GABAaR-ab and another 6 had GlyR-ab without association to distinctive symptoms. None of the patients had gephyrin- or GABARAP-ab. GAD65-ab were not internalized by live neurons. Overall, these findings show that regardless of the neurological syndrome, the CSF immune response against GAD is more widespread than that of the serum and that there is no specific association between clinical phenotype and the presence of antibodies against other proteins of the inhibitory synapsis.


Assuntos
Autoanticorpos/imunologia , Autoimunidade , Glutamato Descarboxilase/imunologia , Doenças do Sistema Nervoso/imunologia , Adolescente , Adulto , Idoso , Animais , Autoanticorpos/sangue , Autoanticorpos/líquido cefalorraquidiano , Pré-Escolar , Epitopos/imunologia , Feminino , Neurônios GABAérgicos/imunologia , Humanos , Isoenzimas , Masculino , Pessoa de Meia-Idade , Doenças do Sistema Nervoso/sangue , Doenças do Sistema Nervoso/líquido cefalorraquidiano , Doenças do Sistema Nervoso/diagnóstico , Células Piramidais/imunologia , Ratos , Sinapses/imunologia , Síndrome , Adulto Jovem
19.
J Alzheimers Dis ; 43(1): 67-80, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25061055

RESUMO

Periodontal disease is a polymicrobial inflammatory disease that leads to chronic systemic inflammation and direct infiltration of bacteria/bacterial components, which may contribute to the development of Alzheimer's disease. ApoE-/- mice were orally infected (n = 12) with Porphyromonas gingivalis, Treponema denticola, Tannerella forsythia, and Fusobacterium nucleatum as mono- and polymicrobial infections. ApoE-/- mice were sacrificed following 12 and 24 weeks of chronic infection. Bacterial genomic DNA was isolated from all brain tissues except for the F. nucleatum mono-infected group. Polymerase chain reaction was performed using universal 16 s rDNA primers and species-specific primer sets for each organism to determine whether the infecting pathogens accessed the brain. Sequencing amplification products confirmed the invasion of bacteria into the brain during infection. The innate immune responses were detected using antibodies against complement activation products of C3 convertase stage and the membrane attack complex. Molecular methods demonstrated that 6 out of 12 ApoE-/- mice brains contained P. gingivalis genomic DNA at 12 weeks (p = 0.006), and 9 out of 12 at 24 weeks of infection (p = 0.0001). Microglia in both infected and control groups demonstrated strong intracellular labeling with C3 and C9, due to on-going biosynthesis. The pyramidal neurons of the hippocampus in 4 out of 12 infected mice brains demonstrated characteristic opsonization with C3 activation fragments (p = 0.032). These results show that the oral pathogen P. gingivalis was able to access the ApoE-/- mice brain and thereby contributed to complement activation with bystander neuronal injury.


Assuntos
Apolipoproteínas E/deficiência , Infecções por Bacteroidaceae/imunologia , Encéfalo/imunologia , Ativação do Complemento , Doenças Periodontais/imunologia , Porphyromonas gingivalis/patogenicidade , Animais , Apolipoproteínas E/genética , Infecções por Bacteroidaceae/microbiologia , Infecções por Bacteroidaceae/patologia , Encéfalo/microbiologia , Encéfalo/patologia , Doença Crônica , DNA Bacteriano/metabolismo , Modelos Animais de Doenças , Infecções por Fusobacterium/imunologia , Infecções por Fusobacterium/patologia , Fusobacterium nucleatum/genética , Masculino , Camundongos Knockout , Microglia/patologia , Microglia/fisiologia , Doenças Periodontais/microbiologia , Doenças Periodontais/patologia , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/isolamento & purificação , Células Piramidais/imunologia , Células Piramidais/patologia , Treponema denticola/genética , Infecções por Treponema/imunologia , Infecções por Treponema/patologia
20.
J Neurophysiol ; 106(3): 1515-24, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21753024

RESUMO

Cholecystokinin (CCK) is one of the most abundant neuropeptides in the brain, where it interacts with two G protein-coupled receptors (CCK-1 and CCK-2). Activation of both CCK receptors increases the activity of PLC, resulting in increases in intracellular calcium ion (Ca(2+)) release and activation of PKC. Whereas high density of CCK receptors has been detected in the superficial layers of the entorhinal cortex (EC), the functions of CCK in this brain region have not been determined. Here, we studied the effects of CCK on neuronal excitability of layer III pyramidal neurons in the EC. Our results showed that CCK remarkably increased the firing frequency of action potentials (APs). The effects of CCK on neuronal excitability were mediated via activation of CCK-2 receptors and required the functions of G proteins and PLC. However, CCK-mediated facilitation of neuronal excitability was independent of inositol trisphosphate receptors and PKC. CCK facilitated neuronal excitability by activating a cationic channel to generate membrane depolarization. The effects of CCK were suppressed by the generic, nonselective cationic channel blockers, 2-aminoethyldiphenyl borate and flufenamic acid, but potentiated by gadolinium ion and lanthanum ion at 100 µM. Depletion of extracellular Ca(2+) also counteracted CCK-induced increases in AC firing frequency. Moreover, CCK-induced enhancement of neuronal excitability was inhibited significantly by intracellular application of the antibody to transient receptor potential channel 5 (TRPC5), suggesting the involvement of TRPC5 channels. Our results provide a cellular and molecular mechanism to help explain the functions of CCK in vivo.


Assuntos
Colecistocinina/fisiologia , Córtex Entorrinal/fisiologia , Neurônios/fisiologia , Canais de Cátion TRPC/fisiologia , Animais , Anticorpos/toxicidade , Colecistocinina/antagonistas & inibidores , Colecistocinina/deficiência , Camundongos , Camundongos Knockout , Neurônios/imunologia , Células Piramidais/imunologia , Células Piramidais/fisiologia , Ratos , Ratos Sprague-Dawley , Receptor de Colecistocinina B/deficiência , Receptor de Colecistocinina B/genética , Canais de Cátion TRPC/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...