Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 275
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 50(4): 955-974, 2019 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-30995509

RESUMO

Neurodegenerative diseases of the central nervous system progressively rob patients of their memory, motor function, and ability to perform daily tasks. Advances in genetics and animal models are beginning to unearth an unexpected role of the immune system in disease onset and pathogenesis; however, the role of cytokines, growth factors, and other immune signaling pathways in disease pathogenesis is still being examined. Here we review recent genetic risk and genome-wide association studies and emerging mechanisms for three key immune pathways implicated in disease, the growth factor TGF-ß, the complement cascade, and the extracellular receptor TREM2. These immune signaling pathways are important under both healthy and neurodegenerative conditions, and recent work has highlighted new functional aspects of their signaling. Finally, we assess future directions for immune-related research in neurodegeneration and potential avenues for immune-related therapies.


Assuntos
Doenças Neurodegenerativas/imunologia , Transdução de Sinais/imunologia , Envelhecimento/imunologia , Animais , Ativação do Complemento , Progressão da Doença , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Gliose/imunologia , Gliose/patologia , Humanos , Imunidade Inata , Inflamação/imunologia , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microglia/imunologia , Modelos Imunológicos , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Doenças Neurodegenerativas/terapia , Agregação Patológica de Proteínas/imunologia , Receptores Imunológicos/imunologia , Fator de Crescimento Transformador beta/imunologia
2.
Int J Mol Sci ; 22(23)2021 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-34884629

RESUMO

In recent years, much attention has been paid to the study of the therapeutic effect of the microelement selenium, its compounds, especially selenium nanoparticles, with a large number of works devoted to their anticancer effects. Studies proving the neuroprotective properties of selenium nanoparticles in various neurodegenerative diseases began to appear only in the last 5 years. Nevertheless, the mechanisms of the neuroprotective action of selenium nanoparticles under conditions of ischemia and reoxygenation remain unexplored, especially for intracellular Ca2+ signaling and neuroglial interactions. This work is devoted to the study of the cytoprotective mechanisms of selenium nanoparticles in the neuroglial networks of the cerebral cortex under conditions of ischemia/reoxygenation. It was shown for the first time that selenium nanoparticles dose-dependently induce the generation of Ca2+ signals selectively in astrocytes obtained from different parts of the brain. The generation of these Ca2+ signals by astrocytes occurs through the release of Ca2+ ions from the endoplasmic reticulum through the IP3 receptor upon activation of the phosphoinositide signaling pathway. An increase in the concentration of cytosolic Ca2+ in astrocytes leads to the opening of connexin Cx43 hemichannels and the release of ATP and lactate into the extracellular medium, which trigger paracrine activation of the astrocytic network through purinergic receptors. Incubation of cerebral cortex cells with selenium nanoparticles suppresses ischemia-induced increase in cytosolic Ca2+ and necrotic cell death. Activation of A2 reactive astrocytes exclusively after ischemia/reoxygenation, a decrease in the expression level of a number of proapoptotic and proinflammatory genes, an increase in lactate release by astrocytes, and suppression of the hyperexcitation of neuronal networks formed the basis of the cytoprotective effect of selenium nanoparticles in our studies.


Assuntos
Astrócitos/citologia , Cálcio/metabolismo , Gliose/tratamento farmacológico , Nanopartículas/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Traumatismo por Reperfusão/prevenção & controle , Selênio/administração & dosagem , Animais , Antioxidantes/administração & dosagem , Antioxidantes/química , Astrócitos/efeitos dos fármacos , Astrócitos/imunologia , Astrócitos/metabolismo , Sinalização do Cálcio , Gliose/imunologia , Gliose/metabolismo , Gliose/patologia , Nanopartículas/química , Neurônios/efeitos dos fármacos , Neurônios/imunologia , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/química , Ratos , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Selênio/química
3.
Neurobiol Dis ; 121: 131-137, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30176351

RESUMO

Mutations in Cu/Zn superoxide dismutase (SOD1) are the cause of ~20% of cases of familial ALS (FALS), which comprise ~10% of the overall total number of cases of ALS. Mutant (mt) SOD1 is thought to cause FALS through a gain and not loss in function, perhaps as a result of the mutant protein's misfolding and aggregation. Previously we used a phage display library to raise single chain variable fragment antibodies (scFvs) against SOD1, which were found to decrease aggregation of mtSOD1 and toxicity in vitro. In the present study, we show that two scFvs directed against SOD1 ameliorate disease in G93A mtSOD1 transgenic mice and also decrease motor neuron loss, microgliosis, astrocytosis, as well as SOD1 burden and aggregation. The results suggest that the use of antibodies or antibody mimetics directed against SOD1 may be a useful therapeutic direction in mtSOD1-induced FALS. Since studies suggest that wild type SOD1 may be misfolded similar to that seen with mtSOD1, this therapeutic direction may be effective in sporadic as well as FALS.


Assuntos
Esclerose Lateral Amiotrófica/imunologia , Esclerose Lateral Amiotrófica/patologia , Anticorpos de Cadeia Única/administração & dosagem , Superóxido Dismutase/imunologia , Animais , Modelos Animais de Doenças , Feminino , Gliose/imunologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios Motores/imunologia , Agregação Patológica de Proteínas/imunologia , Medula Espinal/imunologia , Medula Espinal/patologia , Superóxido Dismutase/genética
4.
J Neuroinflammation ; 16(1): 163, 2019 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-31383034

RESUMO

BACKGROUND: Traumatic brain injury (TBI) is a major cause of death and disability. T cells were shown to infiltrate the brain during the first days after injury and to exacerbate tissue damage. The objective of this study was to investigate the hitherto unresolved role of immunosuppressive, regulatory T cells (Tregs) in experimental TBI. METHODS: "Depletion of regulatory T cell" (DEREG) and wild type (WT) C57Bl/6 mice, treated with diphtheria toxin (DTx) to deplete Tregs or to serve as control, were subjected to the controlled cortical impact (CCI) model of TBI. Neurological and motor deficits were examined until 5 days post-injury (dpi). At the 5 dpi endpoint, (immuno-) histological, protein, and gene expression analyses were carried out to evaluate the consequences of Tregs depletion. Comparison of parametric or non-parametric data between two groups was done using Student's t test or the Mann-Whitney U test. For multiple comparisons, p values were calculated by one-way or two-way ANOVA followed by specific post hoc tests. RESULTS: The overall neurological outcome at 5 dpi was not different between DEREG and WT mice but more severe motor deficits occurred transiently at 1 dpi in DEREG mice. DEREG and WT mice did not differ in the extent of brain damage, blood-brain barrier (BBB) disruption, or neuronal excitotoxicity, as examined by lesion volumetry, immunoglobulin G (IgG) extravasation, or calpain-generated αII-spectrin breakdown products (SBDPs), respectively. In contrast, increased protein levels of glial fibrillary acidic protein (GFAP) and GFAP+ astrocytes in the ipsilesional brain tissue indicated exaggerated reactive astrogliosis in DEREG mice. T cell counts following anti-CD3 immunohistochemistry and gene expression analyses of Cd247 (CD3 subunit zeta) and Cd8a (CD8a) further indicated an increased number of T cells infiltrating the brain injury sites of DEREG mice compared to WT. These changes coincided with increased gene expression of pro-inflammatory interferon-γ (Ifng) in DEREG mice compared to WT in the injured brain. CONCLUSIONS: The results show that the depletion of Tregs attenuates T cell brain infiltration, reactive astrogliosis, interferon-γ gene expression, and transiently motor deficits in murine acute traumatic brain injury.


Assuntos
Astrócitos/patologia , Lesões Encefálicas Traumáticas/patologia , Encéfalo/patologia , Gliose/patologia , Interferon gama/genética , Depleção Linfocítica , Linfócitos T Reguladores/patologia , Animais , Astrócitos/imunologia , Encéfalo/imunologia , Lesões Encefálicas Traumáticas/genética , Lesões Encefálicas Traumáticas/imunologia , Modelos Animais de Doenças , Gliose/genética , Gliose/imunologia , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Camundongos , Linfócitos T Reguladores/imunologia
5.
Epilepsy Behav ; 92: 235-244, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30711777

RESUMO

There is a large body of evidence suggesting that parasites could be a major preventable risk factor for epilepsy in low- and middle-income countries. We review potentially important substrates for epileptogenesis in parasitic diseases. Taenia solium is the most widely known parasite associated with epilepsy, and the risk seems determined mainly by the extent of cortical involvement and the evolution of the primary cortical lesion to gliosis or to a calcified granuloma. For most parasites, however, epileptogenesis is more complex, and other favorable host genetic factors and parasite-specific characteristics may be critical. In situations where cortical involvement by the parasite is either absent or minimal, parasite-induced epileptogenesis through an autoimmune process seems plausible. Further research to identify important markers of epileptogenesis in parasitic diseases will have huge implications for the development of trials to halt or delay onset of epilepsy.


Assuntos
Epilepsia/epidemiologia , Epilepsia/parasitologia , Doenças Parasitárias/epidemiologia , Animais , Epilepsia/imunologia , Gliose/imunologia , Gliose/parasitologia , Gliose/patologia , Humanos , Parasitos/imunologia , Parasitos/isolamento & purificação , Doenças Parasitárias/imunologia , Taenia solium/imunologia , Taenia solium/isolamento & purificação , Teníase/epidemiologia , Teníase/imunologia
6.
Proc Natl Acad Sci U S A ; 113(38): E5665-74, 2016 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-27601660

RESUMO

The generalized seizures of status epilepticus (SE) trigger a series of molecular and cellular events that produce cognitive deficits and can culminate in the development of epilepsy. Known early events include opening of the blood-brain barrier (BBB) and astrocytosis accompanied by activation of brain microglia. Whereas circulating monocytes do not infiltrate the healthy CNS, monocytes can enter the brain in response to injury and contribute to the immune response. We examined the cellular components of innate immune inflammation in the days following SE by discriminating microglia vs. brain-infiltrating monocytes. Chemokine receptor 2 (CCR2(+)) monocytes invade the hippocampus between 1 and 3 d after SE. In contrast, only an occasional CD3(+) T lymphocyte was encountered 3 d after SE. The initial cellular sources of the chemokine CCL2, a ligand for CCR2, included perivascular macrophages and microglia. The induction of the proinflammatory cytokine IL-1ß was greater in FACS-isolated microglia than in brain-invading monocytes. However, Ccr2 knockout mice displayed greatly reduced monocyte recruitment into brain and reduced levels of the proinflammatory cytokine IL-1ß in hippocampus after SE, which was explained by higher expression of the cytokine in circulating and brain monocytes in wild-type mice. Importantly, preventing monocyte recruitment accelerated weight regain, reduced BBB degradation, and attenuated neuronal damage. Our findings identify brain-infiltrating monocytes as a myeloid-cell subclass that contributes to neuroinflammation and morbidity after SE. Inhibiting brain invasion of CCR2(+) monocytes could represent a viable method for alleviating the deleterious consequences of SE.


Assuntos
Quimiocina CCL2/genética , Interleucina-1beta/metabolismo , Monócitos/patologia , Receptores CCR2/genética , Estado Epiléptico/imunologia , Animais , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/patologia , Quimiocina CCL2/metabolismo , Encefalite/imunologia , Encefalite/metabolismo , Encefalite/patologia , Gliose/imunologia , Gliose/metabolismo , Gliose/patologia , Imunidade Inata/genética , Interleucina-1beta/genética , Camundongos , Camundongos Knockout , Neurônios/imunologia , Neurônios/patologia , Receptores CCR2/metabolismo , Convulsões/genética , Convulsões/imunologia , Convulsões/metabolismo , Convulsões/patologia , Estado Epiléptico/metabolismo , Estado Epiléptico/patologia
7.
Eur J Neurosci ; 47(12): 1429-1443, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29790615

RESUMO

Müller glial cells (MGCs) are known to participate actively in retinal development and to contribute to homoeostasis through many intracellular mechanisms. As there are no homologous cells in other neuronal tissues, it is certain that retinal health depends on MGCs. These macroglial cells are located at the centre of the columnar subunit and have a great ability to interact with neurons, astrocytes, microglia and endothelial cells in order to modulate different events. Several investigations have focused their attention on the role of MGCs in diabetic retinopathy, a progressive pathology where several insults coexist. As expected, data suggest that MGCs display different responses according to the severity of the stimulus, and therefore trigger distinct events throughout the course of the disease. Here, we describe physiological functions of MGCs and their participation in inflammation, gliosis, synthesis and secretion of trophic and antioxidant factors in the diabetic retina. We invite the reader to consider the protective/deleterious role of MGCs in the early and late stages of the disease. In the light of the results, we open up the discussion around and ask the question: Is it possible that the modulation of a single cell type could improve or even re-establish retinal function after an injury?


Assuntos
Retinopatia Diabética , Células Ependimogliais/fisiologia , Gliose , Inflamação , Fatores de Crescimento Neural/fisiologia , Estresse Oxidativo/fisiologia , Animais , Retinopatia Diabética/imunologia , Retinopatia Diabética/metabolismo , Retinopatia Diabética/fisiopatologia , Células Ependimogliais/imunologia , Células Ependimogliais/metabolismo , Gliose/imunologia , Gliose/metabolismo , Gliose/fisiopatologia , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/fisiopatologia , Fatores de Crescimento Neural/imunologia , Fatores de Crescimento Neural/metabolismo , Estresse Oxidativo/imunologia
8.
Adv Exp Med Biol ; 1074: 185-191, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29721943

RESUMO

The eye is an immuno-privileged organ. However, certain diseases such as uveitis are intrinsically linked to inflammation. In several retinal degenerative diseases, there is a unique damage at the onset of the disease, but evidence suggests that chronic and low-grade inflammatory processes play an important role in their progression. Studies have identified similar signaling pathways and changes in resident immune cells within the retina among these diseases. Herein, we will discuss some of these studies and propose how understanding this inflammatory response could aid in the development of therapies.


Assuntos
Retinopatia Diabética/imunologia , Degeneração Macular/imunologia , Retinose Pigmentar/imunologia , Animais , Antígenos de Neoplasias/fisiologia , Citocinas/fisiologia , Retinopatia Diabética/patologia , Células Ependimogliais/imunologia , Células Ependimogliais/patologia , Gliose/imunologia , Gliose/patologia , Humanos , Inflamassomos/fisiologia , Inflamação , Degeneração Macular/patologia , Camundongos , Microglia/imunologia , Microglia/patologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Receptor para Produtos Finais de Glicação Avançada/deficiência , Retina/imunologia , Retina/patologia , Drusas Retinianas/imunologia , Drusas Retinianas/patologia , Retinose Pigmentar/patologia
9.
Glia ; 65(9): 1423-1438, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28608978

RESUMO

The activation of resident microglial cells, alongside the infiltration of peripheral macrophages, are key neuroinflammatory responses to traumatic brain injury (TBI) that are directly associated with neuronal death. Sexual disparities in response to TBI have been previously reported; however it is unclear whether a sex difference exists in neuroinflammatory progression after TBI. We exposed male and female mice to moderate-to-severe controlled cortical impact injury and studied glial cell activation in the acute and chronic stages of TBI using immunofluorescence and in situ hybridization analysis. We found that the sex response was completely divergent up to 7 days postinjury. TBI caused a rapid and pronounced cortical microglia/macrophage activation in male mice with a prominent activated phenotype that produced both pro- (IL-1ß and TNFα) and anti-inflammatory (Arg1 and TGFß) cytokines with a single-phase, sustained peak from 1 to 7 days. In contrast, TBI caused a less robust microglia/macrophage phenotype in females with biphasic pro-inflammatory response peaks at 4 h and 7 days, and a delayed anti-inflammatory mRNA peak at 30 days. We further report that female mice were protected against acute cell loss after TBI, with male mice demonstrating enhanced astrogliosis, neuronal death, and increased lesion volume through 7 days post-TBI. Collectively, these findings indicate that TBI leads to a more aggressive neuroinflammatory profile in male compared with female mice during the acute and subacute phases postinjury. Understanding how sex affects the course of neuroinflammation following brain injury is a vital step toward developing personalized and effective treatments for TBI.


Assuntos
Lesões Encefálicas Traumáticas/imunologia , Inflamação/fisiopatologia , Caracteres Sexuais , Animais , Arginase/metabolismo , Astrócitos/imunologia , Astrócitos/patologia , Lesões Encefálicas Traumáticas/patologia , Morte Celular , Modelos Animais de Doenças , Feminino , Gliose/imunologia , Gliose/patologia , Inflamação/patologia , Ativação de Macrófagos/imunologia , Ativação de Macrófagos/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Microglia/imunologia , Microglia/patologia , Neuroimunomodulação/fisiologia , Fator de Crescimento Transformador beta/metabolismo
10.
Neurobiol Dis ; 101: 27-39, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28189498

RESUMO

The immune system plays a crucial role in the progression of Alzheimer's disease (AD). Recently, immune-dependent cascade induced by systemic immune activation has been verified to play a beneficial role in AD mouse models. Here, we tested whether Bacillus Calmette-Guérin (BCG) immunization alters AD pathology and cognitive dysfunction in APP/PS1 AD mouse model, and with 4Aß1-15 vaccination as positive control. It was found that BCG treatment reversed the cognitive decline to the extent observed in 4Aß1-15 group, but did not reduce the ß-amyloid (Aß) burden in the brain. Then, we demonstrated the enhanced recruitment of inflammation-resolving monocytes across the choroid plexus and perivascular spaces to cerebral sites of plaque pathology in APP/PS1 mice immunized with BCG. Furthermore, elevated splenocyte Foxp3+ regulatory T cell levels in the control APP/PS1 mice were down-regulated back to the wild-type (WT) levels by BCG treatment but not 4Aß1-15 vaccination. In addition, BCG treatment induced the production of more circulating interferon (IFN)-γ than the controls and 4Aß1-15 vaccination. Though the similar reductions in brain levels of pro-inflammatory cytokines were observed in the BCG and 4Aß1-15 groups compared to the controls, only BCG had the great effect in upregulating cerebral anti-inflammatory cytokine levels as well as elevating the expression of neurotrophic factors in the brain of APP/PS1 mice. Thus, it is suggested that BCG exerts a beneficial immunomodulatory effect in APP/PS1 mice through mitigation of systemic immune suppression, induction of IFN-γ response and alleviation of the neuroinflammatory response.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/imunologia , Vacina BCG/uso terapêutico , Encéfalo/imunologia , Monócitos/imunologia , Doença de Alzheimer/patologia , Doença de Alzheimer/psicologia , Peptídeos beta-Amiloides/imunologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Modelos Animais de Doenças , Feminino , Fatores de Transcrição Forkhead/metabolismo , Gliose/tratamento farmacológico , Gliose/imunologia , Gliose/patologia , Humanos , Interleucina-10/metabolismo , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Monócitos/efeitos dos fármacos , Monócitos/patologia , Fragmentos de Peptídeos/imunologia , Baço/citologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia
11.
J Virol ; 90(1): 379-91, 2016 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-26491149

RESUMO

UNLABELLED: Varicella-zoster virus (VZV) is a highly neurotropic virus that can cause infections in both the peripheral nervous system and the central nervous system. Several studies of VZV reactivation in the peripheral nervous system (herpes zoster) have been published, while exceedingly few investigations have been carried out in a human brain. Notably, there is no animal model for VZV infection of the central nervous system. In this report, we characterized the cellular environment in the temporal lobe of a human subject who recovered from focal VZV encephalitis. The approach included not only VZV DNA/RNA analyses but also a delineation of infected cell types (neurons, microglia, oligodendrocytes, and astrocytes). The average VZV genome copy number per cell was 5. Several VZV regulatory and structural gene transcripts and products were detected. When colocalization studies were performed to determine which cell types harbored the viral proteins, the majority of infected cells were astrocytes, including aggregates of astrocytes. Evidence of syncytium formation within the aggregates included the continuity of cytoplasm positive for the VZV glycoprotein H (gH) fusion-complex protein within a cellular profile with as many as 80 distinct nuclei. As with other causes of brain injury, these results suggested that astrocytes likely formed a defensive perimeter around foci of VZV infection (astrogliosis). Because of the rarity of brain samples from living humans with VZV encephalitis, we compared our VZV results with those found in a rat encephalitis model following infection with the closely related pseudorabies virus and observed similar perimeters of gliosis. IMPORTANCE: Investigations of VZV-infected human brain from living immunocompetent human subjects are exceedingly rare. Therefore, much of our knowledge of VZV neuropathogenesis is gained from studies of VZV-infected brains obtained at autopsy from immunocompromised patients. These are not optimal samples with which to investigate a response by a human host to VZV infection. In this report, we examined both flash-frozen and paraffin-embedded formalin-fixed brain tissue of an otherwise healthy young male with focal VZV encephalitis, most likely acquired from VZV reactivation in the trigeminal ganglion. Of note, the cellular response to VZV infection mimicked the response to other causes of trauma to the brain, namely, an ingress of astrocytes and astrogliosis around an infectious focus. Many of the astrocytes themselves were infected; astrocytes aggregated in clusters. We postulate that astrogliosis represents a successful defense mechanism by an immunocompetent human host to eliminate VZV reactivation within neurons.


Assuntos
Astrócitos/imunologia , Encefalite por Varicela Zoster/patologia , Gliose/patologia , Herpesvirus Humano 3/imunologia , Animais , Astrócitos/virologia , Modelos Animais de Doenças , Encefalite por Varicela Zoster/imunologia , Encefalite por Varicela Zoster/virologia , Células Gigantes/patologia , Células Gigantes/virologia , Gliose/imunologia , Herpesvirus Suídeo 1 , Humanos , Masculino , Pseudorraiva/imunologia , Pseudorraiva/patologia , Pseudorraiva/virologia , Ratos Sprague-Dawley , Lobo Temporal/patologia , Lobo Temporal/virologia
12.
J Neuroinflammation ; 13(1): 89, 2016 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-27107718

RESUMO

BACKGROUND: Müller glial cells are important regulators of physiological function of retina. In a model disease of retinal inflammation and spontaneous recurrent uveitis in horses (ERU), we could show that retinal Müller glial cells significantly change potassium and water channel protein expression during autoimmune pathogenesis. The most significantly changed channel protein in neuroinflammatory ERU was aquaporin 11 (AQP11). Aquaporins (AQP, 13 members) are important regulators of water and small solute transport through membranes. AQP11 is an unorthodox member of this family and was assigned to a third group of AQPs because of its difference in amino acid sequence (conserved sequence is only 11 %) and especially its largely unknown function. METHODS: In order to gain insight into the distribution, localization, and function of AQP11 in the retina, we first developed a novel monoclonal antibody for AQP11 enabling quantification, localization, and functional studies. RESULTS: In the horse retina, AQP11 was exclusively expressed at Müller glial cell membranes. In uveitic condition, AQP11 disappeared from gliotic Müller cells concomitant with glutamine synthase. Since function of AQP11 is still under debate, we assessed the impact of AQP11 channel on cell volume regulation of primary Müller glial cells under different osmotic conditions. We conclude a concomitant role for AQP11 with AQP4 in water efflux from these glial cells, which is disturbed in ERU. This could probably contribute to swelling and subsequent severe complication of retinal edema through impaired intracellular fluid regulation. CONCLUSIONS: Therefore, AQP11 is important for physiological Müller glia function and the expression pattern and function of this water channel seems to have distinct functions in central nervous system. The significant reduction in neuroinflammation points to a crucial role in pathogenesis of autoimmune uveitis.


Assuntos
Aquaporinas/metabolismo , Doenças Autoimunes/veterinária , Células Ependimogliais/metabolismo , Gliose/veterinária , Uveíte/veterinária , Animais , Aquaporinas/imunologia , Doenças Autoimunes/metabolismo , Doenças Autoimunes/patologia , Western Blotting , Gliose/imunologia , Gliose/metabolismo , Doenças dos Cavalos , Cavalos , Imuno-Histoquímica , Pressão Osmótica , Uveíte/metabolismo , Uveíte/patologia
13.
J Neuroinflammation ; 13(1): 96, 2016 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-27130316

RESUMO

BACKGROUND: Rheumatoid arthritis (RA) patients frequently show weak correlations between the magnitude of pain and inflammation suggesting that mechanisms other than overt peripheral inflammation contribute to pain in RA. We assessed changes in microglial reactivity and spinal excitability and their contribution to pain-like behaviour in the early stages of collagen-induced arthritis (CIA) model. METHODS: Mechanically evoked hypersensitivity, spinal nociceptive withdrawal reflexes (NWRs) and hind paw swelling were evaluated in female Lewis rats before and until 13 days following collagen immunization. In the spinal dorsal horn, microgliosis was assayed using immunohistochemistry (Iba-1/p-p38) and cyto(chemo)kine levels in the cerebrospinal fluid (CSF). Intrathecal administration of microglia-targeting drugs A-438079 (P2X7 antagonist) and LHVS (cathepsin S inhibitor) were examined upon hypersensitivity, NWRs, microgliosis and cyto(chemo)kine levels in the early phase of CIA. RESULTS: The early phase of CIA was associated with mechanical allodynia and exaggerated mechanically evoked spinal NWRs, evident before hind paw swelling, and exacerbated with the development of swelling. Concomitant with the development of hypersensitivity was the presence of reactive spinal microgliosis and an increase of IL-1ß levels in CSF (just detectable in plasma). Prolonged intrathecal administration of microglial inhibitors attenuated the development of mechanical allodynia, reduced microgliosis and attenuated IL-1ß increments. Acute spinal application of either microglial inhibitor significantly diminished the sensitization of the spinal NWRs. CONCLUSIONS: Mechanical hypersensitivity in the early phase of CIA is associated with central sensitization that is dependent upon microglial-mediated release of IL-1ß in the spinal cord. Blockade of these spinal events may provide pain relief in RA patients.


Assuntos
Artrite Experimental/fisiopatologia , Artrite Reumatoide/fisiopatologia , Sensibilização do Sistema Nervoso Central/fisiologia , Hiperalgesia/fisiopatologia , Neurônios/metabolismo , Animais , Artrite Experimental/imunologia , Artrite Experimental/metabolismo , Artrite Reumatoide/imunologia , Artrite Reumatoide/metabolismo , Eletromiografia , Feminino , Gliose/imunologia , Gliose/metabolismo , Gliose/fisiopatologia , Hiperalgesia/imunologia , Hiperalgesia/metabolismo , Imuno-Histoquímica , Interleucina-1beta/metabolismo , Microglia/imunologia , Microglia/metabolismo , Microglia/patologia , Ratos , Ratos Endogâmicos Lew , Medula Espinal/imunologia , Medula Espinal/fisiopatologia
14.
J Neurovirol ; 22(2): 140-8, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26419894

RESUMO

Chikungunya, "that which bends up" in the Makonde dialect, is an emerging global health threat, with increasing incidence of neurological complications. Until 2013, Chikungunya infection had been largely restricted to East Africa and the Indian Ocean, with cases within the USA reported to be from foreign travel. However, in 2014, over 1 million suspected cases were reported in the Americas, and a recently infected human could serve as an unwitting reservoir for the virus resulting in an epidemic in the continental USA. Chikungunya infection is increasingly being associated with neurological sequelae. In this study, we sought to understand the role of astrocytes in the neuropathogenesis of Chikungunya infection. Even after virus has been cleared form the circulation, astrocytes were activated with regard to TLR2 expression. In addition, white matter astrocytes were hypertrophic, with increased arbor volume in gray matter astrocytes. Combined, these would alter the number and distribution of synapses that each astrocyte would be capable of forming. These results provide the first evidence that Chikungunya infection induces morphometric and innate immune activation of astrocytes in vivo. Perturbed glia-neuron signaling could be a major driving factor in the development of Chikungunya-associated neuropathology.


Assuntos
Astrócitos/patologia , Febre de Chikungunya/patologia , Gliose/patologia , Imunidade Inata , Neurônios/patologia , Animais , Astrócitos/imunologia , Astrócitos/virologia , Febre de Chikungunya/genética , Febre de Chikungunya/imunologia , Febre de Chikungunya/virologia , Vírus Chikungunya/imunologia , Vírus Chikungunya/patogenicidade , Modelos Animais de Doenças , Expressão Gênica , Gliose/genética , Gliose/imunologia , Gliose/virologia , Substância Cinzenta/imunologia , Substância Cinzenta/patologia , Substância Cinzenta/virologia , Interações Hospedeiro-Patógeno , Humanos , Macaca fascicularis , Neurônios/imunologia , Neurônios/virologia , Transdução de Sinais , Telemetria , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/imunologia , Substância Branca/imunologia , Substância Branca/patologia , Substância Branca/virologia
15.
Brain Behav Immun ; 56: 42-55, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27067748

RESUMO

Toll-like receptor 4 (TLR4) is a crucial receptor in the innate immune system, and increasing evidence supports its role in inflammation, stress, and tissue injury, including injury to the lung and brain. We aimed to investigate the effects of TLR4 on neuroinflammation due to the lung-brain interaction in mechanically ventilated mice. Male wild-type (WT) C57BL/6 and TLR4 knockout (TLR4 KO) mice were divided into three groups: (1) control group (C): spontaneous breathing; (2) anesthesia group (A): spontaneous breathing under anesthesia; and (3) mechanical ventilation group (MV): 6h of MV under anesthesia. The behavioral responses of mice were tested with fear conditioning tests. The histological changes in the lung and brain were assessed using hematoxylin-eosin (HE) staining. The level of TLR4 mRNA in tissue was measured using reverse transcription-polymerase chain reaction (RT-PCR). The levels of inflammatory cytokines were measured with an enzyme-linked immunosorbent assay (ELISA). Microgliosis, astrocytosis, and the TLR4 immunoreactivity in the hippocampus were measured by double immunofluorescence. MV mice exhibited impaired cognition, and this impairment was less severe in TLR4 KO mice than in WT mice. In WT mice, MV increased TLR4 mRNA expression in the lung and brain. MV induced mild lung injury, which was prevented in TLR4 KO mice. MV mice exhibited increased levels of inflammatory cytokines, increased microglia and astrocyte activation. Microgliosis was alleviated in TLR4 KO mice. MV mice exhibited increased TLR4 immunoreactivity, which was expressed in microglia and astrocytes. These results demonstrate that TLR4 is involved in neuroinflammation due to the lung-brain interaction and that TLR4 KO ameliorates neuroinflammation due to lung-brain interaction after prolonged MV. In addition, Administration of a TLR4 antagonist (100µg/mice) to WT mice also significantly attenuated neuroinflammation of lung-brain interaction due to prolonged MV. TLR4 antagonism may be a new and novel approach for the treatment and management of neuroinflammation in long-term mechanically ventilated patients.


Assuntos
Encéfalo , Citocinas , Gliose , Inflamação , Pulmão/metabolismo , Respiração Artificial , Receptor 4 Toll-Like/metabolismo , Animais , Encéfalo/imunologia , Encéfalo/metabolismo , Citocinas/imunologia , Citocinas/metabolismo , Gliose/imunologia , Gliose/metabolismo , Hipocampo/imunologia , Hipocampo/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
16.
Brain ; 138(Pt 11): 3299-315, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26115674

RESUMO

See Moon (doi:10.1093/awv239) for a scientific commentary on this article.Traumatic brain injury frequently leads to long-term cognitive problems and physical disability yet remains without effective therapeutics. Traumatic brain injury results in neuronal injury and death, acute and prolonged inflammation and decreased blood flow. Drugs that block angiotensin II type 1 receptors (AT1R, encoded by AGTR1) (ARBs or sartans) are strongly neuroprotective, neurorestorative and anti-inflammatory. To test whether these drugs may be effective in treating traumatic brain injury, we selected two sartans, candesartan and telmisartan, of proven therapeutic efficacy in animal models of brain inflammation, neurodegenerative disorders and stroke. Using a validated mouse model of controlled cortical impact injury, we determined effective doses for candesartan and telmisartan, their therapeutic window, mechanisms of action and effect on cognition and motor performance. Both candesartan and telmisartan ameliorated controlled cortical impact-induced injury with a therapeutic window up to 6 h at doses that did not affect blood pressure. Both drugs decreased lesion volume, neuronal injury and apoptosis, astrogliosis, microglial activation, pro-inflammatory signalling, and protected cerebral blood flow, when determined 1 to 3 days post-injury. Controlled cortical impact-induced cognitive impairment was ameliorated 30 days after injury only by candesartan. The neurorestorative effects of candesartan and telmisartan were reduced by concomitant administration of the peroxisome proliferator-activated receptor gamma (PPARγ, encoded by PPARG) antagonist T0070907, showing the importance of PPARγ activation for the neurorestorative effect of these sartans. AT1R knockout mice were less vulnerable to controlled cortical impact-induced injury suggesting that the sartan's blockade of the AT1R also contributes to their efficacy. This study strongly suggests that sartans with dual AT1R blocking and PPARγ activating properties have therapeutic potential for traumatic brain injury.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Apoptose/efeitos dos fármacos , Benzimidazóis/farmacologia , Benzoatos/farmacologia , Lesões Encefálicas/patologia , Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Tetrazóis/farmacologia , Animais , Benzamidas/farmacologia , Compostos de Bifenilo , Lesões Encefálicas/imunologia , Lesões Encefálicas/metabolismo , Circulação Cerebrovascular/efeitos dos fármacos , Gliose/imunologia , Gliose/metabolismo , Gliose/patologia , Inflamação , Camundongos , Camundongos Knockout , PPAR gama/antagonistas & inibidores , Piridinas/farmacologia , Receptor Tipo 1 de Angiotensina/genética , Transdução de Sinais/efeitos dos fármacos , Telmisartan
17.
J Neuroinflammation ; 12: 79, 2015 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-25896970

RESUMO

BACKGROUND: Therapeutic modalities effective in patients with progressive forms of multiple sclerosis (MS) are limited. In a murine model of progressive MS, the sustained disability during the chronic phase of experimental autoimmune encephalomyelitis (EAE) correlated with elevated expression of interleukin (IL)-6, a cytokine with pleiotropic functions and therapeutic target for non-central nervous system (CNS) autoimmune disease. Sustained IL-6 expression in astrocytes restricted to areas of demyelination suggested that IL-6 plays a major role in disease progression during chronic EAE. METHODS: A progressive form of EAE was induced using transgenic mice expressing a dominant negative interferon-γ (IFN-γ) receptor alpha chain under control of human glial fibrillary acidic protein (GFAP) promoter (GFAPγR1Δ mice). The role of IL-6 in regulating progressive CNS autoimmunity was assessed by treating GFAPγR1Δ mice with anti-IL-6 neutralizing antibody during chronic EAE. RESULTS: IL-6 neutralization restricted disease progression and decreased disability, myelin loss, and axonal damage without affecting astrogliosis. IL-6 blockade reduced CNS inflammation by limiting inflammatory cell proliferation; however, the relative frequencies of CNS leukocyte infiltrates, including the Th1, Th17, and Treg CD4 T cell subsets, were not altered. IL-6 blockade rather limited the activation and proliferation of microglia, which correlated with higher expression of Galectin-1, a regulator of microglia activation expressed by astrocytes. CONCLUSIONS: These data demonstrate that astrocyte-derived IL-6 is a key mediator of progressive disease and support IL-6 blockade as a viable intervention strategy to combat progressive MS.


Assuntos
Astrócitos/efeitos dos fármacos , Encefalomielite Autoimune Experimental/imunologia , Interferon gama/farmacologia , Interleucina-6/antagonistas & inibidores , Ativação de Macrófagos/efeitos dos fármacos , Microglia/efeitos dos fármacos , Animais , Anticorpos Neutralizantes/uso terapêutico , Doenças Desmielinizantes/patologia , Encefalomielite Autoimune Experimental/tratamento farmacológico , Proteína Glial Fibrilar Ácida/genética , Gliose/imunologia , Humanos , Interleucina-6/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Esclerose Múltipla/tratamento farmacológico , Regiões Promotoras Genéticas/genética
18.
Clin Exp Immunol ; 180(3): 419-31, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25619397

RESUMO

Recently, parasite infections or parasite-derived products have been suggested as a therapeutic strategy with suppression of immunopathology, which involves the induction of regulatory T cells or/and T helper type 2 (Th2) responses. In a recent study, researchers reported that constructed recombinant galectin (rTl-gal) isolated from an adult worm of the gastrointestinal nematode parasite Toxascaris leonina attenuated clinical symptoms of inflammatory bowel disease in mice treated with dextran sulphate sodium. Noting the role of rTl-gal in inflammatory disease, we attempted to investigate the effect of the parasite via its rTl-gal on neuronal autoimmune disease using experimental autoimmune encephalomyelitis (EAE), a mouse inflammatory and demyelinating autoimmune disease model of human multiple sclerosis. In this model, rTl-gal-treated experimental autoimmune encephalomyelitis (EAE) mice failed to recover after the peak of the disease, leading to persistent central nervous system (CNS) damage, such as demyelination, gliosis and axonal damage. Further, rTl-gal-treated EAE mice markedly increased the number of CD45R/B220(+) B cells in both infiltrated inflammation and the periphery, along with the increased production of autoantibody [anti-myelin oligodendrocyte glycoprotein (MOG)35-55 ] in serum at chronic stage. Upon antigen restimulation, rTl-gal treatment affected the release of overall cytokines, especially interferon (IFN)-γ and tumour necrosis factor (TNF)-α. Our results suggest that galectin isolated from a gastrointestinal parasite can deliver a harmful effect to EAE contrary to its beneficial effect on inflammatory bowel disease.


Assuntos
Autoanticorpos/imunologia , Encefalomielite Autoimune Experimental/imunologia , Galectinas/imunologia , Imunomodulação/efeitos dos fármacos , Parasitos/química , Animais , Autoanticorpos/sangue , Axônios/imunologia , Axônios/metabolismo , Axônios/patologia , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Citocinas/biossíntese , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Progressão da Doença , Encefalomielite Autoimune Experimental/diagnóstico , Feminino , Galectinas/efeitos adversos , Galectinas/isolamento & purificação , Gliose/imunologia , Gliose/metabolismo , Gliose/patologia , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Glicoproteína Mielina-Oligodendrócito/efeitos adversos , Glicoproteína Mielina-Oligodendrócito/imunologia , Fragmentos de Peptídeos/efeitos adversos , Fragmentos de Peptídeos/imunologia , Índice de Gravidade de Doença , Medula Espinal/imunologia , Medula Espinal/patologia
19.
BMC Neurosci ; 16: 48, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26232001

RESUMO

BACKGROUND: Inflammation following intracerebral hemorrhage (ICH) significantly contributes to secondary brain damage and poor outcomes. Prostaglandin E2 (PGE2) is known to modulate neuroinflammatory responses and is upregulated in response to brain injury as a result of changes in inducible cyclooxygenase 2 (COX-2) and the membrane-bound type of PGE synthase. Inhibition of COX-2 activity has been reported to attenuate ICH-induced brain injury; however, the clinical utility of such drugs is limited due to the potential for severe side effects. Therefore, it is now important to search for downstream targets capable of preferentially modulating PGE2 signaling, and the four E prostanoid receptors, EP1-4, which are the main targets of PGE2, remain a viable therapeutic option. We have previously shown that EP1 receptor deletion aggravates ICH-induced brain injury and impairs functional recovery, thus the current study aimed to elaborate on these results by including a pharmacologic approach targeting the EP1 receptor. RESULTS: Chronic post-treatment with the selective EP1 receptor antagonist, SC-51089, increased lesion volume by 30.1 ± 14.5% (p < 0.05) and treatment with the EP1 agonist, 17-pt-PGE2, improved neuromuscular functional recovery on grip strength (p < 0.01) and hanging wire (p < 0.05) behavioral testing. To begin identifying the mechanisms involved in EP1-mediated neuroprotection after ICH, histology was performed to assess ferric iron content, neuroinflammation, leukocyte transendothelial migratory potential, and peripheral neutrophil and immunoglobulin infiltration. Following ICH, mice treated with the antagonist displayed increased ferric iron (p < 0.05) and cortical microgliosis (p < 0.05), whereas treatment with the agonist decreased cortical (p < 0.01) and striatal (p < 0.001) astrogliosis, leukocyte transendothelial migratory potential (p < 0.01), neutrophil infiltration (p < 0.05), and blood brain barrier breakdown (p < 0.05). CONCLUSIONS: In agreement with our previous results, selective antagonism of the EP1 receptor aggravated ICH-induced brain injury. Furthermore, EP1 receptor agonism improved anatomical outcomes and functional recovery. Thus, the present data continues to reinforce a putative role for EP1 as a new and more selective therapeutic target for the treatment of ICH that could reduce the side effects associated with COX-2 inhibition while still exploiting the beneficial effects.


Assuntos
Encéfalo/efeitos dos fármacos , Hemorragia Cerebral/tratamento farmacológico , Receptores de Prostaglandina E Subtipo EP1/agonistas , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/imunologia , Astrócitos/patologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/patologia , Encéfalo/imunologia , Encéfalo/patologia , Hemorragia Cerebral/imunologia , Hemorragia Cerebral/patologia , Colagenases , Modelos Animais de Doenças , Gliose/tratamento farmacológico , Gliose/imunologia , Gliose/patologia , Hidrazinas/farmacologia , Ferro/metabolismo , Leucócitos/efeitos dos fármacos , Leucócitos/imunologia , Leucócitos/patologia , Masculino , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/imunologia , Microglia/patologia , Neuroimunomodulação/efeitos dos fármacos , Neuroimunomodulação/fisiologia , Fármacos Neuroprotetores/farmacologia , Oxazepinas/farmacologia , Receptores de Prostaglandina E Subtipo EP1/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Recuperação de Função Fisiológica/efeitos dos fármacos
20.
Mediators Inflamm ; 2015: 873860, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25873774

RESUMO

Trinucleotide repeat expansion disorders (TREDs) are a group of dominantly inherited neurological diseases caused by the expansion of unstable repeats in specific regions of the associated genes. Expansion of CAG repeat tracts in translated regions of the respective genes results in polyglutamine- (polyQ-) rich proteins that form intracellular aggregates that affect numerous cellular activities. Recent evidence suggests the involvement of an RNA toxicity component in polyQ expansion disorders, thus increasing the complexity of the pathogenic processes. Neurodegeneration, accompanied by reactive gliosis and astrocytosis is the common feature of most TREDs, which may suggest involvement of inflammation in pathogenesis. Indeed, a number of immune response markers have been observed in the blood and CNS of patients and mouse models, and the activation of these markers was even observed in the premanifest stage of the disease. Although inflammation is not an initiating factor of TREDs, growing evidence indicates that inflammatory responses involving astrocytes, microglia, and the peripheral immune system may contribute to disease progression. Herein, we review the involvement of the immune system in the pathogenesis of triplet repeat expansion diseases, with particular emphasis on polyglutamine disorders. We also present various therapeutic approaches targeting the dysregulated inflammation pathways in these diseases.


Assuntos
Sistema Imunitário/imunologia , Doenças do Sistema Nervoso/imunologia , Expansão das Repetições de Trinucleotídeos , Processamento Alternativo , Animais , Gliose/imunologia , Gliose/fisiopatologia , Humanos , Inflamação/imunologia , Inflamação/fisiopatologia , Camundongos , Mutação , Doenças do Sistema Nervoso/fisiopatologia , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/fisiopatologia , Peptídeos/química , Interferência de RNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA